Anda di halaman 1dari 118

THE

JOURNAL OF
EXPERIMENTAL
MEDICINE
SELECTED ARTICLES MARCH 2015 www.jem.org

T CELLS
MACS Technology for immunology
Empowering discovery. Advancing science.

MACS Sample Preparation MACS Cell Separation MACS Cell Analysis & Culture

Preserved cell integrity and Research and GMP grade Broad range of instruments and
surface epitopes products for cell isolation reagents for flow cytometry
Gentle yet effective tissue Isolated cells always compatible Research and GMP grade
dissociation with downstream application antibodies, media, cytokines,
Flexible kits for any tissue type The most cited cell separation and antigens for all your cell
technology culture needs

Tissue dissociation Cell isolation or depletion Downstream applications

miltenyibiotec.com
Unless otherwise specifically indicated, Miltenyi Biotec products and services are for research use only and not for therapeutic or diagnostic use.
MACS GMP products are manufactured and tested under a quality management system (ISO 13485) and in compliance with relevant GMP guidelines. They are designed
following the recommendations of USP <1043> on ancillary materials. They are for research use and ex vivo cell culture processing only, and is not intended for human in vivo
applications. MACS is a registered trademarks of Miltenyi Biotec GmbH. Copyright 2015 Miltenyi Biotec GmbH. All rights reserved.
Welcome
T Cells
The Journal of Experimental Medicine now prints topic-specific mini collections to showcase a handful
of our recent publications. In this installment, we highlight papers focusing on the regulation and
effector function of T cells.
Our collection begins with an insight from Luc Van Kaer describing the findings from Bougeois
et al. establishing a role for bee and wasp venom enzymes in generating neoantigens that activate
CD1a-restricted T cells.They show that phospholipase A2, which is present in insect venom, acts on
human skin-derived phospholipids to release fatty acid antigens that are then loaded onto CD1a and
presented to T cells, which subsequently become activated and produce interleukin (IL)-22.
Mutations in dedicator of cytokinesis 8 (DOCK8) can result in an inherited combined immuno
deficiency in humans. Zang et al. demonstrate that susceptibility to skin infections seen in patients with
DOCK8 mutations is due to a form of catastrophic cell death the authors term cytothripsis (cell shattering),
as T cell traffic through the collagen-dense tissue network of the skin. This impaired flexibility prevents the
generation of skin-resident memory CD8+ T cells that are needed to control herpesvirus skin infections. An insight by Stuart Tangye
accompanies the article and speculates on how the findings could be used to limit pathology incurred by skin infections.
Flu infection can cause respiratory as well as gastrointestinal symptoms, even though the virus only exhibits tropism for
respiratory tissues. An insight from Carolina Amezcua, Nicola Gagliani, and Richard Flavell highlight findings from Wang et al.,
who provide an explanation for gut inflammation in the absence of detectable virus in the gastrointestinal tract. They find that
infection in mice recruits lung-derived IFNg secreting CCR9+CD4+T cells into the small intestine that alter the composition
of the gut microbiota. Th17 cells then expand in the small intestine and neutralization with IL-17A or antibiotic treatment
reduces intestinal injury.
In a human study of hepatitis B and C, Kurktschiev et al. implicate the transcription factor T-bet in viral clearance. The
authors find that acute resolving infections are characterized by high expression of T-bet in CD8+T cells, which is correlated
with enhanced IFNg production, while absence of T-bet is more often seen in patients whose infections become chronic. IFNg
induction and T-bet expression are restored in dysfunctional T cells upon IL-2 and IL-12 supplementation.
Rapid and effective adaptive immune responses to viral pathogens rely on immunological memory and are curtailed by
lymphocyte exhaustion. An article by Penaloza-Macmaster et al. investigates how regulatory T cells (Treg) can modulate CD8+
T cell exhaustion during chronic lymphocytic choriomeningitis virus infection in mice. Their findings show that depletion of
Tregs can expand functional virus-specific CD8+T cells, rescuing exhausted CD8+ T cell subpopulations. Treg depletion also
upregulates the programmed-death ligand 1 receptor (PD-L1) on CD8+ T cells, but it is a combination of PD-L1 blockade and
Treg depletion that is able to reduce viral load. These findings suggest that Tregs have the ability to contribute to the maintenance
of exhausted CD8+ T cells during chronic infection.
Medullary thymic epithelial cells (mTECs) contribute to self-tolerance through thymic expression of tissue-specific antigens
(TSAs). Modulating central tolerance is an attractive therapeutic strategy for cancer treatment. Khan et al. demonstrate that in vivo
blockade of RANKL can inhibit the development and turnover of mTECs and alter central T cell tolerance. In vivo RANKL
blockade in mice transiently depletes Aire and TSA expression in the thymus inhibiting negative selection. RANKL blockade
can also rescue melanoma-specific T cells from thymic deletion, with tumor-specific effector CD4+ T cells facilitating host survival
in response to tumor challenge.
Adult T-cell leukemia/lymphoma (ATLL) is an aggressive malignancy caused by human T cell lymphotropic virus type1
(HTLV-1). Increased expression of CCR4 is a hallmark of ATLL, but it is not clear whether dysregulated CCR4 contributes to
disease pathogenesis. An article by Nakagawa et al. identifies recurring somatic mutations of CCR4 in ATLL patients, finding
that a CCR4 gain-of-function mutation impairs CCR4 internalization and increases cell migration and chemotaxis in vitro. An
accompanying insight by Kevin Shannon discusses the implications of CCR4 mutations, PI-3K signaling downstream of CCR4,
and the translational potential of CCR4 blockade.
Acute cerebral ischemia reperfusion injury is mediated in part by T cells. Clarkson et al. show that brain-infiltrating
CD4+T cells sustain neuroinflammation after stroke in mice by producing IL-21 and increasing neuronal death. Treatment with
an IL-21 decoy receptor or genetic lack of IL-21 protected animals from brain injury following stroke, offering a potential
target for immunotherapy. Additionally, analysis of postmortem human brain tissue confirmed that IL-21 localizes to CD4+T cells
surrounding acute stroke lesions.
Together these studies provide new insights into the biology and mechanisms of T cell responses in several disease and
pathogen conditions and offer insight into therapeutics. We hope you enjoy this complimentary copy of our T cell collection.
We invite you to explore additional collections at www.jem.org and to follow JEM on Facebook, Google+, and Twitter.
NEW ADJUVANTS FROM AVANTI 3D-PHAD

Monophosphoryl 3-Deacyl Lipid A (Synthetic)


Avanti Number 699852

PHOSPHORYLATED HEXAACYL DISACCHARIDE


(PHAD)
Monophosphoryl
Lipid A (Synthetic)
Avanti Number 699800

Vaccination is well-accepted as an effective method to


prevent infections by mounting pathogen-specific im-
mune responses prior to the infection. Usually, immuni-
zation with vaccine antigens alone is not able to induce
robust or long-lasting immune responses resulting in
failure of protective immunity against infections. Thus,
adjuvants are required to enhance cellular or humoral
immune responses upon immunization. Because vac-
cine adjuvants using Lipid A have proven to be safe and
effective in inducing Th-1 type immune responses to het-
erologous proteins in animal and human vaccines, Avan-
Stimulatory effect of PHAD on macrophages.
Macrophage cell line J774 cells were cultured with
ti developed Phosphorylated HexaAcyl Disaccharide
Avanti PHAD for 24hrs. IL-12 levels in superna- (PHAD), the first fully synthetic monophosphoryl Lipid
tants were measured by sandwich ELISA A available for use as an adjuvant in human vaccines.
For details of these and other adjuvants visit
www.avantilipids.com

THERE IS ONLY ONE AVANTI


AVANTI FOR THE MS STANDARDS YOU CAN TRUST
Selected Articles March 2015

Bee venom stirs up buzz in antigen presentation


Luc Van Kaer

Bee venom processes human skin lipids for presentation by CD1a


Elvire A. Bourgeois, Sumithra Subramaniam, Tan-Yun Cheng, Annemieke De Jong, Emilie Layre, Dalam Ly,
Maryam Salimi, Annaliza Legaspi, Robert L. Modlin, Mariolina Salio, Vincenzo Cerundolo, D. Branch Moody,
and Graham Ogg

T cells require DOCK8 for flexibility and function


Stuart Tangye

DOCK8 regulates lymphocyte shape integrity for skin antiviral immunity


Qian Zhang, Christopher G. Dove, Jyh Liang Hor, Heardley M. Murdock, Dara M. Strauss-Albee, Jordan A. Garcia,
Judith N. Mandl, Rachael A. Grodick, Huie Jing, Devon B. Chandler-Brown, Timothy E. Lenardo, Greg Crawford,
Helen F. Matthews, Alexandra F. Freeman, Richard J. Cornall, Ronald N. Germain, Scott N. Mueller, and Helen C. Su

FLUshing in the bathroom


Carolina Amezcua, Nicola Gagliani, and Richard Flavell

Respiratory influenza virus infection induces intestinal immune injury via microbiotamediated
Th17 celldependent inflammation
Jian Wang, Fengqi Li, Haiming Wei, Zhe-Xiong Lian, Rui Sun, and Zhigang Tian

Dysfunctional CD8+ T cells in hepatitis B and C are characterized by a lack of antigen-specific


T-bet induction
Peter D. Kurktschiev, Bijan Raziorrouh, Winfried Schraut, Markus Backmund, Martin Wchtler, Clemens-Martin Wendtner,
Bertram Bengsch, Robert Thimme, Gerald Denk, Reinhart Zachoval, Andrea Dick, Michael Spannagl, Jrgen Haas,
Helmut M. Diepolder, Maria-Christina Jung, and Norbert H. Gruener

Interplay between regulatory T cells and PD-1 in modulating T cell exhaustion and viral control during
chronic LCMV infection
Pablo Penaloza-MacMaster, Alice O. Kamphorst, Andreas Wieland, Koichi Araki, Smita S. Iyer, Erin E. West, Leigh OMara,
Shu Yang, Bogumila T. Konieczny, Arlene H. Sharpe, Gordon J. Freeman, Alexander Y. Rudensky, and Rafi Ahmed

CCR4 drives ATLL jail break


Kevin Shannon

Gain-of-function CCR4 mutations in adult T cell leukemia/lymphoma


Masao Nakagawa, Roland Schmitz, Wenming Xiao, Carolyn K. Goldman, Weihong Xu, Yandan Yang, Xin Yu,
Thomas A. Waldmann, and Louis M. Staudt

Enhancement of an anti-tumor immune response by transient blockade of central T cell tolerance


Imran S. Khan, Maria L. Mouchess, Meng-Lei Zhu, Bridget Conley, Kayla J. Fasano, Yafei Hou, Lawrence Fong,
Maureen A. Su, and Mark S. Anderson

T cellderived interleukin (IL)-21 promotes brain injury following stroke in mice


Benjamin D.S. Clarkson, Changying Ling, Yejie Shi, Melissa G. Harris, Aditya Rayasam, Dandan Sun, M. Shahriar Salamat,
Vijay Kuchroo, John D. Lambris, Matyas Sandor, and Zsuzsanna Fabry
THE
JOURNAL OF
EXPERIMENTAL
MEDICINE
Executive Editor Advisory Editors Tomohiro Kurosaki Ethan M. Shevach
Marlowe S.Tessmer Shizuo Akira Bart N. Lambrecht Roy L. Silverstein
phone (212) 327-8575 Kari Alitalo Klaus F. Ley Jonathan Sprent
email: jem@rockefeller.edu Frederick W. Alt Yong-Jun Liu Janet Stavnezer
Senior Editor K. Frank Austen Clare Lloyd Andreas Strasser
Heather L. Van Epps Albert Bendelac Tak Mak Stuart Tangye
Michael J. Bevan Bernard Malissen Steven L.Teitelbaum
Scientific Editors
Christine A. Biron James S. Malter Thomas J.Templeton
Teodoro Pulvirenti
Christian Bogdan Philippa Marrack Kevin J.Tracey
Catarina Sacristn
Hal E. Broxmeyer Diane Mathis Giorgio Trinchieri
Editors Meinrad Busslinger Ira Mellman Shannon Turley
Jean-Laurent Casanova Arturo Casadevall Matthias Merkenschlager Marcel R.M. van den Brink
Douglas T. Fearon Ajay Chawla Sean J. Morrison Ulrich von Andrian
David Holtzman Yongwon Choi Muriel Moser Harald von Boehmer
Lewis L. Lanier Robert L. Coffman Christian Mnz Christopher M.Walker
William A. Muller Daniel J. Cua Cornelis Murre Raymond M.Welsh
Carl Nathan Myron I. Cybulsky Benjamin G. Neel E. John Wherry
Michel Nussenzweig Riccardo Dalla Favera Michael Neuberger Linda S.Wicker
Anne OGarra Glenn Dranoff Victor Nussenzweig Ian Wilson
Alexander Rudensky Michael Dustin John J. OShea Thomas Wynn
Alan Sher Vincent A. Fischetti Paul H. Patterson
Sasha Tarakhovsky Richard A. Flavell Fiona Powrie
Andreas Trumpp Adolfo Garcia-Sastre Lluis Quintana-Murci Monitoring Editors
David Tuveson Patricia Gearhart Klaus Rajewsky Marco Colonna
Editor Emeritus Ronald N. Germain Gwendalyn J. Randolph Jason Cyster
Alan N. Houghton Christopher Goodnow Jeffrey Ravetch Stephen Hedrick
Siamon Gordon Sergio Romagnani Kristin A. Hogquist
Manuscript Coordinator Or Gozani Nikolaus Romani Andrew McMichael
Sylvia F. Cuadrado Sergio Grinstein David L. Sacks Luigi Notarangelo
phone (212) 327-8575
email: jem@rockefeller.edu Philippe Gros Shimon Sakaguchi Anjana Rao
Kristian Helin Matthew D. Scharff Federica Sallusto
Preflight Editor Chyi Hsieh Olaf Schneewind Louis M. Staudt
Rochelle Ritacco Christopher A. Hunter Stephen P. Schoenberger Toshio Suda
Assistant Production Editor Kayo Inaba Hans Schreiber
Shauna OGarro Gerard Karsenty Gerold Schuler
Jay Kolls Robert A. Seder Consulting
Production Editor
Paul Kubes Rafick-P. Skaly Biostatistics Editors
Brianna Caszatt
Vijay K. Kuchroo Charles N. Serhan Glenn Heller
Production Manager Ralf Kuppers Nilabh Shastri Madhu Mazumdar
Camille Clowery
Production Designer
Erinn A. Grady

Copyright to articles published in this journal is held by the authors. Articles are published by
The Rockefeller University Press under license from the authors. Conditions for reuse of the
articles by third parties are listed at http://www.rupress.org/terms

Print ISSN 0022-1007 Online ISSN 1540-9538


INSIGHTS

Bee venom stirs up buzz in antigen presentation


While most T cells recognize peptide antigens presented by major histocompatibility complex (MHC) class I
and class II molecules, some T cells react with lipid antigens presented by MHC-related CD1 proteins. In this
issue, Bourgeois et al. establish the concept that enzymes in bee venom can cleave host skin-derived phospho-
lipids into lipid neoantigens that activate CD1a-restricted T cells to promote a local inflammatory response.
Humans have four CD1 proteins: CD1a, CD1b, CD1c, and CD1d. The CD1a molecule has long been
known as a specific surface marker for human Langerhans cells, and recent studies have shown that CD1a-
restricted T cells are recruited to the skin. Such T cells are intrinsically autoreactive, recognizing skin-specific
oils produced by sebaceous glands. Insight from Luc
In searching for potential contributions of CD1a-reactive T cells to inflammatory responses in the Van Kaer
skin, Bourgeois et al. investigated human T cell responses to the venoms of bees and wasps. They found
that such venoms induce a CD1a-mediated response in both individual T cell clones and primary human T cells. However,
further analyses revealed that the antigenic substance unex-
pectedly partitioned within the lipid rather than the protein
fractions, raising the possibility that enzymes in the venom
generate lipid antigens by cleaving common phospholipids.
The investigators were able to zoom in on phospholipase
A2 (PLA2), an enzyme that is abundant in insect and snake
venoms, which acts on phospholipids to release fatty acids
and lysophospholipids.
These intriguing findings support a model of T cell acti-
vation involving insect-mediated PLA2 introduction into the
skin, inducing the release of fatty acid neoantigens from com-
mon skin phospholipids, subsequently sampled by Langerhans
cells, loaded onto CD1a molecules, and presented to T cells.
Activated CD1a-reactive T cells produce IL-22, a cytokine
that plays a role in antimicrobial defenses, promotes keratino-
cyte proliferation, and contributes to the pathogenesis of a
variety of skin inflammatory diseases. In this manner, CD1a-
reactive T cells may contribute to the local inflammatory
response to venoms. The findings also provide a potential
molecular explanation for the generation of autoantigens in
normal skin, which expresses secreted phospholipases. It has
been proposed that such natural lipids are present at the surface
Phospholipase A2 (PLA2) in bee venom is introduced into human skin
and cleaves ubiquitous phospholipids into free fatty acids and lyso-
of the skin and are thus inaccessible to CD1a-restricted T cells
phospholipids that function as neoantigens. These fatty acid neoan- in the dermis. However, a breach in the skin barrier might
tigens are now available for sampling by epidermal Langerhans cells, make these natural oils available to Langerhans cells in the epi-
which load them onto CD1a proteins and present them at their surface dermis, resulting in their presentation to CD1a-reactive T cells
to skin-resident CD1a-reactive T cells. These T cells produce cytokines and the subsequent induction of antimicrobial and inflam-
such as IL-22 that contribute to the inflammatory response. matory responses. Moreover, infections and inflammatory
processes can modulate the expression patterns of phospho-
lipases in the skin, which may represent a mechanism to control inflammation via CD1a-reactive T cells. Finally, certain skin
pathogens secrete PLA2 enzymes, raising the possibility that such infections can induce CD1a-mediated T cell responses that
contribute to host immunity and disease pathogenesis. These proposed scenarios, together with their potential therapeutic
applications, will provide rich and fertile avenues for future investigation.
Bourgeois, E.A., et al. 2015. J. Exp. Med. http://dx.doi.org/10.1084/jem.20141505.

Luc Van Kaer, Vanderbilt University School of Medicine: luc.van.kaer@vanderbilt.edu


Article

Bee venom processes human skin lipids


for presentation by CD1a
Elvire A. Bourgeois,1* Sumithra Subramaniam,2,3* Tan-Yun Cheng,1
Annemieke De Jong,1 Emilie Layre,1 Dalam Ly,1 Maryam Salimi,2,3
Annaliza Legaspi,4,5 Robert L. Modlin,4,5 Mariolina Salio,2,3
Vincenzo Cerundolo,2,3 D. Branch Moody,1** and Graham Ogg2,3**
1Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Womens Hospital, Harvard

Medical School, Boston, Massachusetts, 02114


2MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine and 3University of Oxford NIHR Biomedical

Research Centre, Oxford, Oxfordshire OX3 9DS, England, UK


4Division of Dermatology, David Geffen School of Medicine, 5Department of Microbiology, Immunology and Molecular

Genetics, University of California Los Angeles, Los Angeles, CA 90095

Venoms frequently co-opt host immune responses, so study of their mode of action can
provide insight into novel inflammatory pathways. Using bee and wasp venom responses as
a model system, we investigated whether venoms contain CD1-presented antigens. Here, we
show that venoms activate human T cells via CD1a proteins. Whereas CD1 proteins typically
present lipids, chromatographic separation of venoms unexpectedly showed that stimula-
tory factors partition into protein-containing fractions. This finding was explained by
demonstrating that bee venomderived phospholipase A2 (PLA2) activates T cells through
generation of small neoantigens, such as free fatty acids and lysophospholipids, from
common phosphodiacylglycerides. Patient studies showed that injected PLA2 generates
lysophospholipids within human skin in vivo, and polyclonal T cell responses are dependent
on CD1a protein and PLA2. These findings support a previously unknown skin immune
response based on T cell recognition of CD1a proteins and lipid neoantigen generated
in vivo by phospholipases. The findings have implications for skin barrier sensing by T cells
and mechanisms underlying phospholipase-dependent inflammatory skin disease.

CORRESPONDENCE Extensive evidence for the important role of of autoimmunity start with protein and peptide
D. Branch Moody: peptideMHC complexes in T cell activation antigen vaccination. However, the discovery of
bmoody@partners.org
evolved into a widespread belief that peptides are the function of CD1a, CD1b, CD1c, and CD1d
OR
Graham Ogg: the only common and natural target of human proteins (McMichael et al., 1979; Calabi and
graham.ogg@ndm.ox.ac.uk T cell responses. Therefore, until recently, nearly Milstein, 1986) as antigen-presenting molecules
all human clinical studies of T cell action in au- expands the biochemical spectrum of natural
Abbreviations used: APC, anti-
gen presenting cells; DDM, toimmune, allergic, and infectious diseases were antigens for T cells to include many types of lip-
dideoxymycobactin; LC, targeted at peptide antigens. For example, most ids (Porcelli et al., 1989, 1992; Kronenberg and
Langerhans cells; mDC, candidate antigens for human T cellmediated Kinjo, 2005).
monocyte-derived DC; PC,
phosphatidylcholin; PLA2,
autoimmune diseases are proteins (Klein et al., CD1 proteins are conserved among mam-
phospholipase A2. 2014). Subunit vaccines (Tameris et al., 2013) mals (Kasmar et al., 2009) and are expressed at
and diagnostic tests (Lalvani and Pareek, 2010) high density on thymocytes and professional
rely on defined peptide motifs, and mouse models APCs in the periphery, including Langerhans
cells (LCs), B cells, macrophages and myeloid
*E.A. Bourgeois and S. Subramaniam contributed equally to DCs (Dougan et al., 2007). In cells, CD1 pro-
this paper. teins bind and display hundreds of molecular
** D.B. moody and G. Ogg contributed equally to species of self-sphingolipids, phospholipids, and
this paper.
A. De Jongs present address is Columbia University, Depart- 2015 Bourgeois et al. This article is distributed under the terms of an
ment of Dermatology, New York, NY 10032. AttributionNoncommercialShare AlikeNo Mirror Sites license for the first
D. Lys present address is University Health Network, Uni- six months after the publication date (see http://www.rupress.org/terms). After
six months it is available under a Creative Commons License (Attribution
versity of Toronto, Department of Immunology, Toronto, NoncommercialShare Alike 3.0 Unported license, as described at http://creative-
Ontario M5G 1L7, Canada. commons.org/licenses/by-nc-sa/3.0/).

The Rockefeller University Press $30.00


J. Exp. Med. 2015 Vol. 212 No. 2 149163 149
www.jem.org/cgi/doi/10.1084/jem.20141505
acylglycerides (Huang et al., 2011), and >20 types of stimula- In addition, skin-specific oils, including squalene, function as
tory lipid antigens for T cells are now known (Young and autoantigens presented by CD1a proteins (de Jong et al., 2014).
Moody, 2006). The molecular bases by which lipids are rec- Venoms act within the skin to cause pain or death by
ognized by T cells are well established through crystal struc- transfer to highly unrelated organisms; they involve multiple
tures of CD1, CD1 bound to lipid, and CD1-lipid bound to active substances that typically function by coopting host
a TCR (Zeng et al., 1997; Gadola et al., 2002; Borg et al., responses and thus have a long history as useful probes of nat-
2007). The alkyl chains of lipids are sequestered within the ural host inflammatory responses. In particular, venom-induced
grooves of CD1 proteins, allowing the carbohydrate, sulfate, immune responses have been relevant beyond venom re
phosphate, and other polar elements to protrude and interact activity and have provided broader immunological insights
with TCRs. in to novel pathways of inflammation and tolerance (Bil and
Despite the wealth of molecular and cell biological data Bonifazi, 2008; Aslam et al., 2010; Gutierrez and Rodewald,
showing that mammalian T cells can recognize lipids, 2013). Because bee and wasp venom responses are localized
translational research to determine the roles of CD1-restricted initially in skin, where CD1a is so abundantly expressed, and
lipid antigens in vivo or during diseases that are commonly because the lipidic content of the bee and wasp venoms was
seen by physicians is limited. Most reported studies have focused unexplored, we sought to use bee and wasp venom as a model
on CD1d and CD1d-restricted T cells, known as NKT cells, system to investigate novel pathways of inflammation in the
because CD1d proteins are the only CD1 isoform expressed in skin. After initial studies documented a CD1a-mediated re-
commonly used mouse models (Godfrey and Rossjohn, 2011). sponse in individual T cell clones and among cohorts of pa-
Yet CD1a, CD1b, and CD1c differ from CD1d proteins, and tients, mechanistic studies tracked the antigenic substance to
from one another, in their trafficking and tissue distribution, venom proteins. By investigating venom-derived proteins,
suggesting that they exert different physiological roles (Kasmar we discovered an unexpected mechanism by which venom
et al., 2009). Notably, CD1a, unlike CD1b and CD1c pro- phospholipases create lipid neoantigens in vivo providing a
teins, has been known for decades as a phenotype-specific new view of venom responses and insight into the role of
marker of human epidermal LCs (Dougan et al., 2007). In phospholipases in CD1a biology.
addition to studies of guinea pigs (Hiromatsu et al., 2002a,b)
and transgenic mice (Felio et al., 2009), the functions of RESULTS
CD1a, CD1b, and CD1c proteins have been studied in humans Responses of T cells to CD1 molecules and venom
during tuberculosis (Moody et al., 2000) and seasonal allergy Stinging insects in the genera vespula (wasp) and apis (bee)
(Agea et al., 2005), and as in other pathogens that infect humans generate a local intradermal inflammatory response. To in-
(Zeissig et al., 2010). However, human studies rely mostly on vestigate a possible role of CD1, we obtained CD3+ T cells
T cell clones whose functions change over time during in vitro from healthy adult individuals and mixed them with wasp
culture and may not represent the natural populations of T cells venom-treated and irradiated K562-CD1 APCs, which were
in vivo. mock transfected or transfected with either CD1a, CD1b,
To study polyclonal autoreactive human T cells ex vivo, CD1c, or CD1d. As expected based on prior work (de Jong
APCs that lack detectable surface expression of MHC proteins et al., 2014), we observed low but detectable alloreactive back-
(K562 cells) were engineered to express CD1a, CD1b, CD1c, ground responses of 1 in 2,000 T cells after 1214 d in cul-
or CD1d proteins at high density (K562-CD1). K562-CD1 ture to mock-transfected target cells in donor C1175 (Fig. 1 A,
cells provide lipid autoantigens, and such MHClow CD1high left), which is representative of three donors. In the absence
APCs largely avoid MHC alloreactivity (de Jong et al., 2010). of venom, T cells showed responses to K562-CD1a cells at
Therefore, T cells from groups of randomly chosen or un- rates higher than to mock transfected cells or cells expressing
related donors can be tested for antigen responses, allowing other CD1 isoforms, confirming the presence and relatively
cohort studies and quantitation of CD1 autoreactivity in high rates of CD1a autoreactive T cells seen previously (de Jong
healthy subjects. Three recent studies show that CD1a auto- et al., 2010, 2014; de Lalla et al., 2011). Further, polyclonal re-
reactive T cells are present in the peripheral blood of nearly sponses to wasp venom above background levels were detected,
all humans tested, defining CD1a autoreactive T cells as a dis- but were seen only when using CD1a-expressing cells as tar-
tinct component of the human immune system (de Jong et al., gets (P < 0.05, Fig. 1 A, left, representative of three donors).
2010, 2014; de Lalla et al., 2011). These studies also found These responses demonstrate that a T cell response to wasp
that the rates of T cell autoreactivity are higher to CD1a, as venom is restricted by CD1a proteins.
compared with CD1b, CD1c and CD1d. In addition to stud- Apis mellifera (honeybee or bee) venom also causes skin
ies of peripheral blood, recent studies implicate CD1a auto- immune responses and shares certain mechanisms of immune
reactive T cells as functioning in organ-specific immunity in action with wasp venom (Mller et al., 2009). For example,
the skin. CD1a is the major human CD1 protein expressed at patients that develop hypersensitivity toward wasp venom can
high density on LCs (Dougan et al., 2007), and CD1a auto- also show reactivity to bee venom (Egner et al., 1998).Therefore,
reactive T cells express skin-specific homing receptors (cuta- we next investigated the CD1-restricted T cell response to
neous lymphocyte antigen, CCR4, CCR6, and CCR10) bee venom and found a pattern of response that was similar to
and home to the skin in large numbers (de Jong et al., 2010). that of wasp venom, including augmentation of the baseline

150 Bee venom generates CD1a ligands | Bourgeois et al.


Ar ticle

Figure 1. Vespula spp and Apis mellifera venoms induce a preferential activation of CD1a-restricted T cells among the group1-CD1
reactive cells. (A) T cells were isolated by CD3 MACS beads from healthy donor PBMCs and cultured for 1214 d with IL-2 and irradiated K562
cells transfected with CD1a (K562-CD1a), CD1b (K562-CD1b), CD1c (K562-CD1c), CD1d (K562-CD1d), or an empty vector (K562) in the presence
of venom. CD1 reactivity was then examined by IFN- ELISpot with transfected or untransfected K562 cells either in the presence or absence of
Vespula spp or Apis mellifera venoms. Representative data for one donor (C1175) of three are shown. (B) The CD1a-restricted T cell response in
the presence or absence of anti-CD1a (donor C1098) and Vespula spp and Apis mellifera venoms. CD1a-restricted, venom-specific responses were
measured in 21 donors for Vespula spp venom (C) and Apis mellifera venom (D). mDC or in vitro LClike cells derived from CD14+ cells were pulsed
with 1 g/ml wasp venom (E) or bee venom (F) and incubated with the T cells in the presence or absence of anti-CD1a antibody or isotype control.
IFN- production was measured by IFN- ELISpot. Representative data for one donor (C556 [E] and C560 [F]) of three are shown. Data were mean
of triplicate measurements SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

JEM Vol. 212, No. 2 151


autoreactive response by venom in the presence of CD1a bee venoms to lymphocytes resulted in apparent toxicity and
(Fig. 1 A, right). Thus, this pilot study provided proof of prin cell death based on monitoring of T cells by Trypan blue
ciple for CD1a-mediated human cellular responses to wasp exclusion. We could not generate long-term T cell lines by
and bee venom among human polyclonal T cells. direct and repeated exposure to venom in vitro. Instead, we
The total IFN- response of fresh polyclonal T cells con- screened existing panels of CD1a-reactive T cell clones for
sisted of three identifiable components: a response to K562, responses to Vespula species (wasp) and Apis mellifera (bee)
an increment in response to CD1a and a further increment to venoms. To minimize toxicity and interrogate the mecha-
added venom, consistent with CD1a presentation of venom nisms of venom action, we switched from K562-CD1a acti-
antigens. Expansion of T cells with antigen ex vivo offered vation assays (cellular assays) and took advantage of assays in
the opportunity to more definitively establish the molecular which biotinylated CD1a proteins are coated onto streptavi-
mechanisms involved. We cultured T cells in IL-2 for 2 wk din plates (plate assays). In the plate assay, CD1a proteins are
in the presence of wasp venomtreated K562-CD1a cells to pulsed with venom-derived products, washed and then ex-
permit the outgrowth of responder cells. ELISpot studies posed to T cells. This method diminishes venom exposure
clearly confirmed reactivity to CD1a alone, as well as to CD1a to APCs and responding cells, thereby minimizing toxicity.
plus wasp venom, but no response to venom was seen in the Also, the plate method might favor detection of stimulants
absence of CD1a (Fig. 1 B, left, donor C1098). Further, anti- that act by forming complexes with CD1 proteins in prefer-
CD1a mAb blocked the response to background levels (P < ence to indirect effects of venom on APCs or T cells that do
0.05), proving that CD1a is required. A similar pattern of re- not involve binding to CD1 proteins. The T cell clone BC2
sponse was seen to T cells treated with K562-CD1a and bee showed a dose-dependent increase in IFN- secretion when
venom (Fig. 1 B, right). cultured in the presence of CD1a proteins pretreated with
wasp venom (Fig. 2 A). The response was likely to be specific
CD1a-restricted venom responses in a healthy cohort antigen recognition rather than a general mitogenic activa-
Next, we sought to test this pattern of wasp venom responses tion, because no response was seen when plates were coated
in a larger cohort of 21 healthy individuals. Measuring mean with CD1b, CD1c (Fig. 2 A), or CD1d (not depicted), or
IFN- spots for all donors, we observed higher (P < 0.0001) when venom-treated CD1 proteins were exposed to another
response to CD1a transfectants, as compared with untrans- T cell line, CD8-2, which normally recognizes dideoxymy-
fected target cells (Fig. 1 C). Similar to the pilot studies, we cobactin lipopeptides presented by CD1a (Moody et al.,
detected higher spot numbers when wasp venom was added, 2004; Fig. 2 B). Furthermore, we observed a similar dose-
but only when CD1a was present (P < 0.001). We observed dependent recognition by T cell clone BC2 of bee venom in
a pattern of IFN- production to bee venom that was depen- the presence of CD1a proteins (Fig. 2 C).
dent on CD1a expression (P < 0.001) and similar in magni- Using cellular K562 assays, we again observed a CD1-
tude to that seen with wasp venom (Fig. 1 D). dependent and dose-dependent response to bee venom, but
K562 cells are transformed cells that represent useful tools this time with significant CD1a autoreactivity. The higher
which bypass MHC alloreactivity. The two types of primary responses seen in cellular assays might be explained if venom
human skin APCs that express CD1a proteins are myeloid acts on the APC in some way or if lipid autoantigens are pro-
DCs and LCs. To investigate whether these cell types mediate vided by K562 cells (Fig. 2 D). In addition to using monocyte-
the venom response similarly to K562 cells, we differentiated derived DCs, we also aimed to use an alternative system for
monocytes with GM-CSF and IL-4 to produce monocyte- the generation of LC-like cells and so sorted CD34+ pro-
derived DCs (mDCs), which mimic key features of myeloid genitors and activated them with relevant cytokines, which
DCs. In addition we activated cells in vitro with cytokines to are well established to serve as a model of LC function (Hunger
mimic LCs (in vitro LCs), including high CD1a expression et al., 2004). The cells express CD1a at moderate to high
(Porcelli et al., 1992; Sallusto and Lanzavecchia, 1994; Caux levels (unpublished data; Porcelli et al., 1992; Sallusto and
et al., 1997; Geissmann et al., 1998). Both types of human Lanzavecchia, 1994; Yakimchuk et al., 2011) and, when added
primary APCs were able to mediate wasp venom (Fig. 1 E) to cultures, mediated CD1a- and venom-dependent responses
and bee venom (Fig. 1 F) responses, which were abrogated in of BC2 at a ratio of 1 APC to 25 T cells (Fig. 2, E and F).
the presence of anti-CD1a. Overall, this study showed that Thus, both transformed and primary CD1a+ cells were used
CD1a autoreactivity and CD1a-dependent venom reactivity to formally show that T cells can be activated by bee and
are common in humans. This pattern of combined CD1a wasp venom, and the primary cells validated BC2 as a re-
autoreactivity and antigen-dependent reactivity is reminiscent agent that mimics the pattern of patient responses seen ex vivo
of invariant NKT cells, which are activated weakly by CD1d (Fig. 1). Thus, this T cell clone could be used for the subse-
and strongly by CD1d with lipid antigen (Kawano et al., 1997). quent assays needed to isolate and identify stimulatory mole-
cules in venom.
Isolation of stimulatory factors from venom
To neutralize diverse predators or prey, natural venoms are Fractionation of venom to discover antigens
usually high potency toxins that have multiple mechanisms of To fractionate natural venoms and discover the stimulatory
action (King et al., 2003). Repeated exposure of wasp and substance, we treated Apis mellifera (bee) and a mix of Vespula

152 Bee venom generates CD1a ligands | Bourgeois et al.


Ar ticle

Figure 2. The autoreactive CD1a-restricted T cell clone BC2 is activated by wasp and bee venoms. Biotinylated CD1a, CD1b, and CD1c and
CD1d proteins were coated on streptavidin plates. After washing, wasp (A and B) and bee (C) venoms and dideoxymycobactin (DDM; B) were added to the
wells for 2448 h. After washing, BC2 cells (A and C) or CD8.2 cells (B) were added, and the supernatants were collected 24 h later and IFN- quantified.
(D) BC2 cells were co-cultured for 24 h with universal target cells (K562) as APCs, transfected with CD1a (K562-CD1a) or empty vector (K562) and bee
venom. IFN- in supernatants was measured by ELISA. Data are representative of three separate experiments. (E) mDCs and (F) CD34-derived LC-like cells
were incubated with IgG control or anti-CD1a antibodies for 1 h at 37C. Bee venom was then added at 2 g/ml for 1 h. BC2 cells were then added at a
1:25 APC/T cell ratio. Two representative donors out of three are shown.

species (wasp) venoms with chloroform and methanol to BC2, and it suggested that multiple compounds within the
create a two-phase mixture. Lipids partition into the organic venom acted in concert to generate T cell response. An analo-
solvents (extract), whereas proteins, nucleotides and other gous experiment performed using the K562-CD1a cellular
polar molecules are excluded (precipitate; Fig. 3 A). Nearly assay showed different results, which provided possible ex-
all CD1-presented antigens are lipids, so it was surprising planations for the unexpected pattern. In contrast to the
that the lipidic extract failed to activate BC2 cells (Fig. 3 B). plate assay, venom precipitate applied to intact K562-CD1a
Likewise, the protein-enriched precipitate failed to activate APCs was sufficient to activate T cells. Again, extracted lipids
T cells. However, T cell activation was detectable when the alone failed to activate BC2 (Fig. 3 C). These seemingly
extract and precipitate were recombined. This result ex- contradictory results might be explained if venom does not
cluded the possibility that chloroform and methanol some- provide an activated lipid antigen, but instead provides a pro-
how destroyed the capacity of the whole venom to activate tein that precipitates in organic solvents and then is capable

JEM Vol. 212, No. 2 153


Figure 3. The co-incubation of both
proteins and lipids from wasp and bee
venoms triggers BC2 cells activation when
presented by CD1a protein. (A) Protein and
lipid fractions of bee and wasp venoms were
separated by organic solvent extraction using
chloroform and methanol. (B) CD1a proteins
were coated on streptavidin plates. After
washing, wasp or bee venoms and their ex-
tracts were added to the wells for 24 to 48 h.
After washing, BC2 cells were added, superna-
tants were collected after 24 h and IFN-
quantified. (C) BC2 cells were co-cultured
with universal target cells transfected with
CD1a (K562 CD1a) or empty vector (K562) and
bee venom or their extracts. Supernatants
were collected after 24 h. IFN- in superna-
tants was measured by ELISA. Data are repre-
sentative of three separate experiments.

of producing antigenic substances when combined with PLA2 releases large amounts of lipid mediators of inflamma-
venom-derived or K562-derived lipids. tion through downstream action of cyclooxygenases, which
create arachidonic acid metabolites. Therefore, testing an-
Phospholipase A2 activates T cells other candidate mechanism of action relating to PLA2, we
In considering protein factors that might generate antigens hypothesized that free arachidonic acid might bind to CD1a
for CD1a proteins, it is well established that wasp and bee proteins as an antigen or act indirectly as cyclooxygenase sub-
venoms contain phospholipase A1 (PLA1) and A2 (PLA2). strate to stimulate BC2 via receptors other than the TCR.
Phospholipases are major components of venom that have However, we observed no arachidonic acidmediated stimu-
been extensively studied in regard to their antigenicity toward lation and saw dose-dependent inhibition of BC2 activation
MHC-restricted T cells and B cells (Aslam et al., 2006; Sin by arachidonic acid in K562 cellular and plate assays when
et al., 2011). Viral phospholipases can promote activation of added at high absolute dose (Fig. 4 C), ruling out both hy-
NKT cells by CD1d (Zeissig et al., 2012). Based on our potheses. Although arachidonic acid could inhibit responses
preliminary experiments with recombined protein and lipid at pharmacological doses, PLA2 had a reproducible and strong
fractions, we hypothesized that phospholipases act on venom- stimulatory effect, suggesting that the smaller amounts of ara-
derived lipids (Fig. 2) or the K562 membrane phosphodi chidonic acid liberated by this enzyme do not have a domi-
acylglycerols (Fig. 3) cleaving ester bonds and generating free nant negative effect. Finally, mDC (Fig. 4 D) and cord blood
fatty acids, and lysophospholipids. Purified venom PLA2 from CD34-derived LC-like cells (Fig. 4 E) act as APCs and sub-
Apis mellifera strongly activated BC2 T cells (Fig. 4 A). No T cell strates of the PLA2 to activate BC2 cells in a CD1a-dependent
activation was seen when PLA2 was exposed to T cells with manner, suggesting that neo-antigens can be generated in pri-
K562 cells lacking CD1a (Fig. 4 A).Therefore, the mechanism of mary APCs and that neither mDCs nor LCs express inhibi-
PLA2 on T cells is indirect and requires CD1a proteins. tory factors.
To directly test the hypothesis that bee venom PLA2 acts
Mechanism of PLA2 action by cleaving intact cellular phospholipids to create neoanti-
Another well-known polypeptide present in bee venom that gens, we preincubated the enzyme with two synthetic sub-
binds lipids and enhances PLA2 activity is mastoparan strates for bee venom PLA2, phosphatidylcholine comprised
(Argiolas and Pisano, 1983; Cabrera et al., 2011). However, of singly unsaturated C18 fatty acyl chains (PC 18:1/18:1)
mastoparan acting alone (Fig. 4 B) did not activate BC2. and phosphatidic acid with C16 fatty acyl chain in sn-1 and

154 Bee venom generates CD1a ligands | Bourgeois et al.


Ar ticle

Figure 4. Bee venom PLA2 alone induces a CD1a-restricted T cell response by releasing free fatty acids. BC2 cells were co-cultured for 24 h
with universal target cells (K562) transfected with CD1a (K562 CD1a) or empty vector (K562) and bee venom or bee venom PLA2 (A), mastoparan (B), or
arachidonic acid (C). IFN- in supernatants was measured by ELISA. In (C) right panel, biotinylated CD1a proteins were coated on streptavidin plates. After
washing, bee venom or arachidonic acid were added to the wells for 24 to 48 h. After washing, BC2 cells were added for 24 h, and supernatant was sam-
pled for IFN- quantification by ELISA. Data are representative of three separate experiments. (D) mDC and (E) CD34-derived LC-like cells were incubated
with IgG control or anti-CD1a antibodies for 1 h at 37C. PLA2 was then added at 2 g/ml for 1 h. BC2 cells were then added at a 1:25 APC:T cell ratio.
Two representative donors out of three are shown.

unsaturated C18 fatty acyl chain in sn-2 (PA 16:0/18:1). We and antigenic fatty acid, in agreement with a recent study
also tested for a T cell response to products of cleavage by identifying free fatty acids as CD1-presented antigens (de Jong
PLA2 and control lipids (Fig. 5 A), including purified free et al., 2014).
fatty acids and lysophospholipids: lysophosphatidylcholine
(LPC 18:1), oleic acid (FA 18:1), lysophosphatidic acid (LPA Venom generates lysophospholipids in vivo
18:1), and palmitic acid (FA 16:0). Higher production of Membrane phospholipids and free fatty acids are both abun-
IFN- was obtained in response to phospholipids when prein- dant in blood and tissue. However phospholipids largely
cubated with PLA2. BC2 T cells responded to fatty acids to a self-assemble into bilayer membrane structures, and free
greater extent than intact phospholipids or lysophospholipids. fatty acids exist mainly as cytosolic metabolites or lipopro-
This result suggests that PLA2 activates CD1a-restricted T cells tein structures generated during gastrointestinal absorption.
by cleaving nonantigenic phospholipids into lysophospholipids Thus, lysophospholipids and free fatty acids are normally

JEM Vol. 212, No. 2 155


Figure 5. Wasp venom phospholipase is active
in vivo in the skin. (A) Biotinylated CD1a proteins were
coated on streptavidin plates. After washing, lipids were
added at 10 g/ml for 2448 h. Lipids tested were as
follows: phosphatidylcholine 18:1 (PC18:1), lysophos-
phatidylcholine 18:1 (LPC18:1), and oleic acid 18:1 (FA
18:1; left) or phosphatidic acid 16:0 (PA16:0), lysophos-
phatidic acid 18:1 (LPA18:1), and palmitic acid 16:0
(FA16:0; right). Phosphatidylcholine and lysophospha-
tidic acid were preincubated or not with PLA2 at 50 g/ml
for 1 h at 37C. 24 h later, wells were washed and
BC2 cells were added. Supernatants were collected after
24 h. IFN- in supernatants was measured by ELISA.
Data are representative of three separate experiments.
(B) Healthy donor epidermis was injected with 10 g of
wasp venom or saline. Skin blisters were raised and, 24 h
later, the blister fluid was sampled. Lipids were extracted
with chloroform, methanol, and water using the Bligh-
Dyer method and analyzed on a quadruple time of flight
mass spectrometer. Positive mode EIC-MS detected ions
at m/z 758.5686, 786.6000, 810.5994, and 834.5993,
corresponding to the homologous series of phosphati-
dylcholine C42H81NO8P+ (16:0/18:2), C44H85NO8P+
(18:0/18:2), C46H85NO8P+ (18:2/20:2), and C48H85NO8P+
(20:4/20:2). Positive mode EIC-MS detected ions at m/z
496.3374, 524.3683, and 544.3370, corresponding to
the homologous series of lysophosphatidylcholine
C24H51NO7P+ (16:0), C26H55NO7P+ (18:0), and C28H51NO7P+
(20:4). Data from one representative donor out of two
are shown.

stored in macromolecular clusters and do not reach high con- using suction cup blisters (Salimi et al., 2013). This method
centration as free molecules in the extracellular space, where uses low pressure, sustained (60 min) suction to produce
CD1a lipid loading is thought to occur (Manolova et al., extracellular blister fluids that are captured for immunological
2003). Furthermore, although extracellular lipids, including and biochemical analysis before or after antigen challenge
fatty acids, accumulate in the stratum corneum and sebaceous (Salimi et al., 2013).
glands, they are not detectable in normal dermis. Accordingly, After injection of 10 g of venom, which mimics the
most models of self- or altered self-lipid display emphasize approximate volume and dose of a wasp sting, or saline vehi-
release of free fatty acids and other antigens into the extracel cle, 2 mm into the skin at two sites in the arm of one pa-
lular space in the proximity of CD1+ epidermal LC (Colonna, tient, we obtained two blister fluid samples. After extracting
2010; de Jong et al., 2010, 2014). Injection of bee and wasp the lipids from blister fluids using a mixture of chloroform,
venoms into skin might locally generate such cleavage of in- methanol and water, we assessed for the presence of sub-
tact phosphodiacylglycerides to lysolipids and free fatty acids, strates and products of the wasp venom PLA by mass spec-
so we tested this in a human model for skin immune responses trometry using sensitive quadrupole time of flight (QToF)

156 Bee venom generates CD1a ligands | Bourgeois et al.


Ar ticle

methods (Fig. 5 B). Focusing on the main natural substrate data suggest that both wasp and bee venom phospholipases
for PLA2, we monitored the phosphatidylcholine series for can generate common CD1a fatty acid ligands.
key ions corresponding to lipids with differing fatty acid con-
tent: C42H81NO8P+ (16:0/18:2), C44H85NO8P+ (18:0/18:2), PLA2 induced polyclonal responses ex vivo
C46H85NO8P+ (18:2/20:2), and C48H85NO8P+ (20:4/20:2; Having identified PLA2 as sufficient to generate CD1a-
Fig. 5 B). The masses of detected ions (m/z 758.5686, presented antigens in vitro using T cell clones, we next sought
786.6000, 810.5994, and 834.5993) matched these PC vari- to determine if bee venom PLA2 is sufficient to activate poly-
ants within the mass accuracy of the detector. Comparing clonal T cells ex vivo in cellular assays (Fig. 7). As with whole
saline- and venom-injected sites, we found a twofold lower venoms (Fig. 1), we detected higher IFN-producing cells
intensity for signals corresponding with all molecular variants with the addition of 100 ng/ml bee venom PLA2 but only in
of PC in the PLA2-injected skin samples, indicating that the the presence of CD1a and not other CD1 isoforms (Fig. 7 A,
enzyme substantially consumed local PC (Fig. 5 B). In a sec- left). We observed significantly (P < 0.001) higher CD1a-
ond patient, signals were reduced after venom injection for restricted responses in the presence of PLA2 in a cohort of 18
all PC species except PC18:1 18:1 (unpublished data). Con- donors (Fig. 7 A, right). More detailed testing of an individual
versely, ions matching the lysophosphatidylcholine series, responder showed that anti-CD1a antibodies prevented the
C24H51NO7P+ (16:0), C26H55NO7P+ (18:0), and C28H51NO7P+ response to CD1a and PLA2 (P < 0.05), confirming the es-
(20:4), were readily detected in wasp venom blister fluid with sential role of CD1a (Fig. 7 B, left).
high signal but were not detected in the blister fluid from
saline injection sites. Thus, lysolipids derive from PLA itself PLA2 is the essential component of venom
and not some other aspect of the suction blister model. We for CD1a ligand generation
measured signals corresponding to the expected masses of Collectively, these studies showed that PLA2 was sufficient
free fatty acids in the positive and negative mode in these to recapitulate the response to venom, so we next sought to
samples, but could not detect them. Mass spectrometry de- determine if PLA2 was necessary, or instead whether the
tection of fatty acids is less sensitive than detection of anionic many known active compounds in venom could generate a
phospholipids, so we cannot derive any direct conclusions response. We observed an inhibition of IFN- production
about fatty acid concentrations in these in vivo experiments. in response to the venoms treated with neutralizing anti-
Nevertheless, these results demonstrate that the injection of bodies that bind PLA2 (P < 0.05; Fig. 7 B, right). Further,
venom alters the local lipid content in ways that are predicted using manoalide we noted a blockade of response to wasp
from the specificity of PLA2 and that the cleavage reactions venom and bee venom PLA2 to background levels seen with
needed to generate fatty acid and lysophospholipid antigens CD1a alone (P < 0.05; Fig. 7 C) and not to lower levels,
do occur. which might be expected in the event of nonspecific toxicity
to cells. Lastly, we showed that monocyte-derived DC and
Direct measure of wasp and bee venom PLA bioactivity LC-like cells derived in vitro, as well as CD1a+ cells isolated
In individual donors (Fig. 1, A and B) and cohorts (Fig. 1, directly ex vivo from skin could induce a response by PLA2-
C and D) and in vitro studies with clones (Figs. 24), we noted specific polyclonal T cells in a CD1a- and PLA-dependent
strikingly similar responses to bee and wasp venom, which manner (Fig. 7 D). We conclude that PLA2 is a necessary
are currently thought to differ in their PLA subtypes, as de- factor involved in venom-dependent activation of CD1a-
scribed above. However if fatty acids are the CD1a-lipid reactive T cells.
ligand, similar immunological responses are expected, as fatty
acids will be generated whether PLA1 or PLA2 cleave acyl DISCUSSION
chains from phospholipids at sn-1 or sn-2, respectively. Inter- Using venoms as a model system, our data identify a new
estingly, PLA2 has been found in venom of the neotropical mechanism of in vivo antigen processing by which phospho-
social wasps Polybia paulista (de Oliveira and Palma, 1998; dos lipases cause local alterations in lipid content and conver-
Santos et al., 2011) and Agelaia pallipes pallipes (Costa and sion of nonantigenic substances to smaller lipids, which have
Palma, 2000). This led us to investigate whether PLA2 activ- CD1-mediated T cell antigenicity. Specifically, we identify
ity is also exerted by Vespula species of wasp venom, and fur- wasp and bee venom phospholipases, which act as the key
thermore whether there were shared specificities between proteins that are necessary and sufficient to cleave common
bee and wasp venom phospholipase. Using sn-2 thiol-labeled cell membrane phosphodiacylglycerides, to release free fatty
PLA2 substrates (arachidonyl thio-PC, heptanoyl thio-PC, acid and lysophospholipids (de Jong et al., 2014). Using the
diheptanoyl thio-PC, and palmitoyl thio-PC), we observed particular clone BC2, we ruled in free fatty acids as neoanti-
PLA2 activity with both bee and wasp venom indicating that gens. However, polyclonal T cells may be responding to
wasp venom contains PLA2 activity in addition to its known other lipids as well, and it is notable that CD1d presents lyso-
PLA1 activity, and that specificities are partially shared with phospholipids to NKT cells (Gumperz et al., 2000; Fox et al.,
bee venom PLA2 (Fig. 6 A). We confirmed the PLA2 activ- 2009; Zeissig et al., 2012). Because the CD1a dependence of
ity of wasp venom could be inhibited by manoalide, a previ- lipid-specific T cell responses is observed in polyclonal T cells
ously known PLA2 inhibitor (Fig. 6 B). Collectively, these and among many unrelated human donors, these data suggest

JEM Vol. 212, No. 2 157


Figure 6. Vespula spp and Apis mellifera venom PLA2
activity. (A) PLA2 activity was detected as free thiol groups
released from thiol-labeled lipid substrates through addition
of DNTB/EGTA and measuring color generation at 415 nm
using Apis mellifera venom with four phosphatidylcholine-
based substrates of varying acyl chain lengths and satura-
tion, but the same headgroup. Vespula spp venom PLA2
activity for substrates diheptanoyl-phosphatidylcholine and
arachidonyl-phosphatidylcholine (P < 0.001 compared with
no enzyme control). (B) Using lipid substrate with a thiol
group at the sn2 position, PLA2 activity in Vespula spp
venom in the presence or absence of manoalide was de-
tected using DNTB/EGTA, which produces a colored precipi-
tate and measured at 415 nm. Data were mean of triplicate
measurements SEM.

that phospholipases are important in CD1a biology, provid- the skin in a process that can be meaningfully mimicked by
ing a potential molecular pathway underlying previous ob- needle injection (Cortellini et al., 2012). Several aspects of
servations of T cell responses to fatty acids, which may be our data fulfill predictions that phospholipase injection might
important for skin barrier sensing and inflammation (de Jong lead to antigen generation beneath cornified epithelia in prox-
et al., 2014). imity to CD1a proteins. Responses to venom and phospholi-
Recent studies of CD1a function have found that CD1a pase are detected most strongly in response to the particular
autoreactive T cells and CD1a proteins are both abundantly CD1 isoform (CD1a) that is most extensively expressed in the
present within adjacent compartments of the skin (de Jong skin. PLA substrates can derive from both venom and cellular
et al., 2010, 2014). In general, there exists a physical separation sources, and PLA2 would presumably have access to venom
of CD1a proteins, which are mainly expressed on LC in the or cell derived phospholipids during a sting. Also, although
epidermis, and natural autoantigens such as fatty acids, wax free fatty acids could not be detected ex vivo by mass spec-
esters, and squalene, which are concentrated more superfi- trometry, phospholipid and lysophospholipid concentrations
cially in the sebum and cornified epithelium. The observa- are demonstrated to change in suction blisters in a process that
tions suggested a near neighbor model in which intact skin is dependent on PLA itself. Thus, bee or wasp stings might
prevents direct contact of autoantigens from CD1a, but skin represent a means to locally remodel lipids and generate neo-
breach through infection or injury deposits antigenic sub- antigens in proximity to CD1a proteins, mimicking a natural
stances more deeply within the skin so that antigens come system of barrier sensing.
into contact with CD1a expressing epidermal LC (Colonna, Venoms injure and thereby neutralize diverse predators
2010; de Jong et al., 2010; Kronenberg and Havran, 2014). and prey through transfer of toxic substances that act on many
Considering bee or wasp sting as a variant of this model, types of animals or insects encountered in the wild. Thus,
phospholipases are normally injected up to 2 mm deep into venoms are typically comprised of many toxic substances

158 Bee venom generates CD1a ligands | Bourgeois et al.


Ar ticle

Figure 7. CD1a-restricted reactivity to


Apis mellifera venom PLA2 is found
among polyclonal T cells from blood of
healthy donors. CD3+ cells were isolated by
magnetic beads from healthy donor PBMC
and cultured for 1214 d with IL-2 and irradi-
ated K562 cells transfected with CD1a (K562
CD1a) or CD1b (K562 CD1b) or CD1c (K562
CD1c) or CD1d (K562 CD1d) or an empty vec-
tor (K562) in the presence of Apis mellifera
venom PLA2. CD1 reactivity was then exam-
ined by IFN- ELISpot with transfected or
untransfected K562 cells either in the pres-
ence or absence of Apis mellifera venom
100 ng/ml PLA2 (A, left). Data from one repre-
sentative donor of three are shown. Venom
PLA2 specific CD1a-restricted T cell responses
were measured in 18 donors (A, right). CD1a-
restricted responses were examined in the
presence or absence of anti-CD1a (B, left) for
donor C1098, and PLA2-neutralizing antibod-
ies (7B, right), and manoalide (specific PLA2
inhibitor) for wasp venom in donor C559
(C, left) and Apis mellifera venom PLA2 in donor
C334 (C, right). Representative results are
shown from three independent experiments.
(D) IFN- production was measured by
co-incubations of PLA2-specific T cells with
mDCs (left) or CD14-derived LC-like DCs
(middle) in the presence or absence of anti-
CD1a for donor C558. IFN- production was
measured from co-incubations of PLA2-specific
T cells and freshly isolated skin CD1a+ cells
in the presence or absence of anti-CD1a or
manoalide (right). Representative data from
3 (left and middle) or 2 (right) separate donors
are shown. Data are mean of triplicate mea-
surements SEM. *, P < 0.05; **, P < 0.01;
***, P < 0.001.

with parallel actions, and act by coopting conserved neuro- divided into 6 groups (I, II, III, V, X, and XII) based on
logical or immunological mechanisms in the stung animal. disulfide bridge structure (Starkl et al., 2013), with bee venom
We speculate that injection of venom PLA represents a means PLA having the closest homology to type III human sPLA
to coopt, through overstimulation, the natural effects of en- (Scott et al., 1990; Valentin et al., 2000). sPLA2 enzymes act
dogenous mammalian PLAs in barrier sensing and immune in lipid digestion, host defense, and inflammation (Murakami
response. Mammalian PLA2 enzymes exist as lysosomal, cyto- and Lambeau, 2013). Other evidence to suggest that mam-
solic, and secreted (sPLA2) forms. The sPLA2 superfamily is malian PLAs might have CD1 or lipid-mediated effects on

JEM Vol. 212, No. 2 159


T cells includes a recent study of mammalian PLAs from hep- Isolation and generation of human APCs. PBMCs were incubated
atitis virus that activate NKT cells (Fox et al., 2009; Zeissig with GM-CSF (300 U/ml) for 30 min. After removing the nonadherent
cells, medium with GM-CSF (300 U/ml) and IL-4 was added (200 U/ml)
et al., 2012) and the role of thymic PLA in controlling NKT
for 34 d. Adherent mDCs were collected by addition of PBS/0.5 mM of
cell selection (Paduraru et al., 2013). In these studies of blood- EDTA for 20 min, and then irradiated by -irradiation (5,000 rad). High
derived T cells, CD1a autoreactivity predominates, but in CD1a expression was verified by flow cytometry. We generated LC-like
the gastrointestinal tract CD1d is highly expressed, and NKT cells in vitro using two published protocols. Starting with CD14+ monocytes
cells home to the intestine and liver. Thus, both studies em- from human buffy coats from the NHS Blood Bank ( John Radcliffe Hospi-
phasize a role of phospholipases, but the differing effects of tal, Oxford, UK), CD14+ human cells were isolated using MACS cell sepa-
ration (Miltenyi Biotec) according to the suppliers instructions. In brief,
CD1a and CD1d isoforms may relate to the organ-specific
such in vitro LC-like cells were prepared as previously described (Caux et al.,
patterns of expression and T cell response. Free fatty acids 1997; Geissmann et al., 1998) from CD14+ cells, which were cultured in
were identified as natural skin antigens for CD1a earlier this 6-well plates in complete medium in the presence of IL-4 (250 ng/ml;
year (de Jong et al., 2014). Collectively these studies provide PeproTech), GM-CSF (100 ng/ml; PeproTech), and TGF-1 (10 ng/ml;
multiple lines of evidence that PLAs control various aspects PeproTech). At days 2 and 4, cultures were replated in the presence of
of T cell response in different organs. The clinical study of the aforementioned cytokines to generate cells which were 1015% CD1a+
CD207+. The in vitro LC-like cells were incubated with 10 g/ml anti
bee and wasp sting responses represents a model system to HLA-ABC and antiHLA-DR blocking antibodies (W6/32 and L243,
study the basic role of PLAs because it is characterized by respectively) for one hour before co-culture with T cells, to minimize HLA-
single, discreet exposures and there exist practical means to restricted responses. Alternatively, samples of human cord blood were ob-
mimic a stinging exposure and study the response in vivo and tained from CFAR Virology Core Laboratory at UCLA (Los Angeles, CA).
ex vivo. Having established a role for CD1a and PLA in the CD34+ human progenitor cells were isolated using MACS cell separation
normal response to venom, future studies will likely examine (Miltenyi Biotec) according to the suppliers instructions. LCs were then
prepared as previously described (Hunger et al., 2004) but with some modi-
their roles in hypersensitivity and immunotherapeutic sys- fications. Specifically, cultures of CD34+ cells were established in the pres-
tems and take advantage of information about lysophospho- ence of stem cell factor (25 ng/ml; R&D Systems), GM-CSF (200 U/ml;
lipids and free fatty acids as natural pathways that could be Sanofi US Services, Inc.), and TNF (2.5 ng/ml; Thermo Fisher Scientific).
considered for diagnosis through skin testing and antigen- At day 5, cultures were replated in the presence of GM-CSF (200 U/ml;
based immunotherapy. Sanofi US Services, Inc.) and TGF-1 (1 ng/ml; R&D Systems) to in-
crease CD1a expression. LC were harvested at day 12. The LC quality of
the cells was checked by flow cytometry, and found to consist of 3040%
CD1a+CD207+ cells.
MATERIALS AND METHODS
Isolation of human PBMC, lines, and clones. PBMCs were isolated
from healthy adult donors with a previous history of bee or wasp sting under Isolation of CD1a+ cells from skin. After removing subcutaneous fat,
local ethics approval (NRES Committee South Central, Oxford C, 09/ skin sections were cut into 5-mm-wide pieces and cultured in 2 mg/ml
H0606/71). T cells were purified from ficollized PBMCs using CD3 MACs dispase solution at 4C overnight. The epidermis was then separated from
beads (Miltenyi Biotec) and cultured for 1014 d with pulsed irradiated K562- the dermis and cultured in complete media for 5 d. Migratory adherent cells
CD1a cells in RPMI supplemented with 100 IU/ml penicillin, 100 g/ml were harvested and enriched by density gradient centrifugation, and CD1a+
streptomycin, 2 mM l-glutamine, and 5% human serum (Sigma-Aldrich), cells were isolated using MACs cell separation (Miltenyi Biotec). The CD1a+
nonessential amino acids, Hepes, sodium pyruvate, and 2-mercaptoethanol cells were incubated with 10 g/ml antiHLA-ABC and antiHLA-DR
(Life Technologies) in the presence of 1 nM IL-2 (PeproTech; 5,000,000 blocking antibodies (W6/32 and L243, respectively) for 1 h before co-culture
T cells and 200,000 K562-CD1a per well of 24-well plate). CD1a reactivity was with T cells, to minimize HLA-restricted responses.
then assessed by IFN- ELISPOT (Mabtech). ELISpot plates (Millipore)
were coated with antiIFN- antibody overnight (Mabtech). K562 cells CD1-plate assays. Biotinylated CD1a, CD1b, CD1c, and CD1d proteins
were pulsed with 1 g/ml wasp/bee venom (Sigma-Aldrich) or 100 ng/ml (National Institutes of Health Tetramer Core Facility, Emory University,
bee venom PLA2 (Sigma-Aldrich) overnight, and were then washed and re- Atlanta, GA) and anti-CD11a antibodies were added on streptavidin-coated
suspended in R5* (RPMI supplemented with 2 mM l-glutamine, 100 IU/ml plates (Thermo Fisher Scientific) at 10 g/ml and 2.5 g/ml, respectively, for
penicillin and 100 g/ml streptomycin plus 5% human serum). The plates 24 h at room temperature as detailed previously (de Jong et al., 2014). Wasp
were washed six times with RPMI and blocked for 1 h with RPMI supple- and bee venoms, their lipidic extracts, dideoxymycobactin (DDM), arachi-
mented with 2 mM l-glutamine, 100 IU/ml penicillin and 100 g/ml strep- donic acid, phosphatidylcholine 18:1, phosphatic acid 16:0, lysophosphatidyl-
tomycin plus 10% human serum (R10*). 20,00050,000 T cells were added choline 18:1, lysophosphatidic acid 16:0 or fatty acids 18:1, and 16:0 (Avanti)
per well to which 10,00025,000 K562 or primary cells were added. Wells were loaded at 10 g/ml in PBS for 2448 h at room temperature. BC2 cells
were set up in duplicate or triplicate. 10 ng/ml phorbol myristate acetate and and CD8-2 cells were then added to the well at 100,000 cells/well.
500 ng/ml Ionomycin was included as a positive control, and T cells alone
in the absence of K562 was included as a negative control. After overnight Cellular assays. 150,000 BC2 cells were co-cultured with 20,000 K562-
incubation at 37C and 5% CO2, plates were washed 6 times in PBS-Tween CD1 cells for 24 h with wasp and bee venoms, chloroform and methanol
0.05% and incubated with 1 g/ml of biotin-linked antiIFN- monoclonal extracts, chloroform and methanol precipitates, PLA2 (Sigma-Aldrich), mas
antibody (Mabtech) for 2 h. After washing 6 times in PBS-Tween 0.05%, toparan (Sigma-Aldrich) or arachidonic acid (Sigma-Aldrich). In some experi-
the plates were incubated for an additional 1 h with streptavidin-alkaline ments 10 g/ml anti-CD1a blocking antibody (OKT-6), or 10 g/ml IgG1
phosphatase (Mabtech). Spots were visualized using an alkaline phosphatase isotype control (P3), or 100 M manoalide, an inhibitor of PLA2 that acts
conjugate substrate kit (Bio-Rad Laboratories) and enumerated using an au- by covalently modifying lysine residues (Enzo Life Sciences), were added to
tomated ELISpot reader (Autimmun Diagnostika GmbH). The T cell lines, K562 or primary cells before addition of T cells.
BC2 and CD8-2, as well as K562-CD1 cells have been described previously
(Partners Health Care Institutional Review Board; Rosat et al., 1999; de Jong Venom extraction. 120 g of lyophilized wasp (Vespula species; ALK) and
et al., 2010, 2014). bee venom were resuspended in a solution of (1:1) methanol/chloroform.

160 Bee venom generates CD1a ligands | Bourgeois et al.


Ar ticle

After 40 min of rotation and a centrifugation at 300 g for 5 min, supernatant CD1-restricted T cell recognition of lipids from pollens. J. Exp. Med.
was collected. Lipid enriched-supernatant and protein-enriched precipitates 202:295308. http://dx.doi.org/10.1084/jem.20050773
were collected and resuspended in 1.2-ml of sterile PBS and normalized to Argiolas, A., and J.J. Pisano. 1983. Facilitation of phospholipase A2 activity
the input mass, or when possible, weighed to determine the exact mass. by mastoparans, a new class of mast cell degranulating peptides from
wasp venom. J. Biol. Chem. 258:1369713702.
Blister fluids. Healthy donors were injected with 10 g of wasp venom or Aslam, A., B. Kessler, M. Batycka, C.A. OCallaghan, S.A. Misbah, D.A.
saline intraepidermally, a dose which is at the low end of venom normally ad- Warrell, and G. Ogg. 2006. Defining the T cell antigen proteome of
ministered in a sting (ALK). Suction was applied to the skin at 200 mmHg for wasp venom. Clin. Exp. Allergy. 36:12741280. http://dx.doi.org/10
1 h, which induced a split between the epidermis and dermis (Salimi et al., .1111/j.1365-2222.2006.02569.x
Aslam, A., H. Chan, D.A. Warrell, S. Misbah, and G.S. Ogg. 2010. Track
2013). The blister fluids were collected by aspiration at 24 h and immediately
ing antigen-specific T-cells during clinical tolerance induction in hu-
stored at 20C. Lipids were extracted with chloroform, methanol, and
mans. PLoS ONE. 5:e11028. http://dx.doi.org/10.1371/journal.pone
water using the Bligh-Dyer method (Bligh and Dyer, 1959). .0011028
Bil, B.M., and F. Bonifazi. 2008. Epidemiology of insect-venom anaphylaxis.
HPLC-(electrospray ionization)-quadrupole time of flight mass Curr. Opin. Allergy Clin. Immunol. 8:330337. http://dx.doi.org/10.1097/
spectrometry. The analyses were performed using an Agilent Technologies ACI.0b013e32830638c5
6520 ESI-QTof coupled with HPLC (Agilent Technologies 1200). Lipids Bligh, E.G., and W.J. Dyer. 1959. A rapid method of total lipid extraction and
were separated using a 3 m 150 mm 2 mm diol column (Varian) as pre- purification. Can. J. Biochem. Physiol. 37:911917. http://dx.doi.org/
viously described (Layre et al., 2011). Spectra were collected in positive mode 10.1139/o59-099
from 100 to 3,000 m/z at 1 spectrum/s. Internal calibrations were performed Borg, N.A., K.S. Wun, L. Kjer-Nielsen, M.C. Wilce, D.G. Pellicci, R.
using calibrants at 121.050873 m/z and 922.009798 m/z that were coinfused Koh, G.S. Besra, M. Bharadwaj, D.I. Godfrey, J. McCluskey, and J.
into the spray chamber and used to monitor the stability of ion signal. Data Rossjohn. 2007. CD1d-lipid-antigen recognition by the semi-invariant
were analyzed using MassHunter Workstation Qualitative Analysis Software NKT T-cell receptor. Nature. 448:4449. http://dx.doi.org/10.1038/
(Agilent Technologies). nature05907
Cabrera, M.P., D.S. Alvares, N.B. Leite, B.M. de Souza, M.S. Palma, K.A.
ELISA. Culture supernatants were harvested and tested for IFN- produc- Riske, and J.R. Neto. 2011. New insight into the mechanism of action
tion by ELISA using the clone 2G1 (Thermo Fisher Scientific) for coating at of wasp mastoparan peptides: lytic activity and clustering observed with
2 g/ml and the biotin-labeled clone B133.5 (Thermo Fisher Scientific) at giant vesicles. Langmuir. 27:1080510813. http://dx.doi.org/10.1021/
la202608r
0.1 g/ml. Streptavidin-HRP (BD) was incubated for 30 min and the sub-
Calabi, F., and C. Milstein. 1986. A novel family of human major histocom-
strate was revealed by 2,2-Azinobis [3-ethylbenzothiazoline-6-sulfonic acid]-
patibility complex-related genes not mapping to chromosome 6. Nature.
diammonium salt (ABTS).
323:540543. http://dx.doi.org/10.1038/323540a0
Caux, C., C. Massacrier, B. Vanbervliet, B. Dubois, I. Durand, M. Cella,
PLA2 biochemical activity experiments. PLA2 activity in wasp and bee A. Lanzavecchia, and J. Banchereau. 1997. CD34+ hematopoietic pro-
venom were detected using site-specific substrate kit (Cayman Chemicals), genitors from human cord blood differentiate along two independent
according to manufacturers instructions. In a flat-bottom 96-well plate, 50 ng dendritic cell pathways in response to granulocyte-macrophage colony-
wasp or bee venom or 10 ng PLA2 (Sigma-Aldrich), plus 5 l assay buffer, stimulating factor plus tumor necrosis factor alpha: II. Functional analy-
were incubated for 1 h at RT with a substrate solution. Substrates used have sis. Blood. 90:14581470.
a thiol group attached at the sn-2 position of the phospholipid, and included Colonna, M. 2010. Skin function for human CD1a-reactive T cells. Nat.
arachidonyl thio-PC, heptanoyl thio-PC, diheptanoyl thio-PC, and palmi- Immunol. 11:10791080. http://dx.doi.org/10.1038/ni1210-1079
toyl thio-PC. In the presence of PLA2, cleavage of the substrate at the sn-2 Cortellini, G., M. Severino, E. Francescato, S. Turillazzi, I. Spadolini, A.
position resulted in release of the thiol group. After 1 h incubation, 10 l Rogkakou, and G. Passalacqua. 2012. Evaluation and validation of a
DNTB/EGTA was added, which reacts with free thiol groups producing a bee venom sting challenge performed by a micro-syringe. Ann. Allergy
colored precipitate, which was measured at 415 nm wavelength (iMark Asthma Immunol. 109:438441.
Microplate Reader; Bio-Rad Laboratories). Costa, H., and M.S. Palma. 2000. Agelotoxin: a phospholipase A(2) from
the venom of the neotropical social wasp cassununga (Agelaia pallipes
Statistics. Cohort of healthy donors investigated for CD1a-restricted wasp pallipes) (Hymenoptera-Vespidae). Toxicon. 38:13671379. http://dx.doi
.org/10.1016/S0041-0101(99)00199-3
venom, bee venom, and PLA2-specific responses were analyzed using one-
de Jong, A., V. Pea-Cruz, T.Y. Cheng, R.A. Clark, I. Van Rhijn, and D.B.
tailed Wilcoxon matched-pairs signed rank test. All other polyclonal T cells
Moody. 2010. CD1a-autoreactive T cells are a normal component of the
responses were analyzed using one-tailed Mann-Whitney tests.
human T cell repertoire. Nat. Immunol. 11:11021109. http://dx.doi
.org/10.1038/ni.1956
We thank John Altman and the NIH tetramer facility for providing recombinant de Jong, A., T.Y. Cheng, S. Huang, S. Gras, R.W. Birkinshaw, A.G. Kasmar,
CD1 proteins. I. Van Rhijn, V. Pea-Cruz, D.T. Ruan, J.D. Altman, et al. 2014.
This work was funded by the MRC and NIHR Biomedical Research Centre, the CD1a-autoreactive T cells recognize natural skin oils that function as
National Institutes of Health (NIH; NIAMS R01 048632), and the Burroughs headless antigens. Nat. Immunol. 15:177185. http://dx.doi.org/10.1038/
Wellcome Foundation Program in Translational Medicine. GO also acknowledges the ni.2790
support of the National Institute for Health Research Clinical Research Network. de Lalla, C., M. Lepore, F.M. Piccolo, A. Rinaldi, A. Scelfo, C. Garavaglia,
The work is supported by Cancer Research UK (Programme Grant C399/A2291 L. Mori, G. De Libero, P. Dellabona, and G. Casorati. 2011. High-
to V. Cerundolo). frequency and adaptive-like dynamics of human CD1 self-reactive
The authors have no conflicting financial interests. T cells. Eur. J. Immunol. 41:602610. http://dx.doi.org/10.1002/eji
.201041211
Submitted: 7 August 2014 de Oliveira, M.R., and M.S. Palma. 1998. Polybitoxins: a group of phos-
Accepted: 11 December 2014 pholipases A2 from the venom of the neotropical social wasp paulistinha
(Polybia paulista). Toxicon. 36:189199. http://dx.doi.org/10.1016/S0041-
0101(97)00053-6
REFERENCES dos Santos, L.D., A.R. da Silva Menegasso, J.R. dos Santos Pinto, K.S.
Agea, E., A. Russano, O. Bistoni, R. Mannucci, I. Nicoletti, L. Corazzi, Santos, F.M. Castro, J.E. Kalil, and M.S. Palma. 2011. Proteomic char-
A.D. Postle, G. De Libero, S.A. Porcelli, and F. Spinozzi. 2005. Human acterization of the multiple forms of the PLAs from the venom of the

JEM Vol. 212, No. 2 161


social wasp Polybia paulista. Proteomics. 11:14031412. http://dx.doi.org/ Kronenberg, M., and W.L. Havran. 2014. Immunology: oiling the wheels of
10.1002/pmic.201000414 autoimmunity. Nature. 506:4243. http://dx.doi.org/10.1038/506042a
Dougan, S.K., A. Kaser, and R.S. Blumberg. 2007. CD1 expression on Kronenberg, M., and Y. Kinjo. 2005. Infection, autoimmunity, and gly-
antigen-presenting cells. Curr. Top. Microbiol. Immunol. 314:113141. colipids: T cells detect microbes through self-recognition. Immunity.
Egner, W., C. Ward, D.L. Brown, and P.W. Ewan. 1998. The frequency 22:657659. http://dx.doi.org/10.1016/j.immuni.2005.06.001
and clinical significance of specific IgE to both wasp (Vespula) and honey- Lalvani, A., and M. Pareek. 2010. A 100 year update on diagnosis of tuberculo-
bee (Apis) venoms in the same patient. Clin. Exp. Allergy. 28:2634. sis infection. Br. Med. Bull. 93:6984. http://dx.doi.org/10.1093/bmb/
http://dx.doi.org/10.1046/j.1365-2222.1998.00176.x ldp039
Felio, K., H. Nguyen, C.C. Dascher, H.J. Choi, S. Li, M.I. Zimmer, A. Layre, E., L. Sweet, S. Hong, C.A. Madigan, D. Desjardins, D.C. Young,
Colmone, D.B. Moody, M.B. Brenner, and C.R. Wang. 2009. CD1- T.Y. Cheng, J.W. Annand, K. Kim, I.C. Shamputa, et al. 2011. A com-
restricted adaptive immune responses to Mycobacteria in human group parative lipidomics platform for chemotaxonomic analysis of Mycobacte
1 CD1 transgenic mice. J. Exp. Med. 206:24972509. http://dx.doi rium tuberculosis. Chem. Biol. 18:15371549. http://dx.doi.org/10.1016/
.org/10.1084/jem.20090898 j.chembiol.2011.10.013
Fox, L.M., D.G. Cox, J.L. Lockridge, X. Wang, X. Chen, L. Scharf, D.L. Manolova, V., Y. Hirabayashi, L. Mori, and G. De Libero. 2003. CD1a
Trott, R.M. Ndonye, N. Veerapen, G.S. Besra, et al. 2009. Recognition and CD1b surface expression is independent from de novo synthesized
of lyso-phospholipids by human natural killer T lymphocytes. PLoS glycosphingolipids. Eur. J. Immunol. 33:2937. http://dx.doi.org/10
Biol. 7:e1000228. http://dx.doi.org/10.1371/journal.pbio.1000228 .1002/immu.200390004
Gadola, S.D., N.R. Zaccai, K. Harlos, D. Shepherd, J.C. Castro-Palomino, McMichael, A.J., J.R. Pilch, G. Galfr, D.Y. Mason, J.W. Fabre, and C.
G. Ritter, R.R. Schmidt, E.Y. Jones, and V. Cerundolo. 2002. Structure Milstein. 1979. A human thymocyte antigen defined by a hybrid my-
of human CD1b with bound ligands at 2.3 A, a maze for alkyl chains. eloma monoclonal antibody. Eur. J. Immunol. 9:205210. http://dx.doi
Nat. Immunol. 3:721726. .org/10.1002/eji.1830090307
Geissmann, F., C. Prost, J.P. Monnet, M. Dy, N. Brousse, and O. Hermine. Moody, D.B., T. Ulrichs, W. Mhlecker, D.C. Young, S.S. Gurcha, E.
1998. Transforming growth factor beta1, in the presence of granulo- Grant, J.P. Rosat, M.B. Brenner, C.E. Costello, G.S. Besra, and S.A.
cyte/macrophage colony-stimulating factor and interleukin 4, induces Porcelli. 2000. CD1c-mediated T-cell recognition of isoprenoid glycolip-
differentiation of human peripheral blood monocytes into dendritic ids in Mycobacterium tuberculosis infection. Nature. 404:884888. http://dx
Langerhans cells. J. Exp. Med. 187:961966. .doi.org/10.1038/35009119
Godfrey, D.I., and J. Rossjohn. 2011. New ways to turn on NKT cells. Moody, D.B., D.C. Young, T.Y. Cheng, J.P. Rosat, C. Roura-Mir, P.B.
J. Exp. Med. 208:11211125. http://dx.doi.org/10.1084/jem.20110983 OConnor, D.M. Zajonc, A. Walz, M.J. Miller, S.B. Levery, et al. 2004.
Gumperz, J.E., C. Roy, A. Makowska, D. Lum, M. Sugita, T. Podrebarac, Y. T cell activation by lipopeptide antigens. Science. 303:527531. http://dx
Koezuka, S.A. Porcelli, S. Cardell, M.B. Brenner, and S.M. Behar. 2000. .doi.org/10.1126/science.1089353
Murine CD1d-restricted T cell recognition of cellular lipids. Immunity. Mller, U.R., N. Johansen, A.B. Petersen, J. Fromberg-Nielsen, and G.
12:211221. http://dx.doi.org/10.1016/S1074-7613(00)80174-0 Haeberli. 2009. Hymenoptera venom allergy: analysis of double positiv-
Gutierrez, D.A., and H.R. Rodewald. 2013. A sting in the tale of TH2 ity to honey bee and Vespula venom by estimation of IgE antibodies to
immunity. Immunity. 39:803805. http://dx.doi.org/10.1016/j.immuni species-specific major allergens Api m1 and Ves v5. Allergy. 64:543548.
.2013.10.015 http://dx.doi.org/10.1111/j.1398-9995.2008.01794.x
Hiromatsu, K., C.C. Dascher, K.P. LeClair, M. Sugita, S.T. Furlong, M.B. Murakami, M., and G. Lambeau. 2013. Emerging roles of secreted phos-
Brenner, and S.A. Porcelli. 2002a. Induction of CD1-restricted im- pholipase A(2) enzymes: an update. Biochimie. 95:4350. http://dx.doi
mune responses in guinea pigs by immunization with mycobacterial .org/10.1016/j.biochi.2012.09.007
lipid antigens. J. Immunol. 169:330339. http://dx.doi.org/10.4049/ Paduraru, C., J.S. Bezbradica, A. Kunte, R. Kelly, J.A. Shayman, N.
jimmunol.169.1.330 Veerapen, L.R. Cox, G.S. Besra, and P. Cresswell. 2013. Role for ly-
Hiromatsu, K., C.C. Dascher, M. Sugita, C. Gingrich-Baker, S.M. Behar, sosomal phospholipase A2 in iNKT cell-mediated CD1d recognition.
K.P. LeClair, M.B. Brenner, and S.A. Porcelli. 2002b. Characterization Proc. Natl. Acad. Sci. USA. 110:50975102. http://dx.doi.org/10.1073/
of guinea-pig group 1 CD1 proteins. Immunology. 106:159172. http:// pnas.1302923110
dx.doi.org/10.1046/j.1365-2567.2002.01422.x Porcelli, S., M.B. Brenner, J.L. Greenstein, S.P. Balk, C. Terhorst, and
Huang, S., T.Y. Cheng, D.C. Young, E. Layre, C.A. Madigan, J. Shires, P.A. Bleicher. 1989. Recognition of cluster of differentiation 1 antigens
V. Cerundolo, J.D. Altman, and D.B. Moody. 2011. Discovery of de- by human CD4-CD8-cytolytic T lymphocytes. Nature. 341:447450.
oxyceramides and diacylglycerols as CD1b scaffold lipids among diverse http://dx.doi.org/10.1038/341447a0
groove-blocking lipids of the human CD1 system. Proc. Natl. Acad. Sci. Porcelli, S., C.T. Morita, and M.B. Brenner. 1992. CD1b restricts the re-
USA. 108:1933519340. http://dx.doi.org/10.1073/pnas.1112969108 sponse of human CD4-8- T lymphocytes to a microbial antigen. Nature.
Hunger, R.E., P.A. Sieling, M.T. Ochoa, M. Sugaya, A.E. Burdick, T.H. 360:593597. http://dx.doi.org/10.1038/360593a0
Rea, P.J. Brennan, J.T. Belisle, A. Blauvelt, S.A. Porcelli, and R.L. Rosat, J.P., E.P. Grant, E.M. Beckman, C.C. Dascher, P.A. Sieling, D.
Modlin. 2004. Langerhans cells utilize CD1a and langerin to efficiently Frederique, R.L. Modlin, S.A. Porcelli, S.T. Furlong, and M.B. Brenner.
present nonpeptide antigens to T cells. J. Clin. Invest. 113:701708. 1999. CD1-restricted microbial lipid antigen-specific recognition found
http://dx.doi.org/10.1172/JCI200419655 in the CD8+ alpha beta T cell pool. J. Immunol. 162:366371.
Kasmar, A., I. Van Rhijn, and D.B. Moody. 2009. The evolved functions of Salimi, M., J.L. Barlow, S.P. Saunders, L. Xue, D. Gutowska-Owsiak, X.
CD1 during infection. Curr. Opin. Immunol. 21:397403. http://dx.doi Wang, L.C. Huang, D. Johnson, S.T. Scanlon, A.N. McKenzie, et al.
.org/10.1016/j.coi.2009.05.022 2013. A role for IL-25 and IL-33driven type-2 innate lymphoid cells
Kawano, T., J. Cui, Y. Koezuka, I. Toura, Y. Kaneko, K. Motoki, H. Ueno, in atopic dermatitis. J. Exp. Med. 210:29392950. http://dx.doi.org/
R. Nakagawa, H. Sato, E. Kondo, et al. 1997. CD1d-restricted and TCR- 10.1084/jem.20130351
mediated activation of valpha14 NKT cells by glycosylceramides. Science. Sallusto, F., and A. Lanzavecchia. 1994. Efficient presentation of soluble
278:16261629. http://dx.doi.org/10.1126/science.278.5343.1626 antigen by cultured human dendritic cells is maintained by granulocyte/
King, T.P., S.Y. Jim, and K.M. Wittkowski. 2003. Inflammatory role of two macrophage colony-stimulating factor plus interleukin 4 and down-
venom components of yellow jackets (Vespula vulgaris): a mast cell de- regulated by tumor necrosis factor alpha. J. Exp. Med. 179:11091118.
granulating peptide mastoparan and phospholipase A1. Int. Arch. Allergy http://dx.doi.org/10.1084/jem.179.4.1109
Immunol. 131:2532. http://dx.doi.org/10.1159/000070431 Scott, D.L., S.P. White, Z. Otwinowski, W. Yuan, M.H. Gelb, and P.B.
Klein, L., B. Kyewski, P.M. Allen, and K.A. Hogquist. 2014. Positive and Sigler. 1990. Interfacial catalysis: the mechanism of phospholipase A2.
negative selection of the T cell repertoire: what thymocytes see (and Science. 250:15411546. http://dx.doi.org/10.1126/science.2274785
dont see). Nat. Rev. Immunol. 14:377391. http://dx.doi.org/10.1038/ Sin, B.A., M. Akdis, J. Zumkehr, S. Bezzine, C. Bekpen, G. Lambeau,
nri3667 and C.A. Akdis. 2011. T-cell and antibody responses to phospholipase

162 Bee venom generates CD1a ligands | Bourgeois et al.


Ar ticle

A2 from different species show distinct cross-reactivity patterns. Allergy. cells and tissues via IL1-. Eur. J. Immunol. 41:694705. http://dx.doi
66:15131521. http://dx.doi.org/10.1111/j.1398-9995.2011.02689.x .org/10.1002/eji.201040808
Starkl, M., N. Brunner, and T.A. Stenstrm. 2013. Why do water and san- Young, D.C., and D.B. Moody. 2006. T-cell recognition of glycolipids pre-
itation systems for the poor still fail? Policy analysis in economically sented by CD1 proteins. Glycobiology. 16:103R112R. http://dx.doi
advanced developing countries. Environ. Sci. Technol. 47:61026110. .org/10.1093/glycob/cwj111
http://dx.doi.org/10.1021/es3048416 Zeissig, S., S.K. Dougan, D.C. Barral, Y. Junker, Z. Chen, A. Kaser, M.
Tameris, M.D., M. Hatherill, B.S. Landry, T.J. Scriba, M.A. Snowden, Ho, H. Mandel, A. McIntyre, S.M. Kennedy, et al. 2010. Primary de-
S. Lockhart, J.E. Shea, J.B. McClain, G.D. Hussey, W.A. Hanekom, ficiency of microsomal triglyceride transfer protein in human abetali-
et al. MVA85A 020 Trial Study Team. 2013. Safety and efficacy of poproteinemia is associated with loss of CD1 function. J. Clin. Invest.
MVA85A, a new tuberculosis vaccine, in infants previously vaccinated 120:28892899. http://dx.doi.org/10.1172/JCI42703
with BCG: a randomised, placebo-controlled phase 2b trial. Lancet. Zeissig, S., K. Murata, L. Sweet, J. Publicover, Z. Hu, A. Kaser, E. Bosse, J.
381:10211028. http://dx.doi.org/10.1016/S0140-6736(13)60177-4 Iqbal, M.M. Hussain, K. Balschun, et al. 2012. Hepatitis B virus-induced
Valentin, E., F. Ghomashchi, M.H. Gelb, M. Lazdunski, and G. Lambeau. lipid alterations contribute to natural killer T cell-dependent protec-
2000. Novel human secreted phospholipase A(2) with homology to the tive immunity. Nat. Med. 18:10601068. http://dx.doi.org/10.1038/
group III bee venom enzyme. J. Biol. Chem. 275:74927496. http://dx nm.2811
.doi.org/10.1074/jbc.275.11.7492 Zeng, Z., A.R. Castao, B.W. Segelke, E.A. Stura, P.A. Peterson, and I.A.
Yakimchuk, K., C. Roura-Mir, K.G. Magalhaes, A. de Jong, A.G. Kasmar, Wilson. 1997. Crystal structure of mouse CD1: An MHC-like fold with
S.R. Granter, R. Budd, A. Steere, V. Pena-Cruz, C. Kirschning, et al. a large hydrophobic binding groove. Science. 277:339345. http://dx
2011. Borrelia burgdorferi infection regulates CD1 expression in human .doi.org/10.1126/science.277.5324.339

JEM Vol. 212, No. 2 163


INSIGHTS

T cells require DOCK8 for flexibility and function


In this issue, Zhang et al. report a novel function for DOCK8 (dedicator of cytokinesis 8) in controlling the
structural integrity of lymphocytes. DOCK8 deficiency severely compromised survival of T cells during
trafficking through dense tissue networks, impairing skin-specific protective antiviral immune responses.
Identifying the genetic lesions underlying primary immunodeficiencies has an immediate impact on
disease diagnosis, therapy, and patient management. It can also provide mechanistic explanations for disease
pathogenesis, such as the absence of T and NK cells in X-SCID due to the requirement for gc in IL-7R and
IL-15R signaling, and the lack of switched Ig isotypes in hyperimmunoglobulin M syndrome due to the
Insight from requirement for CD40/CD40L interactions in this process. However, there are other examples where iden-
Stuart Tangye tifying the mutant gene in a particular disease opens up a new field of study, for the simple reasons that little
was known about the function of this gene in immune cells. The discovery that biallelic loss-of-function
mutations in DOCK8 causes autosomal recessive hyper IgE syndrome falls into this second category. DOCK8 is a guanine
nucleotide exchange factor that activates small GTPases such as Cdc42. Previous studies revealed that cytoskeletal defects in
DOCK8-deficient B cells, T cells, and NK cells impairs their effector function. However, it is not clear why individuals
with DOCK8 mutations specifically develop severe viral infections of the skin. Zhang et al. theorized that since DOCK8-
deficient lymphocytes exhibited normal chemotaxis and initial migration into human tissue, these cells may be impaired
in their ability to traffic through the dense tissue networks of the
skin. By analyzing humans, mice, and cell lines, DOCK8-
deficient T and NK cells were found to have aberrant mor-
phologies under conditions that replicated skin infiltration
and penetration, and this ultimately contributed to an unusual
form of cell death termed cytothripsis (cell shattering). This
morphological effect was replicated by abolishing expression
of Cdc42 or p21-activated kinase (PAK), but not RAC1/2 or
the WAS protein, thereby establishing that DOCK8 operates
in this setting by activating these regulators of actin polymer-
ization. Overall, the stress experienced by DOCK8-deficient In adoptive transfer experiments, Dock8-deficient T cells were
cells moving through dense tissue networks abrogated the gen- undetectable in skin one month after HSV infection compared with
eration and maintenance of tissue-resident memory CD8+ normal numbers of control T cells. Left panel: two photon imaging
T cells that are important for protective immunity at such sites. of the skin 17 days after infection with HSV; right panel: numbers of
This selective inability of effector T cells lacking HSV-specific T cells in skin at various times after infection with HSV.
DOCK8 to efficiently localize to tissues high in collagen
content and provide potent antiviral immunity at these sites may explain why DOCK8-deficient patients have heightened
susceptibility to skin-trophic viral infections, yet systemic viral infections are more effectively controlled. These findings reveal the
morphological flexibility of immune cells that is required for them to execute effector function in nonlymphoid tissues and the
critical function of DOCK8/Cdc42/PAK in this process. It will be important to elucidate the mechanism by which DOCK8
integrates into TCR signaling, as well as to assess virus-specific skin-resident memory cells in DOCK8-deficient humans to
establish a paucity of these cells at these sites. Identification of the components that regulate lymphocyte integrity, motility,
and survival under conditions of migratory stress may provide an opportunity to enhance tissue-specific immunity and memory
in patients with germline mutations in this pathway. Eventually, shedding light on the pathology arising in the skin may result
in improved outcomes for patients with this often-fatal immunodeficiency.
Zhang, Q., et al. 2014. J. Exp. Med. http://dx.doi.org/10.1084/jem.20141307.

Stuart G. Tangye, Garvan Institute of Medical Research: s.tangye@garvan.org.au


Article

DOCK8 regulates lymphocyte shape


integrity for skin antiviral immunity
Qian Zhang,1 Christopher G. Dove,1* Jyh Liang Hor,5,6*
Heardley M. Murdock,1 Dara M. Strauss-Albee,1 Jordan A. Garcia,1
Judith N. Mandl,2 Rachael A. Grodick,1 Huie Jing,1
Devon B. Chandler-Brown,1 Timothy E. Lenardo,1 Greg Crawford,7
Helen F. Matthews,3 Alexandra F. Freeman,4 Richard J. Cornall,7
Ronald N. Germain,2 Scott N. Mueller,5,6 and Helen C. Su1
1Laboratory of Host Defenses, 2Laboratory of Systems Biology, 3Laboratory of Immunology, and 4Laboratory of Clinical
Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
5Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, and 6The ARC Centre

of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia
7MRC Human Immunology Unit, Nuffield Department of Medicine, Oxford University, Oxford OX3 7BN, England, UK

DOCK8 mutations result in an inherited combined immunodeficiency characterized by in-


creased susceptibility to skin and other infections. We show that when DOCK8-deficient
T and NK cells migrate through confined spaces, they develop cell shape and nuclear defor-
mation abnormalities that do not impair chemotaxis but contribute to a distinct form of
catastrophic cell death we term cytothripsis. Such defects arise during lymphocyte migration
in collagen-dense tissues when DOCK8, through CDC42 and p21-activated kinase (PAK), is
unavailable to coordinate cytoskeletal structures. Cytothripsis of DOCK8-deficient cells
prevents the generation of long-lived skin-resident memory CD8 T cells, which in turn impairs
control of herpesvirus skin infections. Our results establish that DOCK8-regulated shape
integrity of lymphocytes prevents cytothripsis and promotes antiviral immunity in the skin.

CORRESPONDENCE DOCK8, which is highly expressed only within cause disease in up to 30% of the population
Helen C. Su: the immune system, functions as an atypical gua- (Higgins et al., 1993; Kilkenny and Marks, 1996;
hsu@niaid.nih.gov
OR
nine nucleotide exchange factor (GEF) to acti- Harpaz et al., 2008). In contrast to normal individ
Scott N. Mueller: vate small Rho GTPases (Ct and Vuori, 2002; uals, DOCK8-deficient patients with autosomal-
smue@unimelb.edu.au Ruusala and Aspenstrm, 2004; Meller et al., recessive loss-of-function mutations in DOCK8
2005; Harada et al., 2012; Mou et al., 2012) and have impaired cellular and humoral immunity
Abbreviations used: DNFB,
2,4-dinitrofluorobenzene; GEF, its role as an adaptor in TLR9-MYD88 signal- (Engelhardt et al., 2009; Zhang et al., 2009; Su
guanine nucleotide exchange ing suggests additional functions beyond GEF et al., 2011; Jing et al., 2014) that manifests as ex-
factor; PAK, p21-activated activity ( Jabara et al., 2012). DOCK proteins and treme susceptibility to skin and other infections
kinase; PI, propidium iodide;
TRM, tissue-resident memory
their orthologs participate in diverse biological (Chu et al., 2012). Patients often suffer from
CD8 T cells; WASP, Wiskott- processes, including gonadal and epidermal cell disseminated and persistent viral skin infections
Aldrich syndrome protein. migration during embryonic development, tumor including those caused by HSV, varicella-zoster
cell invasion, and leukocyte chemotaxis and traf- virus, human papillomavirus, and molluscum
ficking through LNs (Kunisaki et al., 2006; Ct contagiosum. Their chronic viral infections may
and Vuori, 2007; Gotoh et al., 2008; Kikuchi reflect multiple defects that affect T cell activa-
et al., 2008; Nishikimi et al., 2009, 2013; Harada tion, proliferation, survival, and priming by den-
et al., 2012). dritic cells (Zhang et al., 2009; Lambe et al., 2011;
For most people without any obvious im- Randall et al., 2011; Harada et al., 2012; Crawford
mune deficiency, infections with HSV, varicella- et al., 2013), NK cell cytotoxicity (Ham et al.,
zoster virus, or human papillomavirus cause
self-limited cold sores, chickenpox, or warts. How
ever, these viruses can reemerge from latency to This article is distributed under the terms of an AttributionNoncommercialShare
AlikeNo Mirror Sites license for the first six months after the publication date
(see http://www.rupress.org/terms). After six months it is available under a Cre-
ative Commons License (AttributionNoncommercialShare Alike 3.0 Unported
*C.G. Dove and J.L. Hor contributed equally to this paper. license, as described at http://creativecommons.org/licenses/by-nc-sa/3.0/).

The Rockefeller University Press $30.00


J. Exp. Med. 2014 Vol. 211 No. 13 25492566 2549
www.jem.org/cgi/doi/10.1084/jem.20141307
Figure 1. Skin disease in DOCK8-defi-
cient patients. (A) Chronic ulcerating and
disseminated HSV infections of the skin in-
volving axilla (left), trunk (middle), and vulva
(right) of a DOCK8-deficient patient. 19 of
34 patients in the NIH cohort had similar re-
current or disseminated HSV skin infections.
(B) Skin histology in human DOCK8 deficiency.
Hematoxylin and eosin staining showing epi-
dermal and dermal layers of skin biopsies from
normal healthy control (left) and DOCK8-
deficient patient (right). Note highly packed,
multilayered epithelial cells in the epidermis
and many large pink collagen bundles in the
dermis. Bars, 1 mm. Skin biopsies were evaluated
from 17 DOCK8-deficient patients. (C) T cells
(green) from either a healthy control (left) or
DOCK8-deficient patient (right), shown mi-
grating in dermis (purple) of foreskin tissues
from healthy donors. Red arrows, elongated
cell processes. Bars, 10 m. Shown is a repre-
sentative of four patients and four healthy
controls tested in three experiments.

2013; Mizesko et al., 2013), and antiviral cytokine production normal skin structures (Fig. 1 B), likely reflecting the fact that
(Zhang et al., 2009). DOCK8 is not expressed by normal keratinocytes, fibroblasts,
T effector cells are a critical component of immunity to the and endothelial cells (Su et al., 2011). Dock8-deficient den-
types of viral skin infections characteristically seen in DOCK8 dritic cells migrate poorly into LNs (Harada et al., 2012). This
deficiency. These cells must scan for and target pathogens raised the possibility that impaired presentation of viral antigens
within the large volume of the skin, which is organized into by dendritic cells within draining LNs might lead to defective
two layers. The epidermis is composed of interlocking arrays T cell immunity to viruses that infect the skin. However, given
of keratinocytes that impede the passage of immune effector that DOCK8 is expressed in T cells as well as myeloid cells,
cells (Honda et al., 2014). In contrast, the dermis is composed it was also possible that a similar migration defect among skin-
of a dense network of packed collagen fibers, through which infiltrating cytotoxic lymphocytes might impair their effec-
immune cells must navigate (Wolf et al., 2009; Honda et al., tive ability to control viral replication locally. To investigate
2014). The collagen fibers make up as much as one third of the motility of DOCK8-deficient T cells, we examined fluor
the wet weight of skin, as compared with 10% of aorta or escently labeled T cells from patients that were allowed to
1% or less of other organs such as spleen and brain (Lowry migrate into the dermal layer of human foreskin biopsies. The
et al., 1941; Neuman and Logan, 1950).Thus, the extracellular mutant T cells moved through the extracellular matrix of the
environments of the epidermis and dermis are characterized skin but had abnormally elongated thin processes (Fig. 1 C).
by many highly confined spaces, which are likely to tax the A similar phenotype was observed when the skins dermal layer
structural integrity of cells navigating to their targets. Given was modeled in vitro by placing patient T cells into a three-
the presumptive role of DOCK8 in controlling cell cytoskel- dimensional (3D) collagen gel matrix (Fig. 2 A; and Video 1);
etal function and migration capacity, the fact that DOCK8- this differed from the rounded, nonmotile phenotype reported
deficient patientsin comparison with other combined for Dock8-deficient dendritic cells (Harada et al., 2012).
immunodeficiency patientsseem to suffer disproportion- The elongated T cells also had dramatically elongated nuclei
ately from a broad variety of skin infections, and the evidence (Fig. 2 A). Such elongation was not due to failed cytokinesis
for physical constraints on immune cell movement in skin, we despite a role of some DOCK proteins in promoting cell di-
investigated whether the skin viral susceptibility of these pa- vision (Kittler et al., 2007) because elongated cells only had
tients might relate to a defect in effector cell migration. Our one centrosome (Fig. 2 B), elongation persisted at high levels
studies revealed an unexpected, critical role for DOCK8 in when cell cycle progression was minimal after prolonged cul-
maintaining lymphocyte cellular integrity during migration ture (Fig. 2 C), and cell cycle arrest at G1/S did not prevent
in dense environments that limits host resistance. elongation (Fig. 2, D and E). Time-lapse video microscopy re-
vealed that T cells from patients (Fig. 2 F) spent an increased
RESULTS proportion of time elongated, as did normal T cells in which
DOCK8-deficient T cells and NK cells develop abnormally DOCK8 expression was silenced by transfection with siRNA
elongated shape and nuclear deformation (Fig. 2 G). Abnormal elongation also occurred in T cells and
Despite their susceptibility to skin infections including HSV NK cells from Dock8 mutant mice (Fig. 2, H and I; and Video 2).
(Fig. 1 A), DOCK8-deficient patients have histologically Like T cells, NK cells are present in the skin of healthy
2550 DOCK8 and cytothripsis in skin viral infections | Zhang et al.
Ar ticle

Figure 2. Abnormal morphology of


DOCK8-deficient lymphocytes when
migrating in 3D collagen gel matrices.
(A) Confocal imaging of T cells migrating in a
collagen gel matrix (3.6 mg/ml) with nuclear
staining. BF, brightfield. DAPI, blue. Represen-
tative of six experiments. (B) Confocal imaging
after in-gel immunofluorescence staining of
control and patient T cells for pericentrin (red,
indicated by arrows), -tubulin (green), and
nuclei (blue). Stained gels were physically
compressed before imaging. Left panels: mi-
grating cells, each showing one centrosome.
Right panels: dividing anaphase cells, each
showing two centrosomes. Representative of
three patients and three healthy controls from
three experiments. (C) Proportions of resting
T cells that were abnormally elongated. Less
than 2% of cells were in S/G2/M phases after
6 wk of culture. Three controls and three pa-
tients were tested from three experiments.
(D) Proportions of patient T cells that were
abnormally elongated after cell cycle blockade.
Similar analysis as for C, except that cells were
from three patients that were treated with
either aphidicolin (Aph) or vehicle (DMSO),
tested in three experiments. (E) Nuclear elon-
gation among abnormally elongated cells.
Nuclei in cells from D were stained with
Hoechst for analysis. (F) Percentage of time
cells spent elongated in the collagen matrices.
6 patients and 12 healthy controls were tested
from five experiments. (G) Similar analysis as
for F, except performed with T cells transfected
with DOCK8 or nonspecific (NS) siRNA, from
four experiments. Representative immunoblot
showing DOCK8 knockdown efficiency.
(H and I) Proportions elongated of T cells (H)
and NK cells (I) from 10 Dock8-deficient (KO)
or control (WT) mice from three experiments,
migrating in collagen gel matrices. Bars,
10 m. CI show means SD. Unpaired two-
tailed Students t test were performed. Statis-
tical significance indicated by *, P < 0.05;
****, P < 0.0001; ns, not significant.

persons and those with inflammatory skin conditions (Ebert occasionally remained in place while moving both ends in a
et al., 2006; Grgoire et al., 2007). These lymphocytes also help poorly coordinated manner, suggesting that immune surveil-
protect against HSV and other viral infections, especially at lance could potentially be compromised (Video 1). However,
early times after infection (Biron et al., 1999). Thus, the in- during skin infection, various chemokines are induced (Stock
ability to maintain shape integrity during migration in the 3D et al., 2014), which could contribute to directed T cell migra-
microenvironment was an intrinsic property of the lympho- tion toward viral pathogens.To test whether the abnormal mor-
cytes due to the lack of DOCK8 expression. phology of Dock8-deficient T cells was associated with impaired
chemotaxis, we tracked individual cells moving through col-
DOCK8-deficient cells exhibit normal chemotaxis lagen matrices toward a gradient of CXCL12 (SDF-1). For
but undergo fragmentation and cell death in 3D conditions each cell, directional velocity and track straightness toward the
The random migratory pattern of T cells within collagen gel chemokine, as well as speed along the path traveled and track
matrices mimics their migration pattern in skin tissues, facili- straightness from origin to destination, were analyzed over a
tating surveillance for infected cells. We saw that DOCK8- 30-min period (Fig. 3 E). The mean values for the population
deficient T cells migrating within 3D collagen gel matrices of cells from each DOCK8-deficient patient were similar to

JEM Vol. 211, No. 13 2551


Figure 3. Chemotaxis of DOCK8-deficient T cells
migrating toward CXCL12 in collagen gel matrices.
(AD) Velocity toward chemokine (A), chemokine direc-
tionality (B), track speed (C), and directionality (D). Each
symbol represents the means of these values for indi-
vidually tracked cells from each sample. The horizontal
line designates the average of these means. T cells from
four patients and eight controls were evaluated from
four experiments. In two separate experiments, normal
T cells were transfected with DOCK8 siRNA or nonspecific
(NS) siRNA. (E) Cartoon showing measurements of che-
motaxis, based upon the actual route that a cell took as
indicated by the gray dotted line. (F) Representative flow
cytometric measurement of CXCR4 (receptor for CXCL12)
expression on cells used in the chemotaxis assays. Un-
paired two-tailed Students t tests were performed. All
statistical comparisons were nonsignificant.

normal healthy control cells (Fig. 3, AD). Chemotaxis was also apoptosis such as loss of mitochondrial membrane potential
unaffected when tested using normal T cells in which DOCK8 or caspase activation (Fig. 5, BE). This cell death was not
expression was silenced after transfection with siRNA (Fig. 3, blocked by treatment with the pan-caspase inhibitor zVAD-fmk
AD, and F). Thus, DOCK8-deficient cells were capable of (Fig. 5 F). Together, these results establish that DOCK8-
normally sensing and migrating toward a chemokine source. deficient lymphocytes, when moving for prolonged periods in
In contrast, after hours of moving within the matrix, pa- a 3D environment, undergo a distinct form of cell death asso-
tient T cells often fragmented catastrophically as they moved ciated with abnormal cell shape and movement, which we
in place (Fig. 4 A and Video 3). The cell fragments contained term cytothripsis (cell shattering).
pieces of deformed nucleus, as shown by propidium iodide
(PI) staining. Flow cytometric quantification of dead and dying
cells revealed that silencing DOCK8 in otherwise healthy donor Abnormal elongation requires movement
human T cells caused them to die within the gels but not within through confined spaces but not adhesive forces
liquid medium (Fig. 4 B). T cells and NK cells, from patients We next investigated the spatial configuration of the micro-
or mice genetically deficient in DOCK8, showed a similar fate environment that elicited this abnormal phenotype. Lowering
(Fig. 4 C and not depicted). Cells that died had spent slightly the concentration of collagen within the gel matrices, which
greater proportion of time elongated, but the duration of increases the average pore size through which the cells mi-
elongation episodes immediately preceding fragmentation grate (Pedersen and Swartz, 2005), resulted in a correspond-
were longer, suggesting that abnormal morphology correlated ing decrease in the amount of cell death (Fig. 6 A). Abnormal
with cell death (Fig. 4, D and E). The dying cells recovered elongation with nuclear deformation also occurred when pa-
from collagen gels showed ultrastructural features reminiscent tient T cells migrated through the small pores of an uncoated
of apoptosis and necrosis with cell shrinkage, loss of microvilli polycarbonate transwell insert (Fig. 6 B) or within 3D matri-
but no membrane blebbing, and holes in the plasma mem- ces composed of agarose (Fig. 6 C). This phenotype was not
brane (Fig. 5 A) but lacked biochemical evidence of classical observed when cells moved on ICAM-coated (Fig. 6 D) or

2552 DOCK8 and cytothripsis in skin viral infections | Zhang et al.


Ar ticle

Figure 4. Elongation when migrating through confined spaces leads to cytothripsis. (A) Time-lapse microscopy of a T cell from a DOCK8-deficient
patient migrating in collagen matrix. PI (orange) added when indicated. Shown is a representative of three patients and three healthy controls, from three
experiments. (B) Flow cytometric quantification of dead and dying cells by Annexin V and PI staining, after collagenase treatment to recover unstained
cells. Normal human T cells were transfected with either DOCK8 (open symbols) or nonspecific (NS) siRNA (filled symbols), and allowed to migrate in col-
lagen matrices (solid line) or medium (dotted line) for the indicated times. Shown is a representative of three experiments. (C) Similar analysis as for B,
after migration in medium or collagen for 24 h, of NK cells from six Dock8-deficient (KO) or control (WT) mice from three experiments. (D) Percentages of
time spent elongated, for those Dock8-deficient T cells that died or remained alive after migrating in collagen matrix for 16 h. (E) Similar to D, except that
duration of elongation episodes was measured for each single episode of elongation that immediately preceded the cell fragmenting (F), or for all other
elongation episodes (NF). Bar, 20 m. Three patients were tested in D and E from three experiments. CE show means SD. Two-way ANOVA was per-
formed for C. Unpaired Students t test was performed for D and E. Statistical significance indicated by *, P < 0.05; ***, P < 0.001; ns, nonsignificant.

collagen-coated 2D (Fig. 6 E) flat surfaces. Although integrin- DOCK8, through CDC42 and p21-activated kinase (PAK),
mediated strong adhesive forces are not normally required for regulates lymphocyte shape integrity to coordinate
leukocyte locomotion within a 3D environment (Lmmermann cytoskeletal structures during cell movement
et al., 2008), the sometimes tethered appearance of elongated Lymphocyte migration in the skin presents a challenge espe-
DOCK8-deficient cells (Video 1) raised the question of whether cially for the nucleus, which is normally the largest and least
such adhesion contributed to the abnormal phenotype. How- deformable organelle (Friedl et al., 2011). Cell body shape
ever, this explanation seemed unlikely because agarose and change must at some level be coordinated with nuclear shape
polycarbonate are not ligands for integrin receptors yet could change during cell migration. Given the elongation phenotype
elicit the abnormal phenotype (Fig. 6, B and C). Elongation was of DOCK8-deficient cells described above, DOCK8 likely
also unaffected by disrupting interactions between integrin regulates this architectural machinery. The small Rho GTPases
1 collagen receptors on patient cells with collagen-containing CDC42 and RAC serve as molecular switches to control di-
gel matrix by either adding anti-integrin 1 subunit antibod- verse biological processes, including cell morphogenesis and
ies to the collagen gel matrix (Fig. 6 E) or silencing its expres- migration ( Jaffe and Hall, 2005); moreover, DOCK8 can ac-
sion within T cells from a DOCK8-deficient patient (Fig. 6 F). tivate CDC42 and RAC (Harada et al., 2012; Mou et al.,
Together, these results argue that the abnormal shape integ- 2012), regulating CDC42 to facilitate dendritic cell migration
rity of DOCK8-deficient cells was elicited solely by the con- (Harada et al., 2012). We therefore investigated the possible
fined spaces that constrained cell movement. This could explain contribution of these small GTPases to the DOCK8-deficient
why the cellular phenotype could vary from tissue to tissue in phenotype of lymphocytes. Knockdown of CDC42 but not
the body, depending on their physical characteristics, and why RAC1/2 in T cells from normal donors recapitulated the
disease in the patients manifests as an immunodeficiency with DOCK8-deficient phenotype of cell elongation, nuclear de-
disproportionate involvement of skin, whose tight adhesive formation, and cell death (Fig. 7, AC). CDC42 in turn acti-
junctions between epidermal cells and tight spacing between vates multiple effectors, including PAK and Wiskott-Aldrich
dermal collagen bands demand marked lymphocyte shape de- syndrome protein (WASP). Treatment of normal T cells with
formation for effective migration. the class I PAK small molecule kinase inhibitor IPA3, or with

JEM Vol. 211, No. 13 2553


PAK1/2 siRNA, also phenocopied loss of DOCK8 or CDC42
(Fig. 7, DF), whereas loss of WASP expression in T cells
showed no such effect (Fig. 7, G and H). Similar results were
seen for T cells from a Wiskott-Aldrich syndrome patient or
Was KO mice (unpublished data). Loss of DOCK8, CDC42,
or PAK1/2 expression or function did not reciprocally decrease
each others protein levels (unpublished data; Akbar et al., 2011).
These results suggest that DOCK8 acts proximally with CDC42
and PAK in regulating lymphocyte shape integrity, most likely
through complex spatial and temporal effects on actin, myo-
sin, and microtubule cytoskeletal structures (Bokoch, 2003;
Li and Gundersen, 2008). Indeed, the absence of DOCK8,
CDC42, or PAK activity resulted in cytoskeletal discoordina-
tion between the front and rear of migrating cells, seen as ab-
normal cellular elongation with slightly decreased total F-actin
polymerization present at both poles and abnormal position-
ing of the microtubules including the microtubule organizing
center (Fig. 8).

Defective cell integrity occurs


during response to viral skin infections
Viral replication in the skin occurs primarily within keratino-
cytes in the epidermis; however, some viruses also infect der-
mal cells such as fibroblasts or neurons at the epidermal-dermal
junction (Cunningham et al., 1985; Drijkoningen et al., 1988;
Muraki et al., 1992; Nikkels et al., 1996). This places special
demands on effector immune cells that must successfully nav-
igate into and through dermis after exiting dermal vessels and
then migrate further into the densely connected cell layers of
the epidermis to mediate host defense. To assess the function
of Dock8-deficient T cells during a viral infection, we infected
mice with HSV, which is the only virus causing severe skin in-
fection in patients that is also capable of infecting mice. When
HSV is inoculated in the epidermis on the flank, it establishes
infection within the dorsal root ganglion before spreading
through the nerves, from which it emerges to cause a skin rash
in a dermatomal distribution (Fig. 9 A; Simmons and Nash,
1984). Containment of the rash reflects the ability of CD8
T cells to limit secondary spread of the virus down the flank
(van Lint et al., 2004). Upon acute infection, Dock8-deficient
mice developed herpetic skin disease that was more severe with
increased mortality when compared with WT mice (Fig. 9,
AC). To visualize T cells that were responding within the in-
Figure 5. Cytothripsis is biochemically distinct from apoptosis.
(A) Scanning electron micrographs of control or patient T cells after mi- fected skin, we adoptively transferred Dock8-deficient or con-
grating in collagen gel matrices. (B) Mitochondrial membrane potentials trol WT T cells into normal mice. The recipient mice were
of patient or control T cells migrating in collagen gels. A decrease in the
ratio of red (JC-1 aggregates) to green (JC-1 monomer) is indicative of

loss of potential. (C) Proportions of T cells expressing active caspase-3
after migrating in collagen gels for the indicated times. Eight controls and with collagenase. All panels show a positive control in which apoptosis
four patients were tested from four experiments. (D and E) Immuno was induced in healthy donor T cells within the collagen gels using anti-
blotting for highly active cleaved caspase-8 (D) and caspase-9 (E) proteins. FAS antibodies with Protein A cross-linking. Cells were analyzed after
Lysates were prepared from control or patient T cells that had migrated in migrating within the collagen gels for the indicated times or as described
medium (0) or collagen gels for the indicated times. The black line shows in the Materials and methods. A, B, D, and E show representatives of three
that intervening lanes were spliced out. (F) Inability to block cytothripsis tested patients and controls from three experiments. Bars, 10 m. C and F
with the pan-caspase inhibitor zVAD. Annexin V+ or PI+ staining of mi- show means SD. Unpaired Students t test was performed for C and F
grating cells (nine controls, four patients, from four experiments) was (right), and two-way ANOVA was performed for F (left). Statistical signifi-
quantitated by flow cytometry after recovery of unstained cells from gels cance indicated by *, P < 0.05; ****, P < 0.0001; ns, nonsignificant.

2554 DOCK8 and cytothripsis in skin viral infections | Zhang et al.


Ar ticle

Figure 6. Requirement for migration through confined spaces but not for adhesion in eliciting loss of shape integrity. (A) Proportions of
T cells (nine controls, four patients, from four experiments) that were Annexin V+ or PI+, after migration in increasing collagen concentrations for 24 h in
collagen gels of increasing matrix density. (B) Confocal and diffusion interference contrast (DIC) microscopy of control (green) and patient (red) T cells
while migrating through transwell pores (orange arrowheads) toward CXCL12. Hoechst, blue. Representative of three patients and three controls from
three experiments. (C) Percentage of time T cells (six controls, three patients, from three experiments) spent elongated in 0.2% agarose gel matrices.
(D) Proportions of T cells (eight controls, four patients, from three experiments) elongated during migration on 2D ICAM-coated plates or in 3D collagen
matrices. (E) Proportions of DOCK8-deficient T cells elongated after migration on collagen-coated slides (2D) or in collagen gels (3D), untreated or with
antiintegrin 1 blocking antibodies. Means are shown for three patients and three controls tested under each condition from three experiments. Two-
way ANOVA was performed to compare treatment with or without antibody. (F) Similar to E except that one patient and one control were tested after
transfection of nonspecific or ITGB1 siRNA. Median fluorescence intensities of CD29 for control cells were 6,386 (NS siRNA) and 2,019 (ITGB1 siRNA), and
for patient cells were 6,363 (NS siRNA) and 1,396 (ITGB1 siRNA). Bars, 10 m. A, C, and D show means SD. Linear mixed effects modeling was per-
formed for A, unpaired two-tailed Students t test for C and D, and two-way ANOVA to compare treatment with or without antibody for E. Statistical
significance indicated by ***, P < 0.001; ****, P < 0.0001; ns, nonsignificant.

infected epicutaneously with HSV, and the donor T cells were they migrate (Zaid et al., 2014). Thus, we hypothesized that
visualized in the skin using intravital two-photon microscopy cytothripsis, associated with defective shape integrity, could
(Gebhardt et al., 2011). In contrast to normal T cells, respond- impair the formation or survival of epidermal TRM. This would
ing Dock8-deficient T cells showed increased elongation and decrease the effective virus-specific T cell concentration below
fragmentation as they migrated within the combined epider- that needed for viral clearance, despite substantial remaining
mal and dermal layers, indicating that DOCK8 regulates cell cytotoxic activity and cytokine production during the immune
shape integrity in vivo (Fig. 9, D and E; and Video 4). response (Zhang et al., 2009; Lambe et al., 2011; Randall et al.,
2011). Other infected organs and normal secondary lymphoid
DOCK8 regulates skin tissueresident memory cell tissue, in contrast, would offer a less confined environment
formation for protection against viral infection for cell migration and reduce these damaging effects on anti-
During HSV infection, control of replicating virus requires a pathogen immune cells. To test this, normal mice were trans-
high density of CD8 T cells, which migrate into the epidermis ferred with equal mixtures of Dock8-deficient and control
and develop into tissue-resident memory CD8 T cells (TRM WT T cells, both also expressing the same transgenic TCR that
cells; Gebhardt et al., 2009; Zhu et al., 2013). These cells per- recognizes an MHC class Irestricted immunodominant HSV
sist indefinitely after acute inflammation resolves within the peptide (Mueller et al., 2002). After epicutaneous infection,
epidermis, where they continuously migrate to protect against Dock8-deficient effector T cells expanded and accumulated
viral reinfection or emergence from latent infection in the skin in secondary lymphoid organs and skin with only slightly re-
(Gebhardt et al., 2011; Ariotti et al., 2012; Jiang et al., 2012; duced efficiencies as compared with control T cells (Fig. 10 A).
Mackay et al., 2012; Mueller et al., 2013; Zaid et al., 2014). Consistent with previous reports that Dock8-deficient CD8
In the epidermis, TRM cells adopt a highly ramified cell shape T cells had reduced long-term survival (Lambe et al., 2011;
that is defined by the constraints of the tissue through which Randall et al., 2011), these cells were disproportionately

JEM Vol. 211, No. 13 2555


Figure 7. DOCK8 signals through CDC42
and PAK to maintain shape integrity.
(A) Top: Proportions of T cells elongated during
migration in collagen gel matrices. Normal
human T cells were transfected with nonspe-
cific (NS), DOCK8, CDC42, or RAC1/2 siRNA.
Bottom: representative nuclear morphology of
these cells migrating in collagen gels. BF,
brightfield. DAPI, blue. (B) Proportions of dead
or dying cells were quantitated by flow cyto-
metric analysis of Annexin V and PI staining,
after cells from A were allowed to migrate in
collagen gels or in medium. (C) Knockdown
efficiencies for A and B. Quantitative real-time
RT-PCR to measure transcript levels of DOCK8,
CDC42, RAC1, and RAC2, normalized to -actin.
Knockdown efficiencies of these genes were
calculated as normalized levels in T cells trans-
fected with indicated siRNA, divided by nor-
malized levels in T cells transfected with
nonspecific siRNA. Ratios are means SD of
five independent experiments. (D and E) Pro-
portions elongated within collagen gels of
normal human T cells either treated with
DMSO or the PAK inhibitor IPA3 (D), or trans-
fected with NS or PAK1/2 siRNA (E). (F) Similar
analysis as in B, using cells from E. Represen-
tative immunoblot showing efficiency of
PAK1/2 knockdown. (G) Morphology of T cells
in which WAS gene expression was silenced,
during migration in collagen gel. (H) Propor-
tions of Annexin V+ or PI+ cells, after migrating
in collagen gel matrices or medium. Normal
human T cells were transfected with NS, WAS,
or DOCK8 siRNA. Representative immunoblot
showing WASP knockdown efficiency in human
T cells. Bars, 10 m. A, B, D, E, G, and H are
representative of three independent experi-
ments, and F of five experiments. AF and H
show means SD. One-way ANOVA was per-
formed for A, two-way ANOVA for B, F, and H,
and unpaired two-tailed Students t test for
D and E. Statistical significance indicated by
*, P < 0.05; **, P < 0.01; ***, P < 0.001; ****,
P < 0.0001; ns, nonsignificant.

decreased in the spleens as infection resolved and immuno- T cells from Dock8-deficient or control TCR transgenic mice
logical memory developed (Fig. 10, B and C). However, the directly into the skin. This method circumvents any defects in
survival defect was markedly more pronounced in the skin, T cell priming and migration to the skin, allowing direct as-
where up to 180 times more WT than Dock8-deficient mem- sessment of the ability of the cells to enter the epidermis and
ory T cells were recovered after 1 mo (Fig. 10 B). When we develop into TRM cells (Mackay et al., 2013). Dock8-deficient
examined the skin in these mice by intravital two-photon T cells rapidly disappeared in the skin and the few surviving
microscopy, Dock8-deficient T cells were initially rare but cells did not up-regulate expression of CD69 and CD103,
became largely undetectable by 1 mo after infection, despite which are necessary for TRM formation (Fig. 10, F and G).
normal numbers of control T cells (Fig. 10 D and Video 5). This Defective TRM survival with severe loss of cells over time re-
was primarily due to decreased numbers of CD69hi CD103+ sulted in minimal protection against challenge with HSV, as
Dock8-deficient TRM, which failed to survive in the skin after measured by viral titers in the skin (Fig. 10 H) after transfer
HSV infection (Fig. 10 E). of effector T cells and topical treatment of recipient mice with
To analyze the formation of CD103+CD8+ TRM cells the chemical sensitizer 2,4-dinitrofluorobenzene (DNFB) to
in the skin, we cotransferred in vitro activated effector CD8 generate TRM (Mackay et al., 2012). Together, these results

2556 DOCK8 and cytothripsis in skin viral infections | Zhang et al.


Ar ticle

Figure 8. Abnormal cytoskeletal organization in elongated


cells. In-gel confocal imaging for F-actin (phalloidin, red),
-tubulin (green), and nuclei (DAPI, blue), in normal human
T cells transfected with nonspecific (NS), DOCK8, or CDC42
siRNA, or treated with the PAK1/2 inhibitor IPA3. Representa-
tive of three experiments. Bars, 10 m.

indicate that DOCK8, by regulating cell shape integrity, is migration. This process can be elicited by the physical prop-
critical in vivo for CD8 T cell persistence, TRM formation, and erties of tissues through which the cells migrate, and it is
antiviral immunity in the skin. characterized by cell elongation and nuclear deformation dur-
ing prolonged migration through confined spaces.The result-
DISCUSSION ing mechanical forces experienced by the cell likely cause breaks
While studying the behavior of DOCK8-deficient T cells in the plasma membrane and nucleus that lead to catastrophic
and NK cells, we have unexpectedly discovered a new form cell death without inducing any biochemical markers of apop-
of cell death we term cytothripsis for cell shattering. Cyto- tosis. Cytothripsis can impair effector T cell and NK cell func-
thripsis occurs when shape maintenance fails during lymphocyte tions by limiting their effective motility and decreasing their

Figure 9. Defective cell integrity occurs


during response to viral skin infections.
(A) Representative skin disease, 7 d after
epicutaneous HSV infection of WT or Dock8-
deficient (KO) mice. Blue arrowheads, primary
infection sites. Red arrowhead, blistering
along infected dermatome. Orange arrow-
head, blistering with skin necrosis. (B) Daily
scoring of disease severity after acute HSV
infection of WT (black) or KO (red) mice.
(C) Kaplan-Meier survival curves for the mice
from (B). (D) Two-photon imaging of Dock8-
deficient cpm (green) or Dock8-replete T cells
from WT gBT-I mice (red) migrating in the
dermis, 7 d after adoptive cotransfer into WT
recipient mice and HSV infection. White
arrow, fragmentation. Frames show time
elapsed (minutes). (E) Proportions elongated
of the cotransferred T cells in D. Bars, 15 m.
B shows means SEM, with a total of 10 or
14 mice per group from two experiments.
E shows means SD, where data were pooled
from six mice from two experiments. Wilcoxon
matched-pairs signed rank sum test (two-tailed)
was performed for B, log-rank (Mantel-Cox)
test for C, unpaired two-tailed Students
t test for E. Statistical significance indicated
by *, P < 0.05; ***, P < 0.001.

JEM Vol. 211, No. 13 2557


Figure 10. Dock8 regulates shape integ-
rity and survival of TRM cells in vivo to
control viral skin disease. (A) Numbers of
WT and Dock8-deficient HSV-specific gBT-I T
cells in the spleen, brachial LNs and skin, 7 d
after adoptive transfer (1:1 mixture) into WT
recipient mice and HSV infection. (B) Relative
numbers of WT gBT-I and Dock8-deficient
cpm gBT-I T cells in spleen and infected skin,
quantified by flow cytometry at the indicated
times after infection. (C) Numbers of WT and
Dock8-deficient gBT-I T cells in the spleen at
various times after skin infection. (D) Two-
photon imaging of skin in C on day 17 after
infection. White arrows, Dock8-deficient cells.
(E) Numbers of WT and Dock8-deficient gBT-I
T cells in skin (CD103+ CD69+ TRM) in C.
(F) Numbers of WT and Dock8-deficient skin
(CD103+ CD69+) TRM, at 23 d after intradermal
cotransfer of in vitroactivated CD8 T cells
into WT recipient mice. (G) Proportions of
CD103+ CD69+ gBT-I TRM that were WT or
Dock8/ in skin in F. (H) Impaired control of
HSV skin infection by virus-specific memory
cells. WT mice received no cells (control) or
in vitro activated effector CD8 T cells from WT
gBT-I or Dock8-deficient cpm gBT-I mice.
After topical treatment with DNFB, mice were
infected at the same site 30 d later with HSV
and viral titers measured on day 5. Bar, 100 m. AC and EH show means SD. For A, data were pooled from four experiments with a total of 20 mice
per group. For B, C, and E, data were pooled from four experiments for the day 7 and 31 time points and two experiments for the day 14 time point, with
a total of 1020 mice per time point. F and G are representative of three experiments with seven mice per group. For H, data are from three experiments
with a total of 1315 mice per group. Two-way ANOVA was performed for AC and E, unpaired two-tailed Students t test for F and G, and one-way
ANOVA for H. Statistical significance indicated by **, P < 0.01; ***, P < 0.001; ****, P < 0.001; ns, nonsignificant.

capacity to reach virally infected cells within the tissue in a GEFs become stimulated to activate CDC42, as well as which
viable state. Skin is among the densest tissues within the body effectors downstream of CDC42 become selectively acti-
and therefore most likely to elicit cytothripsis, given that vated to carry out the different shape rearrangements for
both epidermis and dermis have many highly confined spaces. these cellular functions (Sinha and Yang, 2008). This could
Although collagen content is a main determinant of tissue explain why DOCK8- and WAS-deficient cells differ in their
density in the dermis (Lowry et al., 1941), other environmen- migration behavior, and why DOCK8 could contribute not
tal constraints, such as the cellular density and tight junctions only to the striking migration defects we have now observed
in the epidermis, likely also define the migratory capacity of but also to the lymphopenia and other functional defects that
leukocytes in tissues. Tissues of intermediate density, such as have been reported in DOCK8-deficient lymphocytes.
the walls of large arteries, may also elicit cytothripsis, thereby Dock8-deficient T cells were previously reported to ex-
contributing to the vasculopathy associated with local virus hibit a long-term survival defect, as shown by their competi-
reactivation that has been documented in some DOCK8- tive disadvantage after adoptive transfers of mixtures of mutant
deficient patients (Sabry et al., 2014; unpublished data). and WT cells (Lambe et al., 2011; Randall et al., 2011). Fur-
Maintenance of lymphocyte shape integrity, which pre- thermore, fewer memory CD8 T cells persisted in Dock8-
vents cytothripsis, is regulated by DOCK8 which, together deficient mice after experimental influenza virus infections
with CDC42 and PAK, coordinates cytoskeletal structures (Lambe et al., 2011; Randall et al., 2011). Our results during
during cell migration. The demand for precise coordination experimental HSV skin infections also indicated that Dock8
of cytoskeletal structures is greatest in cells when they ma- mutant T cells are at a competitive disadvantage. However, it
neuver through highly confined spaces, as compared with open is unlikely that they would survive better in a noncompeti-
spaces. In lymphocytes, coordination of cytoskeletal struc- tive situation, or at least not sufficiently to restore function,
tures is also required during immune synapse formation for given that transfer separately of either WT or Dock8 mutant
cytotoxicity, directed cytokine secretion, assembly of signaling HSV-specific T cells, followed by challenge of mice after es-
platforms for activation and proliferation, and cell division tablishment of memory, conferred no protection in terms of
(Dustin, 2008). External signals determine which of many viral replication. Interestingly, mice conditionally deficient in

2558 DOCK8 and cytothripsis in skin viral infections | Zhang et al.


Ar ticle

Cdc42 also showed increased ex vivo expression of apoptotic syndrome, which result from impaired chemotaxis, loss of
markers in their peripheral T cells, which could be rescued in peripheral NK cell and dendritic cell differentiation, and/or
vivo by overexpressing PAK1 (Guo et al., 2010). Although defective actin polymerization, can also be associated with
the decreased survival was attributed to small decreases in viral skin infections, although the skin infections tend to be
IL-7 receptor expression for survival signals (Guo et al., 2010; of narrower spectrum or later onset than DOCK8 deficiency
Randall et al., 2011), our results suggest another mechanism (Sullivan et al., 1994; Imai et al., 2004; Dotta et al., 2011;
whereby survival depends not only upon access to survival Sanal et al., 2012; Spinner et al., 2014). Interestingly, in the
signals (unpublished data) but also whether the cell can with- Wiskott-Aldrich syndrome, the defective actin polymeriza-
stand physical stresses as it migrates through the body. Such tion affects many T cell functions including thymopoiesis,
stress may have a cumulative effect on trafficking T cells that chemotaxis, activation, proliferation, cytokine production,
is pronounced in certain tissues, such as the skin. and cytotoxicity (Zhang et al., 1999; Snapper et al., 2005;
The different migratory patterns of T cells allow them to De Meester et al., 2010; Lang et al., 2013). However, when
perform their important immunosurveillance functions of pre- compared with DOCK8 deficiency, the lack of abnormal elon-
venting reinfection or controlling viral reactivation through- gation and cytothripsis and less frequent occurrence of recur-
out the body (Mueller et al., 2013). To accomplish this, naive rent viral skin infections in Wiskott-Aldrich syndrome patients
and central memory T cells continually migrate between blood (Sullivan et al., 1994) support the concept that lymphocyte
and secondary lymphoid organs, whereas effector memory shape integrity contributes to normal protection against viral
T cells also migrate through peripheral tissues. When they skin infections.
circulate through skin, effector memory T cells can be found In summary, our work has now revealed that the ability to
moving rapidly through the dermis en route to draining LNs. maintain cell shape integrity is a critical determinant of im-
In contrast, CD8 memory T cells in the skin do not recircu- mune function in rapidly motile cells such as lymphocytes.
late throughout the body but instead remain indefinitely Although we have focused on the role of lymphocyte cell shape
within the epidermis where they continuously move. TRM integrity in antiviral immunity, our findings are likely to have
cells also form in other nonlymphoid tissues in response to general relevance for skin immunity. For example, DOCK8-
infections, including the intestines, reproductive tract, and lungs, deficient patients also often have bacterial or fungal skin infec-
where they are predominantly associated with epithelial tions, and are at increased risk of developing skin cancers. We
layers (Mueller et al., 2013). Although the ability of Dock8- speculate that these conditions result from locally impaired
deficient T cells to form TRM populations in those tissues has defense against those pathogens, and possibly impaired tumor
yet to be examined, the unique microanatomy of those tissues surveillance, when lymphocytes undergo cytothripsis as they
may differ from the skin in having less confinement, which navigate to lesions in skin tissues. Dock8-deficient dendritic
could facilitate some degree of TRM generation. This would cells also show more globally impaired interstitial migration
help explain the disproportionate number and severity of viral that might further compromise skin immunity (Harada et al.,
skin infections in DOCK8-deficient patients. Nevertheless, 2012). Maintenance of lymphocyte shape integrity may also
reduced TRM in the lungs or mucosal surfaces may contribute become a factor when cells repeatedly traverse less dense tis-
to the recurrent sinopulmonary infections and chronic ano- sues such as blood vessel walls or lymphoid tissues. Because
genital viral infections in DOCK8-deficient patients. More- many T cell populations recirculate between blood, lymphat-
over, even during acute infections, T cells may be subjected ics, and other tissues, our results may mechanistically explain
to physical stresses from the highly confined skin environ- the milder systemic defect in peripheral CD8 memory T cell
ment. This could lead to the increased morbidity and mortal- survival and attrition of naive T cell numbers in DOCK8-
ity we saw during epicutaneous HSV infections, in contrast deficient humans or mice (Lambe et al., 2011; Randall et al.,
to the normal morbidity previously seen during intranasally 2011). Finally, our results suggest a more general conclusion
inoculated influenza virus infections of Dock8-deficient mice that loss of shape integrity during the navigation of nonim-
(Lambe et al., 2011). mune cells in other migratory processes, such as during em-
Collectively, our data are highly suggestive that the ab- bryonic development, could contribute to human disease.
normal elongation phenotype is responsible for T cell death
in vivo and that this process prevents TRM formation during
MATERIALS AND METHODS
viral infections. However, other explanations, including im-
Patients. Whole blood and leukapheresis samples were obtained from
paired migration and proliferation or alternative mechanisms mutation- and immunoblot-confirmed DOCK8-deficient patients, their rela-
of cell death, remain possible. These appear less likely given tives, or paid healthy volunteers. Only those patients without somatic rever-
our results showing normal chemotaxis, impaired TRM for- sion or at most 25% of revertant T cells (Jing et al., 2014) were used for the
mation even when activated T cells were directly transferred studies described here. These individuals gave written informed consent to
to the skin, and normal proliferation and activation of T cells participate in research protocols approved by National Institutes of Health
(NIH) Institutional Review Boards. Buffy coat cells, which were by-products
from Dock8-deficient mice (Lambe et al., 2011). Nevertheless,
of volunteer-donor blood units, and human foreskin tissues (gift from C. Yee,
other immunodeficiencies demonstrate that additional mecha- National Cancer Institute, Bethesda, MD), which were discarded after rou-
nisms exist to control viral skin infections. For example, the tine newborn circumcisions, were distributed in an anonymized manner.
WHIM syndrome, GATA2 deficiency, or the Wiskott-Aldrich These were exempted from need for informed consent and Institutional

JEM Vol. 211, No. 13 2559


Review Board review, as determined by the NIH Office of Human Subjects (Zhang et al., 2009). Samples were acquired on a FACSCanto instrument
Research Protection. using FACSDiva software (BD). Analyses were performed using the FlowJo
software version 9 and higher (Tree Star).
Mice. Mice were bred and used under animal study protocols approved by
the NIAID Animal Care Use Committee, UK Home Office License, and Migration studies in human foreskin tissues. Human foreskin tissues
the University of Melbourne Animal Ethics Committee. Dock8-deficient discarded after routine newborn circumcisions were stored in HBSS at 4C
cpm/cpm (cpm) mutant mice, generated by ENU mutagenesis from a pure for 37 d before use. Tissues were warmed to 37C overnight in PBS sup-
C57BL/6 background, gBT-I TCR transgenic mice recognizing an MHC plemented with 10% FBS, cut into 5 5 mm pieces, and the epidermis
I-restricted HSV-1 gB epitope, and Actb-DsRed transgenic mice were pre- carefully peeled off using a small forceps. T cells, labeled with 0.5 M Cell-
viously described (Mueller et al., 2002; Vintersten et al., 2004; Randall et al., Tracker Green CMFDA (5-chloromethylfluorescein diacetate; Life Tech-
2009). To obtain Dock8-deficient GFP-expressing mice (GFP-cpm) for cell nologies), were dropped onto the exposed dermis and allowed to migrate
tracking experiments, cpm mice were crossed to UBI-GFP transgenic mice into the tissues at 37C for 3 h. Cells remaining on the dermal surface were
on a C56BL/6 background (The Jackson Laboratory; Schaefer et al., 2001). gently rinsed away, and tissues immersed in complete medium within
Mice were further crossed with gBT-I TCR transgenic mice to enable as- Lab-Tek II 8-well chambered coverglasses (Nunc). Live cell imaging was per-
sessments of virus-specific T cells. WT gBT-I TCR transgenic mice were formed at 37C in the presence of 5% CO2, using a TCS SP5 laser scanning
crossed to Actb-DsRed transgenic mice for use as controls. In some experi- confocal microscope (Leica) with excitation at 488 nm. Image stacks were
ments, Dock8-deficient KO mice were used instead of cpm mice. KO mice collected with a 4-m vertical step size at a depth of 4060 m. Collagen was
were generated from targeted deletion of exons 9 and 10 in pure C57BL/6 visualized by refractive imaging.
ES cells. WT control C57BL/6 mice were purchased from Taconic or The
Jackson Laboratory. Mice were sacrificed by carbon dioxide asphyxiation or
cervical dislocation for harvest of LNs and spleens. RNA interference. Human T cells, 47 d after initial activation, were nu-
cleofected using the Lonza 96-well shuttle system according to the manufac-
Cell isolation for in vitro experiments. Human peripheral blood mono- turers optimized protocol. Stealth select prevalidated siRNA against DOCK8,
nuclear cells (PBMCs) were isolated from peripheral blood products by ITGB1, CDC42, RAC1, RAC2, PAK1, PAK2, WAS, or nonspecific nega-
Ficoll-Paque PLUS density gradient centrifugation (GE Healthcare). T cells tive control siRNA were used (Life Technologies). To optimize knockdowns,
were purified from PBMC by negative selection using human panT cell cells were nucleofected 2 d in a row, and the dead cells removed by Ficoll-Paque
isolation kit II (Miltenyi Biotec). In some samples, eosinophils were further PLUS gradient centrifugation on the third day. Experiments were performed
depleted using human CD15 microbeads (Miltenyi Biotec). T cells were ac- and lysates harvested to analyze RNA or protein expression on the fourth
tivated using beads coated with anti-CD2, -CD3, and -CD28 antibodies day after nucleofection. For PAK1/2 knockdown, cells were nucleofected
provided as a T cell activation/expansion kit (Miltenyi Biotec), using a bead every other day for four times, the dead cells removed by Ficoll-Paque PLUS
to cell ratio of 1. Beads were removed on the fourth day after activation, and gradient centrifugation 1 d later, and used for experiments and evaluation of
cells were expanded in 100 U/ml of recombinant human IL-2 (Aldesleukin; knockdown efficiency on the second day after the last nucleofection.
Prometheus). Activated T cells were cultured for a total of 445 d, when
they acquired an activated effector/memory phenotype, as reflected by loss Immunoblotting. Immunoblotting for DOCK8 protein and -actin was
of CD45RA and CCR7 surface marker expression, that was comparable be- performed as previously described (Zhang et al., 2009). For analysis of other
tween patients and healthy normal controls. The proportions of revertant proteins, cells were lysed in 2% SDS (50 mM Tris HCl, pH 6.8, 10% glycerol)
cells were determined in one patient, which appeared stable or mildly in- or 1% NP-40 (1 mM EDTA, 50 mM Hepes, and 150 mM NaCl) containing
creased, ranging from 18% on day 2, 14% on day 8, and 26% on day 21 dur- complete protease inhibitors (Roche). Protein lysates were separated on Nu-
ing culture. Human NK cells were purified by positive selection using CD56 PAGE Novex 12% or 412% gradient Bis-Tris gels using MOPS SDS or
microbeads (Miltenyi Biotec) and expanded in 100 U/ml of recombinant MES running buffers (Life Technologies), followed by wet or semi-dry trans-
human IL-2 for 68 d. Human T cells and NK cells were typically >90 and fer. Membranes were blocked with 5% blocking grade nonfat milk (Bio-Rad
>85% purity, respectively, when used in experiments. Laboratories) or blk-CH (Millipore). Antibodies were to caspase-9 (BD);
Mouse LNs and spleens were minced into single cell suspensions and caspase-8 (clone C15; gift from L. Zheng, NIAID, NIH, Bethesda, MD); and
passed through a cell strainer. Splenocytes were also treated with ACK lysing PAK1, PAK2, and WASP (Santa Cruz Biotechnology, Inc.).
buffer (Quality Biological). Mouse LN T cells or splenic NK cells were puri-
fied by negative selection, using mouse panT cell or NK cell isolation kits II
Quantitative real-time RT-PCR. Total RNA from transfected primary
(Miltenyi Biotec). Plates were coated overnight with anti-CD3 and anti-CD28
T cells was isolated using the RNeasy mini kit with DNaseI in-column di-
antibodies (BD) at 3 g/ml in PBS to which purified panT cells were added.
gestion (QIAGEN). 0.9 g of total RNA was reverse-transcribed with Su-
After activation for the first 3 d, T cells were expanded in 200 U/ml of re-
perscript III first-strand synthesis supermix (Invitrogen). Diluted cDNA was
combinant human IL-2. Purified mouse T cells were cultured for a total of
analyzed by quantitative real-time PCR using Power SYBR Green PCR
57 d. Purified mouse NK cells were activated and expanded with 1,000 U/ml
master mix on a 7500 Real Time PCR System (Applied Biosystems). Stan-
of recombinant human IL-2 for 67 d. Mouse T cells and NK cells were
dard conditions of 40 cycles (95C for 15 s, and 60C for 1 min) were used.
typically >90% purity when used in experiments.
RNA quantity was calculated from the cycle number by using primer-specific
Human and mouse cells were grown in complete medium, consisting of
standard curves. Primer sets designed to span exon junctions were as follows:
RPMI medium supplemented with 10% FBS, l-glutamine, penicillin, strep-
DOCK8 forward (F), 5-TCAGCCTCTGTGGGTAGACA-3, and reverse
tomycin, and recombinant human IL-2. Unless otherwise indicated, cells
(R), 5-CCGCACAAAGAGATTTTGGA-3; CDC42 F, 5-CGCCCA-
were kept in the presence of IL-2 throughout subsequent experiments, in-
CAACAACACACTTA-3, and R, 5-CCCGGTGGAGAAGCTGAG-3;
cluding within 3D matrices.
RAC1 F, 5-TCTCCAGGAAATGCATTGGT-3, and R, 5-CTGATG-
CAGGCCATCAAGT-3; RAC2 F, 5-TTGCTGTCCACCATCA-
Lymphocyte immunophenotyping. Standard flow cytometry methods CATT-3, and R, 5-AGACCTGCCTTCTCATCAGC-3; and -actin
were used to evaluate cell surface marker expression using various combina- F, 5-GTTGTCGACGACGAGCG-3, and R, 5-GCACAGAGCCTC-
tions of FITC, PE, PE-Cy5, PE-Cy7, APC, APC-Cy7conjugated anti-CD3, GCCTT-3. Expression of each gene was normalized to the -actin house-
anti-CD4, anti-CD16, anti-CD25, anti-CD29, anti-CD45RA, anti-CD56, and keeping gene. Knockdown efficiencies of genes were calculated as normalized
PE anti-CXCR4 antibodies (Life Technologies; the rest of the antibodies levels in T cells transfected with indicated siRNA, divided by normalized
are from BD). CFSE dilution assays were performed as previously described levels in T cells transfected with nonspecific siRNA.

2560 DOCK8 and cytothripsis in skin viral infections | Zhang et al.


Ar ticle

Collagen gel migration assays for morphological analyses. Collagen setting of 0.150.2 m. Where indicated, after staining gels were physically
solution from bovine skin (Sigma-Aldrich) was mixed with 10 RPMI 1640 compressed to facilitate simultaneous visualization of all vertical stacks.
medium (Life Technologies) and FBS, to a final concentration of 3.6 mg/ml
(unless otherwise indicated) of collagen, 1 RPMI, and 10% FBS. l-glutamine, Cell cycle analysis. Cell cycle analysis was performed according to standard
penicillin, streptomycin, and 200 U/ml of recombinant human IL-2 were protocols (Darzynkiewicz and Huang, 2004). In brief, after fixation in 70%
also added. The collagen mixture was stored at 4C for no more than 10 d ethanol on ice for 2 h, cells were stained with 20 g/ml of PI in PBS contain-
before use. Cells were then admixed at a final concentration of up to 5 ing 0.1% Triton-100 and 0.2 mg/ml DNase-free RNase A (QIAGEN) at
107 cells/ml and the collagen gel matrix polymerized at 37C for at least 37C for 15 min. DNA content was acquired on a FACSCanto using a linear
1 h in Lab-Tek II 8-well chambered coverglasses (Nunc). In initial experi- fluorescence amplification scale. Area versus width was used to gate out dou-
ments, addition to bovine skin collagen of fibronectin at 6 g/ml and mouse blets. To calculate cell cycle, data were analyzed by using the Dean-Jett-Fox
laminin at 2.5 g/ml (both from Life Technologies), or substitution of Cul- model on the Cell Cycle analysis platform of FlowJo.
trex rat tail collagen (Trevigen) for bovine skin collagen, made no difference.
Therefore, bovine collagen alone was used in all subsequent experiments. Cell cycle blockade. Patient cells that had been expanded in culture were
Where indicated, small molecule inhibitors, or DMSO used as a vehicle enriched for viable cells by Ficoll-Paque PLUS density gradient centrifuga-
control, were mixed with the collagen matrix at 4C, before addition of cells tion before use in experiments. Cells were treated for 48 h with aphidicolin
and polymerization of gels. Minimal doses that exerted effects while avoiding at 2 g/ml (Sigma-Aldrich) or were treated with DMSO. Cells were kept in
nonspecific toxicity were used. In some experiments, Z-Val-Ala-Asp (OMe)- the presence or absence of aphidicolin when mixed into the collagen mixture
FMK (zVAD) (MP Biomedicals) was added at 40 or 80 M to gels for 1218 h and the gel allowed to polymerize. After migration for 24 h in the gel, cells
to inhibit caspase activation. The class I PAK inhibitor IPA3 (Sigma- were visualized by DIC microscopy to calculate the percent of cells elon-
Aldrich) was used to pretreat cells at 5 m for 30 min but was not added to gated, as described above. Alternatively, cells were fixed in the gel using 2%
gels to minimize cell toxicity. In other experiments, anti1-integrin (ITGB1 paraformaldehyde and stained with Hoechst dye 33342. For abnormally
or CD29) antibodies (clone P5D2), which have been shown to block adhe- elongated cells, the nuclear length was measured as a proportion of the cells
sion to collagen (Blaschke et al., 2002; Mukhopadhyay et al., 2004), were total length. From 13 to 38 elongated cells were analyzed per patient. Cell
added to the gels at a final concentration of 20 g/ml (R&D Systems). cycle analysis performed on cells recovered from the gel according to colla-
After polymerization of gels, cells were allowed to migrate within the genase treatment showed that aphidicolin treatment decreased the percentage
gels at 37C in the presence of humidified 5% CO2. Migrating cells were of cells in S/G2/M from 17.0 10.5 and 13.2 4.0 in pretreated controls
visualized by diffusion interference contrast (DIC) microscopy using an and patients, respectively, to 1.4 0.6 and 1.0 0.5 in post-treated controls
AF6000 LX microscope (Leica) on a motorized stage, with either a 20 dry and patients, respectively (mean SD).
objective lens or a 63 glycerol immersion objective lens. Images were ac-
quired at 30-s intervals. Post-capture analysis was conducted with Imaris 7.0 Chemotaxis assays. 3D chemotaxis assays were performed as described
software (Bitplane). Length was determined by measuring the distance be- using a custom-fabricated device, with modifications (Sixt and Lmmermann,
tween cell front and uropod. Width was determined by measuring the middle 2011). Activated T cells, resuspended in 3.6 mg/ml collagen, as described
point across the cell body. Cells were defined as abnormally elongated if cell above, were loaded into the migration chamber. After polymerization of the
length was 8 the cell width at its midpoint. Abnormal elongation was re- gel matrix containing the cells, additional collagen gel, containing recombi-
ported either as percentage of time elongated or percentage of cells elongated. nant human CXCL12 (SDF-1; PeproTech) at a final concentration of
To calculate percentage of time elongated, at least 30 min of captured live 400 ng/ml, was applied as a second layer within the migration chamber.
images were analyzed. For each sample, cells were numbered and 2030 cells Cells were visualized over 30 min as they migrated toward the chemokine
were randomly chosen using an online random number generator. The gradient at 37C in the presence of humidified CO2. Time-lapse DIC im-
lengths and widths of these cells were measured in each frame. The percent- ages of migrating cells were acquired using a microscope (AF6000 LX; Leica)
age of time each individual cell spent abnormally elongated was calculated, at intervals of 30 s. Cell images were loaded into Imaris 7.0, and individual
which in turn was used to calculate the mean value for the sampled cells from cells were tracked using the Track Spots feature in the DIC channel. The
each patient. To calculate percentage of cells elongated, at least 3 h of cap- estimated diameter of the cells was 8 m, and a Quality Threshold for the
tured live images were analyzed. Four time points that divided up the total spots was typically around 100. Tracks of the individual cell spots were as-
time imaged into equal time intervals, and three z-steps associated with each time sembled using the autoregression feature. Tracks shorter than 30 time points,
point, were selected for analysis. The lengths and widths of all the cells in the or in which the cell displaced less than 5 m, were excluded. The path trav-
view at these four time points were measured. The average proportion of eled was used to calculate total displacement, displacement to the chemo-
cells that were abnormally elongated was calculated from the sampled time kine, speed, and directional velocity toward the chemokine. A total of 60326
points from each patient. Approximately 150300 cells were analyzed by this cells were analyzed to obtain means per sample. For statistical analysis, the
latter method. The percentage of cells with elongated nucleus was calculated values from DOCK8-deficient cells from patients and knockdowns were
similar to the percent of cells elongated, except that Hoechst dye 33342 was combined and compared with control cells and nonspecific siRNA knock-
used to stain nuclei and nuclei were classified as elongated as determined by downs, using the unpaired Students t test.
binary scoring of blinded samples.
Live imaging of dying cells. In initial experiments to assess cell death,
Immunofluorescence. T cells migrating within the collagen gel matrix standard collagen gel matrices were set up as described above in Collagen gel
were fixed with prewarmed 4% paraformaldehyde (Electron Microscopy Sci- migration assays for morphological analyses. DIC images were acquired at
ences) in PBS. The collagen gel containing the cells was washed in PBS, per- 2-min intervals for 21 h. PI was added during the last 4.5 h and allowed to
meabilized with 0.5% Triton X-100, blocked with 2% bovine serum albumin diffuse into the gel, during which time simultaneous DIC and fluorescence
(Sigma-Aldrich) in 0.1% Triton X-100, and incubated with primary antibod- images were acquired at 510 min intervals.
ies for 1 h at 4C. Anti-TubulinAlexa Fluor 488 (Life Technologies) was In subsequent experiments, mini-gels were used to track the fate of
used with anti-pericentrin antibody (Abcam), followed by Alexa Fluor 647 cells for 16 h. 10 l of collagen gel, which contained 104 cells, was cast into
conjugated antirabbit secondary antibody (Life Technologies). Hoechst dye a custom-partitioned segment of a high culture-insert StemCell, ibiTreat-coated,
33342 at 1 g/ml (Life Technologies) was also used. Stained gels were kept 35 mm -Dish (ibidi). After polymerization, all cells within the mini-gel
in PBS at 4C for no longer than 24 h before image capture. Imaging was were visualized at 37C in the presence of humidified 5% CO2 by DIC
performed on a Leica SP8 or SP5 white light laser confocal microscope with microscopy. A microscope (AF6000 LX) with 20 dry objective lens and
a 63 glycerol immersion objective lens, using an image stack vertical step motorized stage with Tiling function was used to scan through the entire

JEM Vol. 211, No. 13 2561


mini-gel every 2 min. Image stacks were acquired using a Roper Coolsnap Transwell assays. Polycarbonate Transwell inserts containing 5-m mem-
camera, using the following settings: 2 2 binning, 1 3 tiling, 8 m verti- brane pores were used in 24-well plates (Corning). Recombinant human
cal step, 300 m vertical depth. After data collection, tiles were assembled CXCL12 (SDF-1) at 200 ng/ml (Life Technologies) was added to the lower
using LAS EZ software (Leica) and analyzed manually using Imaris software compartment. Control and patient T cells were loaded with 0.5 M of Cell-
as described above. Cells were classified as those that had died or remained Tracker Green CMFDA or CellTracker Red CMPTX (Life Technologies),
alive at the end of the experiment. The amount of time each cell spent ab- and dye swaps were also performed. Cells were washed once with com-
normally elongated was calculated from the cell length and width measure- plete medium before placing a total of 5 104 cells in the upper compart-
ments at each time frame. Elongation episodes were also classified as those ment. After incubating at 37C, in humidified 5% CO2, for 12 h, the
immediately preceding cell fragmentation and those that did not. The dura- insert was removed, fixed in 2% paraformaldehyde in PBS for 15 min,
tion of each elongation episode was also measured. 20 cells, chosen ran- rinsed in PBS, and stained with Hoechst dye 33342. The membrane was
domly at the beginning of the experiment, were analyzed per sample. carefully cut off from the insert using a sharp scalpel and mounted onto
glass slides with coverslips using ProLong Gold mounting medium (Life
Quantitation of cell death. Cells were mixed into collagen gel matrices Technologies). Cells migrating through the membrane pores were imaged
containing varying concentrations of collagen, or in complete medium, which using a TCS SP5 laser-scanning confocal microscope with a 63 glycerol
were both supplemented with 200 U/ml of recombinant human IL-2. At immersion objective lens.
the indicated times, cells were recovered by treating samples with 1 mg/ml
of collagenase (Sigma-Aldrich) with 2.5 mM CaCl2 at 37C for 1 h. To stain Agarose gel migration assays. Tissue-culture grade agarose (Sigma-
for cell death markers, APC-conjugated Annexin V (BD) was incubated at Aldrich) was heated at a concentration of 2% in RPMI medium. A heating
4C for 15 min with the recovered cells. PI was added right before acquisi- block was used to keep the mixture warm as it was diluted with prewarmed
tion for 30 s at high speed on the FACSCanto. The numbers of Annexin V+, complete medium to a final concentration of 0.2% agarose. After equilibra-
PI+, or live (Annexin V, PI) cells were counted using FlowJo analysis soft- tion to 37C, cells were admixed. Gels were cast into Lab-Tek II 8well
ware. These numbers were used to calculate the percentage of cells that were chambered coverglasses, solidified at 4C for 10 min, and moved back to
dead or dying cells, as defined by Annexin V+ and/or PI+. 37C in the presence of humidified 5% CO2. After migration within the
agarose gel matrix, live cell imaging and analysis was performed as described
above in Collagen gel migration assays.
Scanning electron microscopy. After allowing 105 cells to migrate within
the collagen gel matrix, cells were isolated by treatment with collagenase as
2D migration assays. Lab-Tek II 8well chambered coverglasses (Nunc)
described above and washed with PBS. Recovered cells were resuspended in
were coated with 3 g/ml of recombinant human ICAM-2/Fc chimera
100 l of 0.1 M sodium cacodylate buffer and 50 l of suspensions were set-
(R&D Systems) overnight at 4C and rinsed with PBS before use. Alterna-
tled on silicon chips (Ted Pella, Inc.) for 20 min. The silicon chips were then
tively, collagen-coated slides were stored at 4C until use (BD). 200 l of
fixed and stored at 4C for up to 20 h in fixative containing 2.5% glutaralde-
cycling T cells (105/ml) were gently added into each well. After 1 h incuba-
hyde and 0.1 M sodium cacodylate buffer in PBS, pH 7.4. Samples were
tion at 37C in the presence of humidified 5% CO2, time-lapse DIC micros-
treated for 1 h with 0.5% osmium tetroxide and 0.8% potassium ferricyanide,
copy and analysis was performed as described above in Collagen gel migration
1 h with 1% tannic acid, and then overnight at 4C with 1% uranyl acetate.
assays for morphological analyses.
After a graded series of ethanol dehydration steps, the silicon chips were crit-
ical point dried (cpd 030; Bal-Tec), mounted on aluminum studs, and coated
with 70 iridium in an IBS/e sputter coater (South Bay Technologies). HSV-1 stocks and titers. To generate cell-free virus stocks of the KOS
Digital images were acquired at 5 kV using a SU-8000 field emission scan- laboratory-adapted strain of HSV-1 (gift from J. Cohen, NIAID, NIH,
ning electron microscope (Hitachi High Technologies). Bethesda, MD), 1.2 107 Vero cells grown in DMEM medium were seeded
1 d earlier to obtain a confluent monolayer. 2 104 pfu of virus was used to
infect the Vero cells. Virus was allowed to replicate while cells were cultured
Caspase-3 staining. For intracellular flow cytometric detection of acti- for 2 d at 37C, in the presence of humidified 5% CO2. To harvest the virus,
vated caspase-3, T cells were allowed to migrate within the collagen gel ma- the infected cells were collected into the medium using a cell scraper, and
trix, or in medium, for up to 20 h. Apoptosis was induced in control cells by centrifuged briefly at 4C. The pellet was frozen on dry ice and thawed for
allowing them to migrate for 6 h within the collagen gel matrix, to which three cycles. After centrifugation at 4C, the supernatant was aliquoted and
anti-FAS (clone APO-1-3) antibodies (Enzo Life Sciences) and Protein A stored at 80C until use.
(Sigma-Aldrich) were incorporated at 200 ng/ml. Cells were recovered Virus stocks were quantitated using a standard plaque assay. Vero cells
from gel or medium after collagenase digestion, as described above. Cells were seeded at 5 105 cells per well in 6-well plates, 1 d earlier. An aliquot
were then fixed and permeabilized using the Cytofix/Cytoperm Fixation/ of virus stock was thawed on ice and a 10-fold dilution series was made in
Permeabilization kit (BD). Cells were incubated with a 1:10 dilution of medium. In each well, 500 l of the diluted virus was added to a total volume
PE-conjugated antiactive-caspase-3 antibody (BD) in Perm/Wash buffer of 1 ml, and virus allowed to adsorb for 1 h. Cells were overlaid with medium
for 30 min at 4C. Stained cells were acquired on a FACSCanto, and per- containing human gamma globulin (Gamunex-C, Talecris) and cultured for
centage of positive cells was calculated using FlowJo analysis software. 2 d. The medium was removed, and cells were stained for 1 h at room tem-
perature with 1% crystal violet in 10% formaldehyde, 5% acetic acid, and 60%
Mitochondrial membrane permeability staining. Cells were loaded methanol. After washing with water and air-drying, the plaques in the cell
with 0.5 g/ml of JC-1 (Life Technologies) at 37C for 30 min and then monolayers were counted. The PFU/ml of the virus stock was calculated as:
washed with complete medium. Loaded cells were incorporated within a (number of plaques in one well) 2 (fold of dilution of virus stock used
collagen gel matrix as described above, and allowed to migrate within the gel for infection).
for 6 h. In some cases, apoptosis was induced in control cells by incubating
JC-1loaded cells with 200 ng/ml of anti-FAS (clone APO-1-3) antibodies HSV-1 infections. Mice were infected epicutaneously as previously de-
and 200 ng/ml of Protein A for 20 min, before incorporating cells within the scribed (Goel et al., 2002; van Lint et al., 2004; Gebhardt et al., 2011). Sex-
collagen gel matrix. Live cell imaging, at 37C and in the presence of 5% matched mice, at 68 wk of age, were used for infections, with four to seven
CO2, was performed using a white light laser confocal microscope (Leica), mice per treatment group in each experiment. In brief, mice were anesthe-
with excitation wavelength of 488 nm and an image stack vertical step setting tized with an intraperitoneal injection of 2.5 mg ketamine and 0.4 mg xyla-
of 0.2 m. Loss of orange-red staining of J aggregates, with continued green zine in sterile PBS, per 20 g of body weight. The fur was clipped and depilated
staining of J monomers, was indicative of mitochondrial depolarization. with Nair in the region of the dorsal left flank. The skin was then abraded

2562 DOCK8 and cytothripsis in skin viral infections | Zhang et al.


Ar ticle

using a 1.5 1.5 mm square, 100 grit sand paper (3M), with 10 steady strokes individual doseresponse curves were calculated using the nonlinear regres-
of manually applied pressure. HSV-1, at 106 pfu in a volume of 5 l, was dropped sion curve fitting option (straight-line) in Prism. The slopes for controls mi-
onto the abraded skin, and the site of infection covered for 12 h. Disease se- grating in gel and patients migrating in gel were 0.015 0.023 and 0.13
verity was scored daily according to the following criteria: 0, no symptoms; 0.054, respectively, which indicated little dose-effect correlation for cells mi-
1, several isolated blisters close to the original infection site; 2, blisters clus- grating in gel. In contrast, in controls induced to undergo apoptosis, the slope
tered along a dermatomal distribution to form a continuous line; 3, blisters was 11.0 1.4, which indicated a strong dose-effect correlation (mean
merged together with extensive necrosis surrounding the blisters; 4, mouse SD). Fig. 6 A: a linear mixed effects model was used to compare the effect
found dead or with neurological symptoms requiring euthanasia. of collagen concentration on response of cells from controls versus patients.
There was no dose effect in the control group, and no difference between
Adoptive transfers of T cells. CD8 T cells were isolated and enriched controls and patients when dose level was 0. The dose effect at concentra-
from DOCK8-deficient GFP-cpm mice by incubating splenocytes with an tions >0 was different in the controls versus the patients, as indicated by in-
antibody cocktail (Ter119, M5/114, GK1.5, RB6-8C5, M1/70, and F4/80) teraction of 4.736 (95% CI of 3.87, 5.61) with p-value <0.0001. Figs. 7 A
and performing negative enrichment with Dynabead magnetic beads (Invit- and 10 H: ordinary one-way ANOVA was performed with correction for
rogen). These T cells were adoptively transferred into C57BL/6 mice (3 multiple comparisons, using Prism. P-values are indicated in the legends.
106 cells) along with 3 103 CD8 T cells from gBT-I.DsRed TCR trans- Fig. 9 B: The Wilcoxon matched-pairs signed rank test (two-tailed) was
genic mice, before skin HSV infection. Naive gBT-I.DsRed or gBT-I.GFP. performed using Prism to compare repeated measurements of clinical scores
cpm T cells isolated from LNs were adoptively transferred via the tail vein. associated with individual Dock8-deficient (KO) versus WT mice. The
A total of 5 104 cells were transferred in cotransfer experiments before HSV p-value was 0.001. Fig. 9 C: the log-rank (Mantel-Cox) test for the Kaplan-
infection. In vitrogenerated gBT-I and gBT-I.GFP.cpm effector spleno- Meier survival curve was performed using Prism. *, P = 0.016.
cytes were activated by peptide-pulsed splenocytes as previously described
(Mackay et al., 2012). Such cells were comparably activated by this regi- Online supplemental material. Videos 1 and 2 show T cell and NK cell
men, as reflected by their CD44hi and CD62Llo surface marker expression, elongation during migration within collagen gel matrices. Video 3 shows
consistent with previous reports that Dock8 mutant cells activate and pro- cytothripsis. Videos 4 and 5 show intravital two-photon microscopy during
liferate normally after TCR stimulation (Lambe et al., 2011). Activated viral skin infections. Online supplemental material is available at http://
CD44hi T cells (106) were transferred into recipients by intradermal injec- www.jem.org/cgi/content/full/jem.20141307/DC1.
tion (five 20-l injections over an area of skin 1 1.5 cm2) with a 30-gauge
needle. For DNFB (Sigma-Aldrich) treatment, mice were shaved and depil- We thank the following people: for pictures of patient skin and skin histology,
ated before the application of 15 l of 0.5% DNFB in acetone/oil (4:1) to Maria Turner and Stefania Pittaluga; for technical assistance with microscopy
a 1-cm2 area of skin. 30 d after DNFB treatment, mice were infected on the studies, Juraj Kabat and Owen Schwartz; for electron microscopy, Beth Fischer; for
skin with HSV. statistical analyses, Jin Qing; for mouse breeding and screening, Gayle Davey and
Melanie Damtsis; for reagents, Fabio Candotti, Jeffrey Cohen, Carole Yee, and Lixin
Intravital two-photon microscopy. Mice were infected epicutaneously Zheng; for clinical support, Thomas Dimaggio and Angela Wang; for helpful advice
with HSV-1 and the skin imaged by two-photon microscopy as previously and/or critical reading of the manuscript, Tim Lmmermann, Michael Lenardo,
described (Gebhardt et al., 2011). For intravital two-photon microscopy, mice Pamela Schwartzberg, Andrew Snow, Li Yu, and Yu Zhang. We also thank the
were anaesthetized, depilated, and two parallel incisions were made 15 mm patients and their families for participating in this study, especially Kelsey Koch, in
apart along the flank. The skin was separated from the peritoneum, and ad- whose memory we dedicate this work.
This work was supported by the Intramural Research Program of the NIAID,
hered to an 18-mm-wide piece of 1-mm stainless steel inserted to form a
NIH, the Australian Research Council (S.N. Mueller), the Medical Research Council
stable raised platform, attached to a custom-made imaging platform main-
UK (R.J. Cornall), and the Oxford NIHR Biomedical Research Centre (R.J. Cornall).
tained at 35C. Images were acquired with an upright LSM710 NLO multi-
R.A. Grodick was an HHMI-NIH Research Scholar.
photon microscope (Carl Zeiss) with a 20 1.0 NA water immersion The authors have no conflicting financial interests.
objective. 3D stacks were captured every 1 min for 3060 min. Raw imag-
ing data were processed with Imaris 7 (Biplane). Cell migration through Author contributions: C.G. Dove and Q. Zhang discovered and characterized the
combined epidermal and dermal layers was analyzed through automatic cell 3D migration defect in collagen gel matrices. D.M. Strauss-Albee performed
tracking aided by manual corrections. Only tracks that lasted longer than foreskin migration experiments. Q. Zhang, D.M. Strauss-Albee, and C.G. Dove
5 min were analyzed. For the generation of movies, image sequences ex- discovered and characterized the nuclear deformation abnormality. R.A. Grodick
ported from Imaris were composed in After Effects (CS5; Adobe). performed cell cycle blockade and agarose gel experiments. C.G. Dove and
T.E. Lenardo performed chemotaxis experiments. Q. Zhang, J.A. Garcia, and
Statistical analyses. Figs. 2 (CI), 4 (D and E), 5 (C and F, right), 6 (C and D), C.G. Dove discovered and characterized cytothripsis. Q. Zhang and C.G. Dove
7 (D and E), 9 E, and 10 (F and G): the unpaired two-tailed Students t test performed immunofluorescence experiments. Q. Zhang performed Transwell
was performed using Prism 6.0 software (GraphPad). Fig. 9 E: 17 movies and 2D migration and integrin blockade/knockdown experiments. H.M. Murdock,
from six mice pooled from two experiments were analyzed. In each movie, Q. Zhang, J.A. Garcia, and D.B. Chandler-Brown performed CDC42, RAC, and
all the cells in the frame, numbering at least several dozen, were counted and PAK knockdown and in-gel immunoblotting experiments. Q. Zhang and J.A.
the proportion elongated calculated. In the remaining experiments where Garcia performed WAS knockdown experiments. H.M. Murdock performed
small molecule cytoskeletal inhibitor experiments. S.N. Mueller, J.L. Hor,
individual cells were analyzed, the mean value for the population from each
G. Crawford, and R.J. Cornall generated mouse strains. Q. Zhang and J.N. Mandl
sample tested was used to represent that individual. P-values are indicated
evaluated disease, and J.L. Hor and S.N. Mueller performed intravital two-
in the legends. Fig. 3 (AD): the mean values for controls and control NS
photon imaging and TRM studies in HSV-infected mice. A.F. Freeman and H.C. Su
siRNA were included in the DOCK8-replete group, and the mean values diagnosed patients and provided clinical information and samples. H.F. Matthews
for patients and knockdowns were included in the DOCK8-deficient group. coordinated clinical study protocol and sample collection. H. Jing and Q. Zhang
For each parameter, the two groups were compared using the unpaired two- assisted with identifying patients and processing of samples. H.C. Su and S.N.
tailed Students t test. ns, not significant (P 0.4). Figs. 4 C, 5 F (left), 6 E, Mueller planned and supervised the experimental work and data analyses, and
7 (B, F, and H), and 10 (AC and E): ordinary two-way ANOVA was per- R.N. Germain provided advice. Q. Zhang and H.C. Su prepared the manuscript. All
formed with correction for multiple comparisons using Prism. P-values are authors discussed and revised the manuscript.
indicated in the legends. For Fig. 6 E comparisons, p-values were nonsignifi-
cant (P > 0.99). Fig. 5 C: for each individual tested, the time was plotted Submitted: 11 July 2014
against percentage of active caspase-3positive cells. The slopes of the Accepted: 29 October 2014

JEM Vol. 211, No. 13 2563


REFERENCES syndrome. J. Allergy Clin. Immunol. 124:12891302: e4. http://dx.doi
Akbar, H., X. Shang, R. Perveen, M. Berryman, K. Funk, J.F. Johnson, N.N. .org/10.1016/j.jaci.2009.10.038
Tandon, and Y. Zheng. 2011. Gene targeting implicates Cdc42 GTPase Friedl, P., K. Wolf, and J. Lammerding. 2011. Nuclear mechanics dur-
in GPVI and non-GPVI mediated platelet filopodia formation, secretion ing cell migration. Curr. Opin. Cell Biol. 23:5564. http://dx.doi.org/
and aggregation. PLoS ONE. 6:e22117. http://dx.doi.org/10.1371/ 10.1016/j.ceb.2010.10.015
journal.pone.0022117 Gebhardt, T., L.M. Wakim, L. Eidsmo, P.C. Reading, W.R. Heath, and
Ariotti, S., J.B. Beltman, G. Chodaczek, M.E. Hoekstra, A.E. van Beek, R. F.R. Carbone. 2009. Memory T cells in nonlymphoid tissue that pro-
Gomez-Eerland, L. Ritsma, J. van Rheenen, A.F. Mare, T. Zal, et al. vide enhanced local immunity during infection with herpes simplex
2012. Tissue-resident memory CD8+ T cells continuously patrol skin virus. Nat. Immunol. 10:524530. http://dx.doi.org/10.1038/ni.1718
epithelia to quickly recognize local antigen. Proc. Natl. Acad. Sci. USA. Gebhardt, T., P.G. Whitney, A. Zaid, L.K. Mackay, A.G. Brooks, W.R.
109:1973919744. http://dx.doi.org/10.1073/pnas.1208927109 Heath, F.R. Carbone, and S.N. Mueller. 2011. Different patterns of
Biron, C.A., K.B. Nguyen, G.C. Pien, L.P. Cousens, and T.P. Salazar-Mather. peripheral migration by memory CD4+ and CD8+ T cells. Nature. 477:
1999. Natural killer cells in antiviral defense: function and regulation 216219. http://dx.doi.org/10.1038/nature10339
by innate cytokines. Annu. Rev. Immunol. 17:189220. http://dx.doi Goel, N., J.J. Docherty, M.M. Fu, D.H. Zimmerman, and K.S. Rosenthal.
.org/10.1146/annurev.immunol.17.1.189 2002. A modification of the epidermal scarification model of herpes sim-
Blaschke, F., P. Stawowy, S. Goetze, O. Hintz, M. Grfe, U. Kintscher, E. plex virus infection to achieve a reproducible and uniform progression
Fleck, and K. Graf. 2002. Hypoxia activates 1-integrin via ERK 1/2 and of disease. J. Virol. Methods. 106:153158. http://dx.doi.org/10.1016/
p38 MAP kinase in human vascular smooth muscle cells. Biochem. Biophys. S0166-0934(02)00160-X
Res. Commun. 296:890896. http://dx.doi.org/10.1016/S0006-291X Gotoh, K., Y. Tanaka, A. Nishikimi, A. Inayoshi, M. Enjoji, R. Takayanagi, T.
(02)02033-8 Sasazuki, and Y. Fukui. 2008. Differential requirement for DOCK2 in mi-
Bokoch, G.M. 2003. Biology of the p21-activated kinases. Annu. Rev. Biochem. gration of plasmacytoid dendritic cells versus myeloid dendritic cells. Blood.
72:743781.http://dx.doi.org/10.1146/annurev.biochem.72.121801 111:29732976. http://dx.doi.org/10.1182/blood-2007-09-112169
.161742 Grgoire, C., L. Chasson, C. Luci, E. Tomasello, F. Geissmann, E. Vivier,
Chu, E.Y., A.F. Freeman, H. Jing, E.W. Cowen, J. Davis, H.C. Su, S.M. Holland, and T. Walzer. 2007. The trafficking of natural killer cells. Immunol. Rev.
and M.L. Turner. 2012. Cutaneous manifestations of DOCK8 defi- 220:169182. http://dx.doi.org/10.1111/j.1600-065X.2007.00563.x
ciency syndrome. Arch. Dermatol. 148:7984. http://dx.doi.org/10.1001/ Guo, F., D. Hildeman, P. Tripathi, C.S. Velu, H.L. Grimes, and Y. Zheng.
archdermatol.2011.262 2010. Coordination of IL-7 receptor and T-cell receptor signaling by
Ct, J.F., and K. Vuori. 2002. Identification of an evolutionarily con- cell-division cycle 42 in T-cell homeostasis. Proc. Natl. Acad. Sci. USA.
served superfamily of DOCK180-related proteins with guanine nucleo- 107:1850518510. http://dx.doi.org/10.1073/pnas.1010249107
tide exchange activity. J. Cell Sci. 115:49014913. http://dx.doi.org/ Ham, H., S. Guerrier, J. Kim, R.A. Schoon, E.L. Anderson, M.J. Hamann,
10.1242/jcs.00219 Z. Lou, and D.D. Billadeau. 2013. Dedicator of cytokinesis 8 interacts
Ct, J.F., and K. Vuori. 2007. GEF what? Dock180 and related proteins with talin and Wiskott-Aldrich syndrome protein to regulate NK cell
help Rac to polarize cells in new ways. Trends Cell Biol. 17:383393. cytotoxicity. J. Immunol. 190:36613669. http://dx.doi.org/10.4049/
http://dx.doi.org/10.1016/j.tcb.2007.05.001 jimmunol.1202792
Crawford, G., A. Enders, U. Gileadi, S. Stankovic, Q. Zhang, T. Lambe, T.L. Harada, Y., Y. Tanaka, M. Terasawa, M. Pieczyk, K. Habiro, T. Katakai,
Crockford, H.E. Lockstone, A. Freeman, P.D. Arkwright, et al. 2013. K. Hanawa-Suetsugu, M. Kukimoto-Niino, T. Nishizaki, M. Shirouzu,
DOCK8 is critical for the survival and function of NKT cells. Blood. et al. 2012. DOCK8 is a Cdc42 activator critical for interstitial dendritic
122:20522061. http://dx.doi.org/10.1182/blood-2013-02-482331 cell migration during immune responses. Blood. 119:44514461. http://
Cunningham, A.L., R.R. Turner, A.C. Miller, M.F. Para, and T.C. Merigan. dx.doi.org/10.1182/blood-2012-01-407098
1985. Evolution of recurrent herpes simplex lesions. An immunohisto- Harpaz, R., I.R. Ortega-Sanchez, and J.F. Seward. Advisory Committee
logic study. J. Clin. Invest. 75:226233. http://dx.doi.org/10.1172/ on Immunization Practices (ACIP) Centers for Disease Control and Pre
JCI111678 vention (CDC). 2008. Prevention of herpes zoster: recommendations of
Darzynkiewicz, Z., and X. Huang. 2004. Analysis of cellular DNA content the Advisory Committee on Immunization Practices (ACIP). MMWR
by flow cytometry. Curr. Protoc. Immunol. Chapter 5:7. Recomm. Rep. 57:130, quiz :CE2CE4.
De Meester, J., R. Calvez, S. Valitutti, and L. Dupr. 2010. The Wiskott- Higgins, C.R., J.K. Schofield, F.M. Tatnall, and I.M. Leigh. 1993. Natural
Aldrich syndrome protein regulates CTL cytotoxicity and is required history, management and complications of herpes labialis. J. Med. Virol.
for efficient killing of B cell lymphoma targets. J. Leukoc. Biol. 88:1031 41:2226. http://dx.doi.org/10.1002/jmv.1890410506
1040. http://dx.doi.org/10.1189/jlb.0410197 Honda, T., J.G. Egen, T. Lmmermann, W. Kastenmller, P. Torabi-Parizi,
Dotta, L., L. Tassone, and R. Badolato. 2011. Clinical and genetic fea- and R.N. Germain. 2014. Tuning of antigen sensitivity by T cell receptor-
tures of Warts, Hypogammaglobulinemia, Infections and Myelokathexis dependent negative feedback controls T cell effector function in inflamed
(WHIM) syndrome. Curr. Mol. Med. 11:317325. http://dx.doi.org/ tissues. Immunity. 40:235247. http://dx.doi.org/10.1016/j.immuni
10.2174/156652411795677963 .2013.11.017
Drijkoningen, M., C. De Wolf-Peeters, H. Degreef, and V. Desmet. 1988. Imai, K., T. Morio, Y. Zhu, Y. Jin, S. Itoh, M. Kajiwara, J. Yata, S. Mizutani,
Epidermal Langerhans cells, dermal dendritic cells, and keratinocytes in H.D. Ochs, and S. Nonoyama. 2004. Clinical course of patients with
viral lesions of skin and mucous membranes: an immunohistochemical WASP gene mutations. Blood. 103:456464. http://dx.doi.org/10.1182/
study. Arch. Dermatol. Res. 280:220227. http://dx.doi.org/10.1007/ blood-2003-05-1480
BF00513961 Jabara, H.H., D.R. McDonald, E. Janssen, M.J. Massaad, N. Ramesh, A.
Dustin, M.L. 2008. Hunter to gatherer and back: immunological synapses Borzutzky, I. Rauter, H. Benson, L. Schneider, S. Baxi, et al. 2012.
and kinapses as variations on the theme of amoeboid locomotion. Annu. DOCK8 functions as an adaptor that links TLR-MyD88 signaling to
Rev. Cell Dev. Biol. 24:577596. http://dx.doi.org/10.1146/annurev B cell activation. Nat. Immunol. 13:612620. http://dx.doi.org/10.1038/
.cellbio.24.110707.175226 ni.2305
Ebert, L.M., S. Meuter, and B. Moser. 2006. Homing and function of human Jaffe, A.B., and A. Hall. 2005. Rho GTPases: biochemistry and biology. Annu.
skin T cells and NK cells: relevance for tumor surveillance. J. Immunol. Rev. Cell Dev. Biol. 21:247269. http://dx.doi.org/10.1146/annurev
176:43314336. http://dx.doi.org/10.4049/jimmunol.176.7.4331 .cellbio.21.020604.150721
Engelhardt, K.R., S. McGhee, S. Winkler, A. Sassi, C. Woellner, G. Jiang, X., R.A. Clark, L. Liu, A.J. Wagers, R.C. Fuhlbrigge, and T.S.
Lopez-Herrera, A. Chen, H.S. Kim, M.G. Lloret, I. Schulze, et al. Kupper. 2012. Skin infection generates non-migratory memory CD8+
2009. Large deletions and point mutations involving the dedicator of T(RM) cells providing global skin immunity. Nature. 483:227231.
cytokinesis 8 (DOCK8) in the autosomal-recessive form of hyper-IgE http://dx.doi.org/10.1038/nature10851

2564 DOCK8 and cytothripsis in skin viral infections | Zhang et al.


Ar ticle

Jing, H., Q. Zhang, Y. Zhang, B.J. Hill, C.G. Dove, E.W. Gelfand, T.P. herpes simplex virus. Immunol. Cell Biol. 80:156163. http://dx.doi
Atkinson, G. Uzel, H.F. Matthews, P.J. Mustillo, et al. 2014. Somatic .org/10.1046/j.1440-1711.2002.01071.x
reversion in dedicator of cytokinesis 8 immunodeficiency modulates Mueller, S.N., T. Gebhardt, F.R. Carbone, and W.R. Heath. 2013. Memory
disease phenotype. J. Allergy Clin. Immunol. 133:16671675. http://dx T cell subsets, migration patterns, and tissue residence. Annu. Rev.
.doi.org/10.1016/j.jaci.2014.03.025 Immunol. 31:137161. http://dx.doi.org/10.1146/annurev-immunol-
Kikuchi, T., S. Kubonishi, M. Shibakura, N. Namba, T. Matsui, Y. Fukui, 032712-095954
M. Tanimoto, and Y. Katayama. 2008. Dock2 participates in bone mar- Mukhopadhyay, N.K., D. Gilchrist, G.J. Gordon, C.J. Chen, R. Bueno,
row lympho-hematopoiesis. Biochem. Biophys. Res. Commun. 367:9096. M.L. Lu, R. Salgia, D.J. Sugarbaker, and M.T. Jaklitsch. 2004. Integrin-
http://dx.doi.org/10.1016/j.bbrc.2007.12.093 dependent protein tyrosine phosphorylation is a key regulatory event in
Kilkenny, M., and R. Marks. 1996. The descriptive epidemiology of warts in the collagen-IV-mediated adhesion and proliferation of human lung tumor
community. Australas. J. Dermatol. 37:8086. http://dx.doi.org/10.1111/ cell line, Calu-1. Ann. Thorac. Surg. 78:450457. http://dx.doi.org/10
j.1440-0960.1996.tb01010.x .1016/j.athoracsur.2004.01.042
Kittler, R., L. Pelletier, A.K. Heninger, M. Slabicki, M. Theis, L. Miroslaw, Muraki, R.,T. Baba,T. Iwasaki,T. Sata, and T. Kurata. 1992. Immunohistochemical
I. Poser, S. Lawo, H. Grabner, K. Kozak, et al. 2007. Genome-scale study of skin lesions in herpes zoster. Virchows Arch. A Pathol. Anat.
RNAi profiling of cell division in human tissue culture cells. Nat. Cell Histopathol. 420:7176. http://dx.doi.org/10.1007/BF01605987
Biol. 9:14011412. http://dx.doi.org/10.1038/ncb1659 Neuman, R.E., and M.A. Logan. 1950. The determination of collagen and
Kunisaki, Y., A. Nishikimi, Y. Tanaka, R. Takii, M. Noda, A. Inayoshi, K. elastin in tissues. J. Biol. Chem. 186:549556.
Watanabe, F. Sanematsu, T. Sasazuki, T. Sasaki, and Y. Fukui. 2006. Nikkels, A.F., P. Delvenne, C. Sadzot-Delvaux, S. Debrus, J. Piette, B. Rentier,
DOCK2 is a Rac activator that regulates motility and polarity during G. Lipcsei, P. Quatresooz, and G.E. Pirard. 1996. Distribution of varicella
neutrophil chemotaxis. J. Cell Biol. 174:647652. http://dx.doi.org/ zoster virus and herpes simplex virus in disseminated fatal infections.
10.1083/jcb.200602142 J. Clin. Pathol. 49:243248. http://dx.doi.org/10.1136/jcp.49.3.243
Lambe, T., G. Crawford, A.L. Johnson, T.L. Crockford, T. Bouriez-Jones, Nishikimi, A., H. Fukuhara, W. Su, T. Hongu, S. Takasuga, H. Mihara,
A.M. Smyth, T.H. Pham, Q. Zhang, A.F. Freeman, J.G. Cyster, et al. Q. Cao, F. Sanematsu, M. Kanai, H. Hasegawa, et al. 2009. Sequential
2011. DOCK8 is essential for T-cell survival and the maintenance of regulation of DOCK2 dynamics by two phospholipids during neu-
CD8+ T-cell memory. Eur. J. Immunol. 41:34233435. http://dx.doi.org/ trophil chemotaxis. Science. 324:384387. http://dx.doi.org/10.1126/
10.1002/eji.201141759 science.1170179
Lmmermann, T., B.L. Bader, S.J. Monkley, T. Worbs, R. Wedlich- Nishikimi, A., M. Kukimoto-Niino, S. Yokoyama, and Y. Fukui. 2013.
Sldner, K. Hirsch, M. Keller, R. Frster, D.R. Critchley, R. Fssler, Immune regulatory functions of DOCK family proteins in health and dis-
and M. Sixt. 2008. Rapid leukocyte migration by integrin-independent ease. Exp. Cell Res. 319:23432349. http://dx.doi.org/10.1016/j.yexcr
flowing and squeezing. Nature. 453:5155. http://dx.doi.org/10.1038/ .2013.07.024
nature06887 Pedersen, J.A., and M.A. Swartz. 2005. Mechanobiology in the third dimen
Lang, P.A., N. Shaabani, S. Borkens, N. Honke, S. Scheu, S. Booth, D. sion. Ann. Biomed. Eng. 33:14691490. http://dx.doi.org/10.1007/
Brenner, A. Meryk, C. Barthuber, M. Recher, et al. 2013. Reduced s10439-005-8159-4
type I interferon production by dendritic cells and weakened antiviral Randall, K.L., T. Lambe, A.L. Johnson, B. Treanor, E. Kucharska, H.
immunity in patients with Wiskott-Aldrich syndrome protein deficiency. Domaschenz, B. Whittle, L.E. Tze, A. Enders, T.L. Crockford, et al.
J. Allergy Clin. Immunol. 131:815824: 824.e2. http://dx.doi.org/10.1016/ 2009. Dock8 mutations cripple B cell immunological synapses, germinal
j.jaci.2012.08.050 centers and long-lived antibody production. Nat. Immunol. 10:1283
Li, R., and G.G. Gundersen. 2008. Beyond polymer polarity: how the cy- 1291. http://dx.doi.org/10.1038/ni.1820
toskeleton builds a polarized cell. Nat. Rev. Mol. Cell Biol. 9:860873. Randall, K.L., S.S. Chan, C.S. Ma, I. Fung, Y. Mei, M. Yabas, A. Tan, P.D.
http://dx.doi.org/10.1038/nrm2522 Arkwright, W. Al Suwairi, S.O. Lugo Reyes, et al. 2011. DOCK8 defi-
Lowry, O.H., D.R. Gilligan, and E.M. Katersky. 1941. The determination ciency impairs CD8 T cell survival and function in humans and mice. J.
of collagen and elastin in tissues, with results obtained in various normal Exp. Med. 208:23052320. http://dx.doi.org/10.1084/jem.20110345
tissues from different species. J. Biol. Chem. 139:795804. Ruusala, A., and P. Aspenstrm. 2004. Isolation and characterisation of DOCK8,
Mackay, L.K., A.T. Stock, J.Z. Ma, C.M. Jones, S.J. Kent, S.N. Mueller, a member of the DOCK180-related regulators of cell morphology. FEBS
W.R. Heath, F.R. Carbone, and T. Gebhardt. 2012. Long-lived epi- Lett. 572:159166. http://dx.doi.org/10.1016/j.febslet.2004.06.095
thelial immunity by tissue-resident memory T (TRM) cells in the ab- Sabry, A., P.J. Hauk, H. Jing, H.C. Su, N.V. Stence, D.M. Mirsky, M.A. Nagel,
sence of persisting local antigen presentation. Proc. Natl. Acad. Sci. USA. J.K. Abbott, L.L. Dragone, J. Armstrong-Wells, et al. 2014. Vaccine
109:70377042. http://dx.doi.org/10.1073/pnas.1202288109 strain varicella-zoster virus-induced central nervous system vasculopathy
Mackay, L.K., A. Rahimpour, J.Z. Ma, N. Collins, A.T. Stock, M.L. Hafon, as the presenting feature of DOCK8 deficiency. J. Allergy Clin. Immunol.
J. Vega-Ramos, P. Lauzurica, S.N. Mueller, T. Stefanovic, et al. 2013. 133:12251227. http://dx.doi.org/10.1016/j.jaci.2013.11.031
The developmental pathway for CD103+CD8+ tissue-resident mem- Sanal, O., H. Jing, T. Ozgur, D. Ayvaz, D.M. Strauss-Albee, S. Ersoy-Evans, I.
ory T cells of skin. Nat. Immunol. 14:12941301. http://dx.doi.org/ Tezcan, G. Turkkani, H.F. Matthews, G. Haliloglu, et al. 2012. Additional
10.1038/ni.2744 diverse findings expand the clinical presentation of DOCK8 deficiency.
Meller, N., S. Merlot, and C. Guda. 2005. CZH proteins: a new family J. Clin. Immunol. 32:698708. http://dx.doi.org/10.1007/s10875-
of Rho-GEFs. J. Cell Sci. 118:49374946. http://dx.doi.org/10.1242/ 012-9664-5
jcs.02671 Schaefer, B.C., M.L. Schaefer, J.W. Kappler, P. Marrack, and R.M. Kedl.
Mizesko, M.C., P.P. Banerjee, L. Monaco-Shawver, E.M. Mace, W.E. Bernal, 2001. Observation of antigen-dependent CD8+ T-cell/dendritic cell
J. Sawalle-Belohradsky, B.H. Belohradsky, V. Heinz, A.F. Freeman, interactions in vivo. Cell. Immunol. 214:110122. http://dx.doi.org/10
K.E. Sullivan, et al. 2013. Defective actin accumulation impairs human .1006/cimm.2001.1895
natural killer cell function in patients with dedicator of cytokinesis 8 defi- Simmons, A., and A.A. Nash. 1984. Zosteriform spread of herpes simplex
ciency. J. Allergy Clin. Immunol. 131:840848. http://dx.doi.org/10.1016/ virus as a model of recrudescence and its use to investigate the role of
j.jaci.2012.12.1568 immune cells in prevention of recurrent disease. J. Virol. 52:816821.
Mou, F., M. Praskova, F. Xia, D. Van Buren, H. Hock, J. Avruch, and Sinha, S., and W. Yang. 2008. Cellular signaling for activation of Rho
D. Zhou. 2012. The Mst1 and Mst2 kinases control activation of rho GTPase Cdc42. Cell. Signal. 20:19271934. http://dx.doi.org/10.1016/
family GTPases and thymic egress of mature thymocytes. J. Exp. Med. j.cellsig.2008.05.002
209:741759. http://dx.doi.org/10.1084/jem.20111692 Sixt, M., and T. Lmmermann. 2011. In vitro analysis of chemotactic leu-
Mueller, S.N., W. Heath, J.D. McLain, F.R. Carbone, and C.M. Jones. kocyte migration in 3D environments. Methods Mol. Biol. 769:149165.
2002. Characterization of two TCR transgenic mouse lines specific for http://dx.doi.org/10.1007/978-1-61779-207-6_11

JEM Vol. 211, No. 13 2565


Snapper, S.B., P. Meelu, D. Nguyen, B.M. Stockton, P. Bozza, F.W. Alt, F.S. in mouse ES cells, embryos, and adult animals. Genesis. 40:241246.
Rosen, U.H. von Andrian, and C. Klein. 2005. WASP deficiency leads http://dx.doi.org/10.1002/gene.20095
to global defects of directed leukocyte migration in vitro and in vivo. J. Wolf, K., S. Alexander, V. Schacht, L.M. Coussens, U.H. von Andrian, J.
Leukoc. Biol. 77:993998. http://dx.doi.org/10.1189/jlb.0804444 van Rheenen, E. Deryugina, and P. Friedl. 2009. Collagen-based cell
Spinner, M.A., L.A. Sanchez,A.P. Hsu, P.A. Shaw, C.S. Zerbe, K.R. Calvo, D.C. migration models in vitro and in vivo. Semin. Cell Dev. Biol. 20:931
Arthur,W. Gu, C.M. Gould, C.C. Brewer, et al. 2014. GATA2 deficiency: 941. http://dx.doi.org/10.1016/j.semcdb.2009.08.005
a protean disorder of hematopoiesis, lymphatics, and immunity. Blood. Zaid, A., L.K. Mackay, A. Rahimpour, A. Braun, M. Veldhoen, F.R. Carbone,
123:809821. http://dx.doi.org/10.1182/blood-2013-07-515528 J.H. Manton, W.R. Heath, and S.N. Mueller. 2014. Persistence of skin-
Stock, A.T., J.M. Smith, and F.R. Carbone. 2014. Type I IFN suppresses Cxcr2 resident memory T cells within an epidermal niche. Proc. Natl. Acad. Sci.
driven neutrophil recruitment into the sensory ganglia during viral infection. USA. 111:53075312. http://dx.doi.org/10.1073/pnas.1322292111
J. Exp. Med. 211:751759. http://dx.doi.org/10.1084/jem.20132183 Zhang, J., A. Shehabeldin, L.A. da Cruz, J. Butler, A.K. Somani, M. McGavin,
Su, H.C., H. Jing, and Q. Zhang. 2011. DOCK8 deficiency. Ann. N.Y. Acad. I. Kozieradzki, A.O. dos Santos, A. Nagy, S. Grinstein, et al. 1999. Antigen
Sci. 1246:2633. http://dx.doi.org/10.1111/j.1749-6632.2011.06295.x receptor-induced activation and cytoskeletal rearrangement are impaired
Sullivan, K.E., C.A. Mullen, R.M. Blaese, and J.A. Winkelstein. 1994. A in Wiskott-Aldrich syndrome protein-deficient lymphocytes. J. Exp.
multiinstitutional survey of the Wiskott-Aldrich syndrome. J. Pediatr. Med. 190:13291342. http://dx.doi.org/10.1084/jem.190.9.1329
125:876885. http://dx.doi.org/10.1016/S0022-3476(05)82002-5 Zhang, Q., J.C. Davis, I.T. Lamborn, A.F. Freeman, H. Jing, A.J. Favreau,
van Lint, A., M. Ayers, A.G. Brooks, R.M. Coles, W.R. Heath, and F.R. H.F. Matthews, J. Davis, M.L. Turner, G. Uzel, et al. 2009. Combined
Carbone. 2004. Herpes simplex virus-specific CD8+ T cells can clear es- immunodeficiency associated with DOCK8 mutations. N. Engl. J. Med.
tablished lytic infections from skin and nerves and can partially limit the 361:20462055. http://dx.doi.org/10.1056/NEJMoa0905506
early spread of virus after cutaneous inoculation. J. Immunol. 172:392 Zhu, J., T. Peng, C. Johnston, K. Phasouk, A.S. Kask, A. Klock, L. Jin,
397. http://dx.doi.org/10.4049/jimmunol.172.1.392 K. Diem, D.M. Koelle, A. Wald, et al. 2013. Immune surveillance by
Vintersten, K., C. Monetti, M. Gertsenstein, P. Zhang, L. Laszlo, S. Biechele, CD8+ skin-resident T cells in human herpes virus infection. Nature.
and A. Nagy. 2004. Mouse in red: red fluorescent protein expression 497:494497. http://dx.doi.org/10.1038/nature12110

2566 DOCK8 and cytothripsis in skin viral infections | Zhang et al.


INSIGHTS

FLUshing in the bathroom


Seasonal influenza represents a contagious family of respiratory viruses that infect
530% of the global population yearly and account for as many as 500,000 deaths
annually. Flu infection is characterized by symptoms such as fever, cough, sore throat,
runny nose, body aches, and fatigue. Interestingly, some people also develop diarrhea,
even though the virus tropism is for respiratory tissue. Despite being a well-studied
viral infection, the underlying mechanisms involved in the development of gastro-
enteritis-like syndrome with flu infection are poorly understood.
Now, Wang et al. have used a mouse model to show that influenza infection Insight from (left to right) Carolina Amezcua,
not only causes lung inflammation but also causes intestinal inflammation, even Nicola Gagliani, and Richard Flavell
though influenza virus was not detectable in the gastrointestinal tract. They showed
that during intranasal flu infection, CCR9+CD4+ T cells migrate from the lung into the intestinal mucosa in a CCL25/CCR9-
dependent manner and alter the composition of the gut microbiota by secreting IFN-g. Homeostasis of the intestinal microbiota
was altered, and increased numbers of Escherichia coli were detected after flu infection. Antibiotic treatment of intranasal flu-infected
mice protected them against infection-induced diarrhea. These data suggest that the dysbiosis generated after flu infection leads
to intestinal injury. The changes generated in the gut microbiota induced the production of IL-15 by intestinal epithelial
cells. IL-15 induced the expansion of Th17 cells in the small intestine, which then mediated intestinal immune injury.
It is noteworthy that not all flu-infected patients develop gastroenteritis-like symptoms. Why is this? One possibility could be
that the migration of pathogenic cells into the intestine depends on the severity of the infection. Consequently only highly infected
patients may get diarrhea. Alternatively, the specific microbial landscape in some individuals and the existence of regulatory bacteria,
could obstruct the growth of E. coli or promote regu-
latory T cells which in turn can control intestinal
inflammation.
The data presented in this paper corroborate
the hypothesis that the intestine is a suitable place
to defuse an immune response. The abundance of
antiinflammatory cytokines, such as IL-10 and

ADAPTED FROM AN ILLUSTRATION BY THE AUTHORS


TGF-b, regulatory cells and continuous regenera-
tion of the tissue predispose the intestine with an
ability to control effector cells. Moreover, if effector
T cells escape all possible regulatory mechanisms,
diverting them to the lumenin essence flushing
them awaycould still be less dangerous than
allowing them to remain in situ and risking lung
tissue damage.
Wang, J., et al. 2014. J. Exp. Med. http://dx.doi.org/10.1084/ Working model of intestinal injury induced by respiratory influenza virus infection
jem.20140625.

Carolina Amezcua, Nicola Gagliani, and Richard Flavell; Howard Hughes Medical Institute, Yale School of Medicine: richard.flavell@yale.edu, caro.amezcua@yale.edu,
and nicola.gagliani@yale.edu


Article

Respiratory influenza virus infection induces


intestinal immune injury via microbiota-
mediated Th17 celldependent inflammation
Jian Wang,1* Fengqi Li,1* Haiming Wei,1,2 Zhe-Xiong Lian,1,2 Rui Sun,1,2
and Zhigang Tian1,2,3
1Instituteof Immunology and CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences
and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China
2Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
3Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis

and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang
310003, China

Influenza in humans is often accompanied by gastroenteritis-like symptoms such as diar-


rhea, but the underlying mechanism is not yet understood. We explored the occurrence of
gastroenteritis-like symptoms using a mouse model of respiratory influenza infection. We
found that respiratory influenza infection caused intestinal injury when lung injury oc-
curred, which was not due to direct intestinal viral infection. Influenza infection altered the
intestinal microbiota composition, which was mediated by IFN- produced by lung-derived
CCR9+CD4+ T cells recruited into the small intestine. Th17 cells markedly increased in the
small intestine after PR8 infection, and neutralizing IL-17A reduced intestinal injury.
Moreover, antibiotic depletion of intestinal microbiota reduced IL-17A production and
attenuated influenza-caused intestinal injury. Further study showed that the alteration of
intestinal microbiota significantly stimulated IL-15 production from intestinal epithelial
cells, which subsequently promoted Th17 cell polarization in the small intestine in situ.
Thus, our findings provide new insights into an undescribed mechanism by which respiratory
influenza infection causes intestinal disease.

CORRESPONDENCE Influenza is an infectious respiratory disease af- 2001; Chervonsky, 2009). Distinct components
Zhigang Tian: fecting many bird and mammal species (Laver of commensal bacteria were associated with spe-
tzg@ustc.edu.cn
and Webster, 1979; Reid et al., 1999). Clinically, cial status of the immune system. Although most
Abbreviations used: BALF, the most common symptoms include cough, commensal bacteria are beneficial (Ichinohe et al.,
bronchoalveolar lavage fluid; fever, headache, and weakness (Monto et al., 2011), a few can be potentially harmful in some
IBD, inflammatory bowel dis- 2000). These symptoms are often accompanied conditions; for example, some commensal bac-
ease; IEC, intestinal epithelial
cell; i.g., intragastrical(ly); i.n., by gastroenteritis-like symptoms in many influ- teria have been suggested to influence suscepti-
intranasal(ly); SFB, segmented enza patients, such as abdominal pain, nausea, bility to inflammatory bowel disease (IBD; Garrett
filamentous bacteria. vomiting, and diarrhea, especially in young chil- et al., 2007; Mazmanian et al., 2008).Thus, when
dren (Baden et al., 2009; Shinde et al., 2009; conditions in the host are unfavorable, such as
Dilantika et al., 2010). However, the immune during infection, the resulting changes within
mechanisms underlying these clinical manifes- the intestinal tract environment may promote
tations in the intestine during a lung-tropic growth of the harmful bacteria that induce in-
viral influenza infection remain unclear. testinal disease.
The intestinal tracts in humans and other It is well known that the respiratory and intes-
animals are inhabited by hundreds of diverse tinal tracts are both mucosal tissues. Over 30 years
species of commensal bacteria, which are essen- ago, John Bienenstock hypothesized that the
tial in shaping intestinal immune responses dur-
ing both health and disease (Hooper and Gordon, 2014 Wang et al. This article is distributed under the terms of an Attribution
NoncommercialShare AlikeNo Mirror Sites license for the first six months
after the publication date (see http://www.rupress.org/terms). After six months
it is available under a Creative Commons License (AttributionNoncommercial
Share Alike 3.0 Unported license, as described at http://creativecommons.org/
*J. Wang and F. Li contributed equally to this paper. licenses/by-nc-sa/3.0/).

The Rockefeller University Press $30.00


J. Exp. Med. 2014 Vol. 211 No. 12 23972410 2397
www.jem.org/cgi/doi/10.1084/jem.20140625
Figure 1. Respiratory influenza virus
infection causes lung and intestinal im-
mune injury. C57BL/6 mice were i.n. infected
with saline or 0.1 HA of PR8. (A) Body weight
was monitored after PR8 infection. (B) The
pathology of lung and small intestine was
assayed after PR8 infection. (C) The length of
colon was recorded after PR8 infection.
(D) The severity of the diarrhea was scored
after PR8 infection (0, normal stool or absent;
1, slightly wet and soft stool; 2, wet and un-
formed stool with moderate perianal staining
of the coat; and 3, watery stool with severe
perianal staining of the coat). (E) The pathol-
ogy of liver and kidney was assayed after PR8
infection. (F) Serum ALT and BUN levels were
measured after PR8 infection (dashed lines
represent damage threshold). All tissue sec-
tions were stained with H&E. Bars, 100 m.
Data represent three independent experi-
ments with at least five mice/group in A, C,
and D or three mice/group in B, E, and F. Data
are expressed as mean SEM by a Students
t test. ***, P < 0.001.

immune cells and structures contained in mucosal tissues were RESULTS


universally connected within the whole body. This common Intranasal (i.n.), but not intragastric (i.g.), infection
mucosal immune system concept speculated that the mucosal with influenza virus causes intestinal immune injury
immune system was itself an organ in which the mucosal To test whether intestinal injury was also a feature in a mouse
immune cells distributed throughout the body could inter- model of influenza, we infected mice i.n. with the A/PR/8/34
play between or among different mucosal tissues or organs (PR8) influenza virus strain. Indeed, their body weight grad-
(McDermott and Bienenstock, 1979; McDermott et al., 1980). ually decreased from days 2 to 9 as compared with saline-treated
Although this term was coined three decades ago, appreciation controls, which maintained their body weight over the same
of its importance is only just beginning. Much was learned period (Fig. 1 A). Furthermore, both the lung and small intes-
from the numerous studies conducted on the mucosal immune tine had severe injury after PR8 infection (Fig. 1 B). Colon
system during this time, which mainly focused on understand- length was shortened (Fig. 1 C) and mild diarrhea occurred
ing its individual components (Holmgren and Czerkinsky, (Fig. 1 D), further indicating intestinal injury (Zaki et al., 2010;
2005; Sato and Kiyono, 2012). Although a few studies have Murray and Rubio-Tapia, 2012). In contrast, nonmucosal liver
suggested that the mucosal immune system is a system-wide and kidney tissues appeared normal after PR8 infection (Fig. 1 E),
organ (Gallichan et al., 2001; Sobko et al., 2010), some questions which was also supported by ALT and BUN analysis (Fig. 1 F).
still need to be clarified. For example, how do the different com- Together, these data indicate that respiratory influenza infec-
ponents affect each other, and how is cross talk achieved among tion causes severe immune injury not only in the lung but also
the various mucosal sites (Gill et al., 2010)? in the intestine.
In this study, we found that lymphocytes derived from the To rule out the possibility that the influenza virus entered
respiratory mucosa specifically migrated into the intestinal the gastrointestinal tract and directly caused immune injury
mucosa during respiratory influenza infection by the CCL25 at this site, we tested for the presence of virus within the small
CCR9 chemokine axis and destroyed the intestinal microbiota intestine after i.n. infection and found that the influenza virus
homeostasis in the small intestine, finally leading to intestinal could not be detected at this site (Fig. 2 A). To test this possi-
immune injury. Our findings may provide new insights into not bility in a more rigorous way, we i.g. infected mice with PR8
only the mechanisms underlying intestinal immune injury in- and found that live virus could be detected in the intestinal
duced by influenza infection of the lung but also the interplay contents and intestinal tissues in a short time after infection, and
of immune cells between or among different mucosal sites. virus was completely cleared from these sites 3 d after infection

2398 Microbiota cause intestine injury during influenza | Wang et al.


Ar ticle

Figure 2. Influenza virus does not infect the


small intestine directly. (A) C57BL/6 mice were i.n.
infected with 0.1 HA of PR8. The levels of the influ-
enza virusderived matrix protein gene in lung and
small intestine were detected by PCR. (BE) C57BL/6
mice were i.g. infected with saline or 0.1 HA of PR8.
Viral titer in intestinal contents was determined by
50% tissue culture infective dose (TCID50) assay after
PR8 infection (B). The levels of the influenza virus
derived matrix protein gene in small intestine were
detected by PCR after PR8 infection (C). The pathol-
ogy of lung and small intestine was assayed after
PR8 infection, and tissue sections were stained with
H&E. Bar, 100 m (D). The length of colon was re-
corded after PR8 infection (E). Data represent three
independent experiments with at least three mice/
group in AE. Data are expressed as mean SEM by a
Students t test. NS: not significant.

(Fig. 2, B and C). However, pathological injury was not found into healthy WT mice increased the number of Enterobacteriaceae
in any of the examined tissues (Fig. 2, D and E). These results and caused intestinal immune injury in recipient mice even
collectively suggest that influenza infection does not directly in the absence of viral infection as compared with the intesti-
cause immune injury in the small intestine.Thus, we unexpect- nal microbiota from saline-treated mice (Fig. 3, E and F).Thus,
edly observed that influenza infection induced severe immune these data suggest that respiratory influenza infection induces
injury within the intestine only when the virus infected the re- intestinal immune injury by altering the composition of in-
spiratory tract and immune injury occurred in the lung. testinal microbiota.
Escherichia coli is an important component of Enterobacteria-
Intestinal microbiota is required for influenza-induced ceae, and pathogenic E. coli infection often causes vomiting and
intestinal immune injury diarrhea in humans (Ochoa and Contreras, 2011).The number
Changes in intestinal microbiota are often involved in the oc- of E. coli in the intestinal tract significantly increased after PR8
currence of intestinal inflammation in many mouse models infection (Fig. 3 G). Treating mice with streptomycinan
(Lupp et al., 2007; Maslowski et al., 2009). To determine antibiotic to which E. coli is sensitiveprotected mice against
whether intestinal microbiota was involved in influenza PR8 infection-induced immune injury to the small intestine
induced intestinal immune injury, we first assayed whether viral by inhibiting the increase of Enterobacteriaceae (Fig. 3, H and I).
infection affected the relative composition of several major Furthermore, directly infecting mice i.g. with E. coli caused
bacterial groups within the intestinal microbiota. Although immune injury in the small intestine (Fig. 3 J).Thus, these data
the number of total bacteria remained the same after infec- suggest that the increase of E. coli may be the primary cause for
tion as quantified by both real-time PCR and selective cul- intestinal immune injury during influenza infection.
ture (Fig. 3 A), the numbers of segmented filamentous bacteria
(SFB) and Lactobacillus/Lactococcus decreased after PR8 infec-
tion, whereas the number of Enterobacteriaceae increased; more- Th17 cells mediate influenza-induced intestinal immune injury
over, the numbers of mouse intestinal Bacteroides, Eubacterium To explore the mechanism by which intestinal bacteria caused
rectale/Clostridium coccoides, and Bacteroides were unchanged intestinal immune injury during influenza infection, many dif-
(Fig. 3 B). We next administered combinatorial antibiotics to ferent types of proinflammatory cells involved in intestinal
the mice via their drinking water to deplete intestinal micro- inflammation (Zhou et al., 2007a; Kleinschek et al., 2009;
biota (Ichinohe et al., 2011) 4 wk before infecting them with Leppkes et al., 2009) were examined. Depletion of NK1.1+
PR8. In antibiotic-treated mice, the lungs still sustained severe by specific antibodies or T cell deficiency could not re-
immune-mediated injury after PR8 infection, but the small duce the PR8 infection-induced intestinal immune injury in
intestine and colon were protected (Fig. 3, C and D). In another our study (Fig. 4, A and B). However, no intestinal injury was
way, transferring intestinal microbiota from PR8-infected mice observed in IL-17A/ mice after PR8 infection (Fig. 4 C),

JEM Vol. 211, No. 12 2399


Figure 3. Antibiotic treatment reduces influenza-induced intestinal immune injury. (A) Bacteria in the small intestine were assayed by real-time
PCR and selective culture in blood plate 7 d after PR8 infection. (B) Several major bacterial groups in intestinal microbiota were assayed by real-time PCR
7 d after PR8 infection. (C and D) C57BL/6 mice were subjected to a 4-wk oral treatment of combinatorial antibiotics in drinking water, followed by
i.n. infection with saline or 0.1 HA of PR8. The pathology of lung and small intestine was assayed 7 d after PR8 infection (C). The length of colon was
recorded 7 d after PR8 infection (D). (E and F) Transfer of intestinal microbiota from saline-treated or PR8-infected mice into healthy WT mice by the
i.g. route. Major bacterial groups in the intestinal microbiota (E) and the pathology of small intestine were assayed 6 d later (F). (G) The number of E. coli
in stool was detected by E. coli/Coliform Count Plates 6 d after PR8 infection. (H and I) C57BL/6 mice were subjected to a 1-wk oral treatment of streptomycin
in their drinking water and then were i.n. infected with 0.1 HA of PR8. The pathology of lung and small intestine (H) and major bacterial groups in intesti-
nal microbiota (I) were assayed 6 d after PR8 infection. (J) C57BL/6 mice were i.g. infected with saline or 5 108 E. coli, and the pathology of small intes-
tine was assayed 3 d later. All tissue sections were stained with H&E. Bars, 100 m. Data represent two independent experiments with three mice/group
in I and J or three independent experiments with at least three mice/group in AH. Data are expressed as mean SEM by a Students t test. *, P < 0.05;
**, P < 0.01; NS: not significant.

2400 Microbiota cause intestine injury during influenza | Wang et al.


Ar ticle

Figure 4. IL-17A deficiency reduces


influenza-induced immune injury in small
intestine but not in lung. (A) The pathology
of lung and small intestine from control and
PK136-treated mice was assayed 6 d after
PR8 infection. (B) The pathology of lung and
small intestine from WT and Tcrd/ mice
was assayed 6 d after PR8 infection. (C) The
pathology of lung and small intestine from
WT and IL-17A/ mice was assayed 6 d after
PR8 infection. (D) IL-17A and IL-17F expres-
sions in the lung from WT mice were de-
tected by real-time PCR 6 d after PR8
infection. (E) Body weight of WT and
IL-17A/ mice was monitored after PR8 infec-
tion. (F) Evans blue dye concentration in BALF
from WT and IL-17A/ mice was determined
by spectrophotometer 6 d after PR8 infec-
tion. (G and H) Total protein (G) and lactate
dehydrogenase (H) levels in BALF from WT
and IL-17A/ mice were determined by
ELISA 6 d after PR8 infection. All tissue sec-
tions were stained with H&E. Bars, 100 m.
Data represent two independent experiments
with five mice/group in EH or three mice/
group in AD. Data are expressed as mean
SEM by a Students t test. *, P < 0.05; **,
P < 0.01; ***, P < 0.001; NS: not significant.

suggesting that Th17 cells might be involved in influenza- et al., 2011). Furthermore, treating mice i.p. with a neutralizing
induced intestinal immune injury. antiIL-17A antibody during PR8 infection effectively reduced
To rule out the possibility that lung injury might also be intestinal injury (Fig. 5 E). Together, these data suggest that
reduced in IL-17A/ mice after influenza infection, which influenza infectioninduced intestinal immune injury is depen-
subsequently resulted in reducing the small intestinal injury dent on Th17 cells.
indirectly, we compared the degree of the lung injury after Because we observed that influenzainduced intestinal im-
influenza infection between WT and IL-17A/ mice.The re- mune injury is dependent on both intestinal microbiota and
sults showed that both IL-17F and IL-17A expressions in lung Th17 cells, we wondered whether there was an association
from WT mice were increased after PR8 infection (Fig. 4 D). between intestinal bacteria and Th17 cells.The results showed
Compared with WT mice, IL-17A/ mice exhibited re- that the percentage and number of Th17 cells in the small in-
duced body weight loss during PR8 infection (Fig. 4 E). How- testine were unchanged in antibiotic-treated mice after PR8
ever, the degree of lung leak and the levels of total protein and infection as compared with uninfected control mice (Fig. 5 F);
lactate dehydrogenase in bronchoalveolar lavage fluid (BALF) transferring intestinal microbiota from PR8-infected mice into
were not significantly different between WT and IL-17A/ healthy WT mice promoted IL-17A expression in the small
mice (Fig. 4, FH), suggesting that the lung injury did not re- intestine of recipient mice (Fig. 5 G); and streptomycin treat-
duce in IL-17A/ mice after influenza infection when com- ment inhibited IL-17A expression in the small intestine dur-
pared with WT mice.Thus, these data suggest that the decrease ing PR8 infection (Fig. 5 H). Collectively, these data suggest
of immune injury in the small intestine from IL-17A/ mice that changes in intestinal microbiota induced by influenza in-
after influenza infection is independent of the decrease of fection promote Th17 cell production, which subsequently
lung injury. causes intestinal immune injury.
To further determine that Th17 cells were responsible for
influenza-induced intestinal immune injury, we detected the CCL25/CCR9 mediates the recruitment of lung-derived
expression of Th17-specific transcription factor RORt and CD4+ T cells into the small intestine
IL-17A and found that their expressions increased in the small Because respiratory influenza infection influences the compo-
intestine after PR8 infection (Fig. 5 A). The percentage and sition of intestinal microbiota, which subsequently promotes
number of Th17 cells increased in the small intestine and colon Th17 cell production and causes intestinal immune injury, we
after PR8 infection (Fig. 5, B and C), but not in the liver or kid- wanted to know how respiratory influenza infection destroyed
ney (Fig. 5 D), consistent with previous observations (Esplugues the microecological homeostasis of the intestinal microbiota.

JEM Vol. 211, No. 12 2401


Figure 5. Increased Th17 cells occur in the
small intestine during influenza virus infec-
tion. (A) RORt and IL-17A expressions in the
small intestine were detected by real-time PCR
7 d after PR8 infection. (B) The percentage and
number of Th17 cells in intestinal IEL and LPL
were detected 7 d after PR8 infection. (C) The
percentage and number of Th17 cells in colonic
LPL were detected 7 d after PR8 infection. (D) The
number of Th17 cells in liver and kidney was de-
tected 7 d after PR8 infection. (E) C57BL/6 mice
were i.p. treated with a neutralizing antiIL-17A
antibody during PR8 infection. The pathology of
lung and small intestine was assayed 6 d after
PR8 infection, and tissue sections were stained
with H&E. Bars, 100 m. (F) The percentage and
number of Th17 cells in IEL and LPL were detected
7 d after PR8 infection in antibiotic-treated mice.
(G) Transfer of intestinal microbiota from saline-
treated or PR8-infected mice into healthy WT
mice by the i.g. route. IL-17A expression in the
small intestine was detected by real-time PCR 6 d
later. (H) IL-17A expression in the small intestine
was detected by real-time PCR at day 6 after PR8
infection in streptomycin-treated mice. Data
represent two independent experiments with
three mice/group in A, D, E, G, and H or three
independent experiments with three mice/group
in B, C, and F. Data are expressed as mean SEM
by a Students t test. *, P < 0.05; **, P < 0.01;
***, P < 0.001; NS: not significant.

Given that influenza infection specifically caused immune in- in the small intestine (Fig. 6 D). These results suggest that the
jury in the respiratory and intestinal mucosal tissues, but not in CCL25CCR9 axis contributes to altering the composition
the nonmucosal liver or kidney in our study, an interconnected of the intestinal microbiota after influenza infection and the
relationship existed between them was intriguing according subsequent development of intestinal inflammation via recruit-
to the common mucosal immune system theory (McDermott ing effector lymphocytes into the intestinal mucosa.
and Bienenstock, 1979; McDermott et al., 1980). The CCL25 Next, we explored which lymphocyte subsets were recruited
chemokine is expressed by intestinal epithelial cells (IECs) and by CCL25 in our influenza model. Although the total number
functions to specifically guide CCR9-expressing effector lym- of T cells increased in the LPL after PR8 infection, the total
phocytes into the small intestine as a homing mechanism number of B cells decreased (Fig. 6 E).Within the T cell popula-
(Campbell and Butcher, 2002). Consistent with previous ob- tion, the CCR9+CD4+ T cell subset increased (Fig. 6 F), whereas
servations, CCL25 expression in the small intestine tissue was the CCR9+CD8+ T cell subset remained unchanged (Fig. 6 G),
much higher than any other tissues, including liver, kidney, and indicating that CCR9+CD4+ T cells might play a key role in al-
lung (Fig. 6 A). Treating mice i.v. with a neutralizing anti- tering the intestinal microbiota. Evaluating this subpopulation
CCL25 antibody during PR8 infection reduced intestinal in other tissues revealed that the number of CCR9+CD4+ T cells
immune injury (Fig. 6 B), inhibited the changes in intestinal was significantly increased in the lung and in the mediastinal
microbiota (Fig. 6 C), and reduced the number of Th17 cells LNs after PR8 infection, but not in the mesenteric LNs (Fig. 6 G),

2402 Microbiota cause intestine injury during influenza | Wang et al.


Ar ticle

Figure 6. Anti-CCL25 antibody treatment reduces influenzainduced intestinal immune injury. (A) CCL25 expression in various tissues was de-
tected by real-time PCR 4 d after PR8 infection. (BD) C57BL/6 mice were i.v. treated with a neutralizing anti-CCL25 antibody during PR8 infection. The
pathology of lung and small intestine (B), major bacterial groups in intestinal microbiota (C), and the number of Th17 cells in IEL and LPL were assayed 7 d
after PR8 infection (D). (EG) C57BL/6 mice were i.n. infected with saline or 0.1 HA of PR8. The number of T and B cells in LPL (E), the percentage and
number of CCR9+CD4+ T cells in small intestine (F), and the number of CCR9+CD8+ T cells in LPL and CCR9+CD4+ T cells in lung, mediastinal LNs, and mes-
enteric LNs were assayed 7 d after PR8 infection (G). (H) ALDH1A2 expression in lung was detected by real-time PCR 6 d after PR8 infection. (I) CD4+
T cells from the lungs of saline- or PR8-infected CD45.1+ mice were adoptively transferred into WT CD45.2+ mice, and the percentage of CD45.1+CD4+
T cells in total CD4+ T cells in LPL from recipient CD45.2+ mice was detected by flow cytometry 48 h later. (J) C57BL/6 mice were i.n. infected with saline or
0.1 HA of PR8. CD4+ T cells in the lung and LPL were purified 6 d later by MACS and then co-cultured with antigen-presenting cells and heat-killed PR8
in an IFN- ELISPOT plate. The number of positive spots was counted 20 h later. (K) Parabiotic pairs of WT mice were established first, and the left partner
was i.n. infected with PR8 2 wk later. The pathology of small intestine was assayed 6 d after PR8 infection. All tissue sections were stained with H&E.
Bars, 100 m. Data represent three independent experiments with three mice/group in AH and K or three wells/treatment in J. Data are expressed as
mean SEM by a Students t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001; NS: not significant.

JEM Vol. 211, No. 12 2403


Figure 7. Lung-derived CD4+ T cells influence microbiota and intestine injury by secreting IFN-. (A) IFN- expression in CD4+ T cells from lung
was detected by flow cytometry 6 d after PR8 infection in WT mice. (B) The pathology of small intestine was assayed at day 7 after PR8 infection in WT
and IFN-/ mice, and tissue sections were stained with H&E. Bars, 100 m. (C and D) IL-17A expression in the small intestine (C) and major bacterial
groups in intestinal microbiota (D) were assayed at day 7 after PR8 infection in IFN-/ mice. (E and F) IL-17A expression in CD4+ T cells from lung (E)
and the percentages of CCR9 Th17 cells and CCR9+ Th17 cells in lung and LPL (F) were detected 6 d after PR8 infection in WT mice. (G) IL-17A expression
in CD4+ T cells from mesenteric LNs, Peyers patches, and blood was detected by flow cytometry 6 d after PR8 infection in WT mice. (H) IL-17A level in
serum was detected by ELISA 6 d after PR8 infection in WT mice. (I) LPL from WT mice at day 6 after PR8 infection was stimulated by heat-killed E. coli
in vitro; 24 h later, the expression of IL-17A in CD4+ T cells was detected by flow cytometry. (J) Lung lymphocytes and LPL from WT mice at day 6 after PR8

2404 Microbiota cause intestine injury during influenza | Wang et al.


Ar ticle

suggesting that the lung and mediastinal LNs might be the main small intestine (Fig. 7 J). Thus, these data suggest that Th17 cell
sources of CCR9+CD4+ T cells recruited to the small intestine polarization, but not recruitment, occurs in the small intestine
during PR8 infection. Retinoic acid is reported to promote the in situ during influenza infection.
expression of CCR9 on T cells (Ohoka et al., 2011), and the
production of retinoic acid is regulated by the aldehyde dehy- Intestinal microbiotainduced IL-15 production
drogenase (ALDH) 1A2 (Yokota et al., 2009). In our study, the promotes intestinal Th17 cell polarization
expression of ALDH1A2 in lung increased after influenza infec- Because Th17 cell polarization occurs in the small intestine in
tion (Fig. 6 H), suggesting that the increase of retinoic acid in situ during influenza infection, we next explore what kind of
lung after influenza infection might be responsible for promot- factors mediated this process. IL-6 expression in the small intes-
ing the CCR9 expression on lung CD4+ T cells. tine was increased after PR8 infection, but IL-23 and TGF-
To determine whether influenza infectionactivated lung expressions were unchanged (Fig. 8 A). However, treating mice
CD4+ T cells tended to migrate into the small intestine, we i.v. with a neutralizing antiIL-6 antibody during PR8 infection
adoptively transferred lung CD4+ T cells from saline- or PR8- could not reduce intestinal immune injury (Fig. 8 B). Thus, the
infected CD45.1+ mice into recipient WT CD45.2+ mice and increase of IL-6 is not the main reason for Th17 cell polarization
found that LPL in recipient mice contained a higher frequency in our study. IL-15 has been reported to contribute to intestinal
of CD45.1+CD4+ T cells from PR8-infected CD45.1+ mice inflammation in various mouse models (Zhou et al., 2007b;
(Fig. 6 I). Moreover, PR8-specific CD4+ T cells were detected Schulthess et al., 2012) and, importantly, it has been shown to in-
not only in lung but also in the small intestine after PR8 in- duce IL-17A expression in both mice and human CD4+ T lym-
fection, as assessed by the IFN- ELISPOT plate (Fig. 6 J), phocytes (Ziolkowska et al., 2000; Ferretti et al., 2003). In our
and, in a parabiotic mice model, PR8 infection in one partner study, IL-15 expression in the small intestine, but not in serum,
caused small intestinal injury to occur in a noninfected partner was up-regulated after PR8 infection (Fig. 8 C).Transferring in-
(Fig. 6 K). Thus, these data suggest that the CCL25CCR9 testinal microbiota from PR8-infected mice also increased IL-15
axis mediates the recruitment of lung-derived effector CD4+ expression in the small intestine of recipient mice (Fig. 8 D). To
T cells into the small intestine as well as the alterations to the explore whether IL-15 contributed to Th17 cell polarization in
intestinal microbiota composition during influenza infection. our study, we first assayed the expression of IL-15 receptor and
found that intestinal CD4+ T cells expressed the IL-15 receptor
Lung-derived CD4+ T cells destroy microbiota homeostasis after PR8 infection (Fig. 8 E). Next, treating mice with a neu-
and promote resident Th17 cell polarization tralizing antiIL-15 antibody during PR8 infection effectively
As we found that lung-derived effector CD4+ T cells are reduced intestinal immune injury (Fig. 8 F).Thus, IL-15, which
recruited into the small intestine and alter the intestinal mi- was induced by intestinal bacteria, contributes to intestinal im-
crobiota during influenza infection, we wondered how they mune injury during influenza infection. Further experiments
influenced the intestinal microbiota composition and whether showed that IL-15 neutralization inhibited IL-17A and IL-6 ex-
Th17 cells in the small intestine originated from the polariza- pression in the small intestine after PR8 infection (Fig. 8 G) and,
tion of them. For the first question, IFN- expression was found consistent with the previous observations (Ziolkowska et al.,
to be significantly increased in lung CD4+ T cells after PR8 2000; Ferretti et al., 2003), exogenous IL-15 promoted IL-17A
infection (Fig. 7 A). When IFN- was deficient, the mice ex- secretion in purified CD4+ T cells from LPL in vitro (Fig. 8 H),
hibited reduced intestinal immune injury, normal IL-17A ex- suggesting that intestinal bacteriainduced IL-15 might promote
pression, and unchanged intestinal microbiota in the small Th17 cell polarization in the small intestine in situ by a direct
intestine after PR8 infection (Fig. 7, BD). Thus, these data and/or indirect way. However, IL-15 neutralization did not in-
suggest that lung-derived effector CD4+ T cells destroy the fluence the changes of the intestinal microbiota (Fig. 8 I), sug-
homeostasis of intestinal microbiota by secreting IFN-. For gesting that IL-15 functioned upstream of IL-17A production but
the second question,Th17 cells were not found to be increased downstream of the change in microbiota after PR8 infection.
in lung after PR8 infection (Fig. 7 E) and, although some CCR9+ Exploring the in vivo cellular sources of IL-15, high IL-15
Th17 cells were present in the small intestine, most Th17 cells expression was detected in IECs after PR8 infection (Fig. 8 J),
(90%) exhibited a CCR9 phenotype (Fig. 7 F). Mean- suggesting that IECs might be an important source of IL-15
while,Th17 cells were also not found increased in the mesen- in the small intestine during influenza infection.
teric LNs, Peyers patches, and blood (Fig. 7 G), and IL-17A
levels in blood did not increased after PR8 infection (Fig. 7 H). DISCUSSION
More convincing evidences showed that E. colispecific Th17 Mucosal tissues, including the gastrointestinal, respiratory, and
cells could be detected in the small intestine (Fig. 7 I), but PR8- urogenital tracts, etc., are the first line of host defense against ex-
specific Th17 cells could not be detected both in the lung and ternal invaders. Although much has been learned from studying

infection were stimulated by heat-killed PR8 in vitro; 24 h later, the expression of IL-17A in CD4+ T cells was detected by flow cytometry. Data represent
three independent experiments with three mice/group in AH or three wells/treatment in I and J. Data are expressed as mean SEM by a Students t test.
*, P < 0.05; **, P < 0.01; ***, P < 0.001; NS: not significant.

JEM Vol. 211, No. 12 2405


Figure 8. Intestinal microbiota induces
Th17 cell polarization in situ via trigger-
ing IL-15 production. (A) IL-6, IL-23, and
TGF- expressions in the small intestine were
detected by real-time PCR 6 d after PR8 in-
fection. (B) The pathology of lung and small
intestine from control and antiIL-6treated
mice was assayed 6 d after PR8 infection.
(C) IL-15 expression in the small intestine and
serum was detected 6 d after PR8 infection.
(D) Transfer of intestinal microbiota from
saline-treated or PR8-infected mice into
healthy WT mice by the i.g. route. IL-15 ex-
pression in the small intestine was detected
6 d later. (E) IL-15R expression on CD4+ T cells
in LPL from WT mice was detected at day 6
after PR8 infection. (F and G) C57BL/6 mice
were i.p. treated with a neutralizing antiIL-15
antibody during PR8 infection. The pathology
of lung and small intestine (F) as well as
IL-17A and IL-6 expressions in the small in-
testine were assayed 6 d after PR8 infection
(G). (H) MACS-purified CD4+ T cells from LPL
were stimulated by IL-15 in vitro, and IL-17A
levels in supernatant were measured at days 2
and 3 by ELISA. (I) Major bacterial groups in
the intestinal microbiota from control and
antiIL-15treated mice were assayed by real-
time PCR 6 d after PR8 infection. (J) IL-15
expression in IECs was detected 6 d after PR8
infection in WT mice. All tissue sections were
stained with H&E. Bars, 100 m. Data repre-
sent three independent experiments with
three mice/group in AG, I, and J or three
wells/treatment in H. Data are expressed as
mean SEM by a Students t test. *, P < 0.05;
**, P < 0.01; ***, P < 0.001; NS: not significant.

each of these components individually, the mucosal immune particularly at the early stage of infection. Although some stud-
system has not yet been examined from a holistic point of view ies suggest that influenza virus disseminates into extrapulmo-
as a system-wide organ (Gill et al., 2010), as conceptualized by nary tissues or organs during infection (Korteweg and Gu,
the common mucosal immune system hypothesis. Unexpect- 2008), others contradict this finding (Mauad et al., 2010), and
edly, we observed that respiratory influenza infection in mice direct evidence for viral replication in extrapulmonary tissues
caused immune injury not only in the lung but also specifically or organs has not yet been shown (Kuiken and Taubenberger,
in the intestine, as it had no influence on the pathology in non- 2008). It therefore remains a mystery how influenza infection
mucosal organs such as the liver or kidney. Because this resem- can be associated with immune injury to extrapulmonary tis-
bles the symptoms exhibited by humans after influenza infection, sues or organs if these injuries are not induced by direct virus
these influenza virusinfected mice provide a good model in infection of these tissues or organs (Polakos et al., 2006; Mauad
which to study the mechanisms underlying how respiratory in- et al., 2010). In our mouse model of respiratory influenza in-
fluenza infection causes intestinal immune injury; furthermore, fection, no influenza virus was detected in the small intestine,
these observations provide further evidence to support the ex- and i.g. administration of the influenza virus directly into the
istence of a common mucosal immune system. intestine did not lead to intestinal immune injury. Thus, the
Pathogens extensively disseminate beyond the limits of the intestinal immune injury observed in our study was not directly
primary infection site in almost all cases of infectious diseases, caused by influenza infection of the intestine.

2406 Microbiota cause intestine injury during influenza | Wang et al.


Ar ticle

An explosive increase in neutrophils is responsible for et al., 2010). These findings suggest that potential exists for an
influenza-induced acute lung injury and death. IL-17 is a po- undetermined link between mucosal immune components and
tent regulator for the neutrophils recruitment. Previous studies that each component is efficient at sharing information distally
have shown that respiratory influenza infection increases the (Gill et al., 2010). In our study, we found that lung-derived virus-
IL-17A and IL-17F expressions in lung, and IL-17RA/ specific effector CCR9+CD4+ T cells were recruited into the
mice exhibit reduced lung injury and higher survival rates small intestine and destroyed the homeostasis of intestinal mi-
after influenza infection (Crowe et al., 2009; Li et al., 2012). crobiota by secreting IFN- after influenza infection. Thus,
In our mouse model of respiratory influenza infection, IL-17A we speculated that the effector CCR9+CD4+ T cells might enter
and IL-17F expressions in lung also increased after infection, into the small intestine by a special way as described above and
but IL-17A deficiency could not reduce influenza-induced remained in the active state to secrete IFN- even in the ab-
lung injury. Thus, based on above results, we speculated that sence of antigen stimulation.
IL-17A and IL-17F played the same function during influ- The intestinal microbiota is extensively accepted in the
enza infection, and IL-17F alone might be enough to function field as a virtual metabolic organ in and of itself (OHara and
to activate IL-17RA and recruit neutrophils when IL-17A Shanahan, 2006). Beyond this role in metabolism, the intestinal
was deficiency. microbiota has a conspicuous effect on host immune functions,
Recruitment and infiltration of inflammatory cells into the as indicated by comparing immune responses between germ-
gastrointestinal mucosa critically regulates the development as free and conventional animals. A previous study showed that
well as progression of IBD (Wurbel et al., 2011). Differential commensal SFBs induce IL-6 and IL-23 production to stimu-
expression of chemokine receptors and adhesion molecules late Th17 cell polarization (Ivanov et al., 2009). However, in
on lymphocytes not only determine their migration into dif- our mouse model of respiratory influenza infection, the num-
ferent tissues but also their localization within these tissues. ber of SFB decreased while the number of E. coli increased in
CCL25 is constitutively expressed by the epithelium of the small the intestinal tract after influenza infection; E. coli promoted
intestine (Papadakis et al., 2000), and the CCL25CCR9 che- IL-15 expression in IECs, and this IL-15 then promoted Th17
mokine axis is considered to be one of the few non-promiscuous cell polarization. Moreover, overgrowth of existing strain
chemokine/receptor pairs involved in gut-specific migration and/or acquisition of new pathogenic strain are involved in
of lymphocytes (Stenstad et al., 2006). In our study, the per- E. colicaused gastrointestinal symptoms (Nguyen et al., 2006;
centage and number of CCR9+CD4+ T cells in both lung and Ochoa and Contreras, 2011). In our study, considering that the
small intestine increased after influenza infection, and neutral- mice live in an SPF environment and that intestinal inflamma-
izing CCL25 with antibody treatment reduced CCR9+CD4+ tion occurs in different kinds of mice, we think that the over-
T cell recruitment and intestinal immune injury. Thus, these growth of existing E. coli in the gut may be the primary cause
data might explain why influenza infection specifically caused for intestinal immune injury during influenza infection.
immune injury in the intestine, but not the liver or kidney, in The function of IL-15 in regulation of Th17 responses has
our study. been studied extensively, but there are still some controversies.
IBD is a common disease characterized by severe inflam- Some studies found that IL-15 induces IL-17A expression in
mation of the intestine (Hooper and Macpherson, 2010). How- both mice and human CD4+T lymphocytes directly (Ziolkowska
ever, the exact causes of this disease remain unclear. Some studies et al., 2000; Ferretti et al., 2003), which was also demonstrated
suggest that IBD arises from dysregulated control of host in our study. However, another study showed that IL-15 inhib-
microorganism interactions. For example, patients with this its Th17 cell polarization in a mouse model of EAE (Pandiyan
disease have an increased number of epithelial cell surface et al., 2012). We thought that there were two main reasons to
associated bacteria (Swidsinski et al., 2005), suggesting the fail- explain why IL-15 played the opposite effect in different mouse
ure of a mechanism designed to limit direct contact between model: (1) the immuno-microenvironment in different mouse
the epithelium and the microbiota. Similarly, in our study, we model is different; (2) IL-15 is reported to activate both STAT3
also found that CCR9+CD4+ T cell recruitment correlated and STAT5 (Johnston et al., 1995), which is inferred either to
to intestinal inflammation by the following mechanism: the inhibit or to promote Th17 cells.
CCR9+CD4+ T cells destroyed the homeostasis of the intestinal
microbiota, the altered microbiota then promoted Th17 cell po-
MATERIALS AND METHODS
larization in the small intestine in situ, and the resulting IL-17A Mice, virus, and bacteria. Male C57BL/6 mice were purchased from
secretion finally mediated the intestinal immune injury. the Shanghai Laboratory Animal Center, Chinese Academy of Sciences.
This concept of the common mucosal immune system pro- IFN-/, Tcrd/, and IL-17A/ mice were purchased from The Jackson
posed by John Bienenstock suggests that the mucosal immune Laboratory. All mice were housed under specific pathogen-free conditions at
system may be considered as one large interconnected network the School of Life Sciences, University of Science and Technology of China
(McDermott and Bienenstock, 1979; McDermott et al., 1980), (USTC), and were used at 610 wk of age. Animal care and experimental
procedures were followed in accordance with the experimental animal guide-
which is supported by some recent researches. For example, lines at USTC. Mouse-adapted influenza A/PR/8/34 strain (H1N1) was a
i.n. immunization results in vaginal protection against genital gift from H. Meng (Institute of Basic Medicine, Shandong Academy of Med-
HSV-2 infection (Neutra and Kozlowski, 2006), and antibiot- ical Sciences, Shandong, China). For influenza infection studies, mice were
ics used in neonates increases the risk to develop asthma (Sobko anesthetized and infected i.n. with 0.1 HA of PR8 in 50 l sterile saline.

JEM Vol. 211, No. 12 2407


E. coli strain was isolated from stool of PR8-infected mice by the 3M Petri- or isotype control Abs. Samples were collected by a flow cytometer (LSR II;
film E. coli/Coliform Count Plate and was cultured in broth medium for BD) and analyzed by FlowJo and WinMDI 2.9 software.
amplification. For E. coli infection, mice were anesthetized and infected i.g.
with 5 108 E.coli in 500 l sterile saline. Real-time PCR. Total RNA was extracted from tissues using TRIzol (Invi-
trogen), and cDNA was then synthesized. Real-time PCR was performed
Histopathology. Lung, small intestine, liver, and kidney tissues were re- according to the manufacturers instructions using a SYBR Premix Ex Taq
moved and fixed immediately in 10% neutral-buffered formalin in PBS for (Takara Bio Inc.). For analysis, target gene expression was normalized to the
>24 h, embedded in paraffin, and cut into 57-m sections. The sections housekeeping gene -actin. Gene expression values were then calculated
were deparaffinized and stained with hematoxylin and eosin (H&E) to deter- using the mean from the control samples as a calibrator. Real-time PCR
mine histological changes. primers were synthesized by Sangon Biotech (Table S1).

Neutralizing antibodies and antibiotic treatment. For in vivo neutral-


Analysis of lung injury. Lung leakage: 1 h before sacrificing mice, 20 mg/kg
ization, the following neutralizing antibodies were administered: 100 g/
Evans blue dye was administered i.v. The lung was instilled with 1 ml of
mouse antiIL-17A (TC11-18H10.1), 100 g/mouse anti-CCL25 (89818),
saline, and the BALF was collected. After centrifugation, Evans blue dye con-
100 g/mouse antiIL-6 (MP5-20F3), or 100 g/mouse antiIL-15 (AIO.3)
centration in supernatant was determined by spectrophotometer at 620 nm.
were administered into mice at days 0, 2, and 4 after PR8 infection. For
Total protein and lactic dehydrogenase in BALF: The lung was instilled with
in vivo cell depletion, 200 g/mouse anti-NK1.1 was administered i.v. into
1 ml saline, and the BALF was collected. After centrifugation, the level of
mice 2 d before PR8 infection. For intestinal microbiota depletion, mice
total protein in supernatant was assayed by the BCA protein assay kit, and the
were treated with a mixture of antibiotics (1 mg/ml ampicillin, 0.5 mg/ml
level of lactic dehydrogenase in supernatant was assayed by ELISA kit (Cloud-
vancomycin, 1 mg/ml neomycin sulfate, and 1 mg/ml metronidazole [San-
Clone Corp).
gon Biotech]) added to their drinking water beginning 4 wk before PR8 in-
fection and continuing until sacrifice, as previously described (Ichinohe et al.,
Analysis of liver and kidney function. Serum from infected mice or 2011). For intestinal E. coli depletion, mice were treated with 1 mg/ml strep-
control mice were collected and stored at 80C until analysis. Liver func- tomycin (Sangon Biotech) added to their drinking water beginning 1 wk
tion was determined by measuring serum ALT (alanine aminotransferase) using before PR8 infection and continuing until sacrifice. Antibiotic-containing
a commercially available kit (Rong Sheng). Kidney function was assessed by water was changed twice a week.
measuring serum BUN (blood urea nitrogen) using a commercially available
kit (Jiancheng Bioengineering Institute). Microbiota transplantation. Cecal contents from saline- or PR8-infected
mice were suspended in 1 ml saline and were administered (0.5 ml per mouse)
Determination of virus and bacteria. Influenza virus in the lung and immediately to WT mice by the i.g. route.Transplanted mice were maintained
small intestine were detected by PCR. The primer sequences to detect the in sterile cages and detected intestinal immune injury 7 d later.
gene encoding the matrix protein within the influenza virus were as follows:
5-GGACTGCAGCGTAGACGCTT-3 (forward) and 5-CATCCTGTT- Transfer of T cells and PR8-specific CD4+ T cells assay. For T cells
GTATATGAGGCCCAT-3 (reverse). Intestinal bacterial genomic DNA transfer, 5 105 CD4+ T cells from the lungs of saline- or PR8-infected
was extracted from the stool using a stool kit (QIAGEN) according to the CD45.1+ mice were adoptively transferred i.v. into WT CD45.2+ mice, and
manufacturers instruction (the optional high-temperature step was per- the percentage of CD45.1+CD4+ T cells in total CD4+ T cells in LPL from
formed). The abundance of total and specific intestinal bacterial groups was recipient CD45.2+ mice was detected 48 h later by flow cytometry. For
measured by real-time PCR with corresponding 16S rDNA gene primers PR8-specfic CD4+ T cells assay, CD4+ T cells in the lung and LPL from
(Sangon Biotech; Table S1). The number of E. coli in stool was detected by saline-treated and PR8-infected mice were purified 6 d later by MACS and
the 3M Petrifilm E. coli/Coliform Count Plate according to the manufac- then co-cultured with antigen-presenting cells and heat-killed PR8 in an
turers instructions. IFN- ELISPOT plate. The number of positive spots was counted 20 h later
according to the manufacturers instructions.
Isolation of IEC, IEL, and LPL. IECs were isolated as described in a pre-
vious study (Zhou et al., 2007a). IELs and LPLs were isolated as previously Statistical analysis. A two-tailed Students t test was used for statistical
described with minor modifications (Das et al., 2003; Kamanaka et al., 2006; analyses. Data were expressed as the mean SEM, and the data were con-
Esplugues et al., 2011). In brief, small intestines were harvested and washed sidered statistically significant when differences achieved values of P < 0.05.
with PBS, and mesentery and Peyers patches were carefully dissected out. In-
testines were opened longitudinally and then cut into 1-cm pieces. Intestinal Online supplemental material. Table S1 shows primers used for real-
pieces were incubated in 10 ml of extraction buffer (5% FCS, 1 mM DTT, time PCR. Online supplemental material is available at http://www.jem
and 5 mM EDTA in PBS) at 37C for 30 min.The released cells were loaded .org/cgi/content/full/jem.20140625/DC1.
onto a Percoll gradient and centrifuged.The cells at the interface of a 40/70%
Percoll solution were collected and used as IELs. The remaining segments We thank H. Meng (Shandong Academy of Medical Sciences) for the influenza
were incubated twice in extraction buffer to remove IELs and isolate LPLs. A/PR/8/34 strain.
This work was supported by Ministry of Science & Technology of China
The tissue was digested with prewarmed complete RPMI1640 containing
(#2010CB911901 and #2013CB530506), Natural Science Foundation of China
2 mg/ml collagenase IV at 37C for 60 min, loaded onto a Percoll gradient,
(#31300753 and #31400783), Fundamental Research Funds for the Central
and centrifuged. The cells at the interface of a 40/70% Percoll solution were
Universities (#WK2070000039), and China Postdoctoral Science Foundation
collected and used as LPLs. (#2013M531532 and #2014T70599).

Flow cytometry. After blocking the Fc receptor with anti-CD16/CD32, Author contributions: J. Wang and F. Li performed experiments. J. Wang, F. Li,
single-cell suspensions were incubated with the fluorescently labeled mAbs H. Wei, Z.-X. Lian, R. Sun, and Z. Tian designed the research. J. Wang, F. Li, and Z. Tian
at 4C for 30 min in PBS and then washed twice. For intracellular cytokine wrote the manuscript. Z. Tian supervised the project. J. Wang and F. Li contributed
staining, cells were first stimulated for 4 h at 37C with 50 ng/ml PMA, equally to this paper.
1 g/ml ionomycin, and 10 g/ml monensin (all from Sigma-Aldrich); cells
were then stained for extracellular markers, fixed, permeabilized, and stained Submitted: 3 April 2014
with the fluorescently labeled mAbs against the indicated intracellular cytokines Accepted: 14 October 2014

2408 Microbiota cause intestine injury during influenza | Wang et al.


Ar ticle

REFERENCES Kleinschek, M.A., K. Boniface, S. Sadekova, J. Grein, E.E. Murphy, S.P.Turner,


Baden, L.R., J.M. Drazen, P.A. Kritek, G.D. Curfman, S. Morrissey, and E.W. L. Raskin, B. Desai, W.A. Faubion, R. de Waal Malefyt, et al. 2009.
Campion. 2009. H1N1 influenza A diseaseinformation for health pro- Circulating and gut-resident human Th17 cells express CD161 and pro-
fessionals. N. Engl. J. Med. 360:26662667. http://dx.doi.org/10.1056/ mote intestinal inflammation. J. Exp. Med. 206:525534. http://dx.doi
NEJMe0903992 .org/10.1084/jem.20081712
Campbell, D.J., and E.C. Butcher. 2002. Intestinal attraction: CCL25 func- Korteweg, C., and J. Gu. 2008. Pathology, molecular biology, and pathogen-
tions in effector lymphocyte recruitment to the small intestine. J. Clin. esis of avian influenza A (H5N1) infection in humans. Am. J. Pathol.
Invest. 110:10791081. http://dx.doi.org/10.1172/JCI0216946 172:11551170. http://dx.doi.org/10.2353/ajpath.2008.070791
Chervonsky, A. 2009. Innate receptors and microbes in induction of auto Kuiken,T., and J.K.Taubenberger. 2008. Pathology of human influenza revisited.
immunity. Curr. Opin. Immunol. 21:641647. http://dx.doi.org/10.1016/ Vaccine. 26:D59D66. http://dx.doi.org/10.1016/j.vaccine.2008.07.025
j.coi.2009.08.003 Laver, W.G., and R.G. Webster. 1979. Ecology of influenza viruses in lower
Crowe, C.R., K. Chen, D.A. Pociask, J.F. Alcorn, C. Krivich, R.I. Enelow, mammals and birds. Br. Med. Bull. 35:2933.
T.M. Ross, J.L. Witztum, and J.K. Kolls. 2009. Critical role of IL-17RA Leppkes, M., C. Becker, I.I. Ivanov, S. Hirth, S.Wirtz, C. Neufert, S. Pouly, A.J.
in immunopathology of influenza infection. J. Immunol. 183:53015310. Murphy, D.M. Valenzuela, G.D. Yancopoulos, et al. 2009. RORgamma-
http://dx.doi.org/10.4049/jimmunol.0900995 expressing Th17 cells induce murine chronic intestinal inflammation via
Das, G., M.M. Augustine, J. Das, K. Bottomly, P. Ray, and A. Ray. 2003. An im- redundant effects of IL-17A and IL-17F. Gastroenterology. 136:257267.
portant regulatory role for CD4+CD8 T cells in the intestinal epithelial http://dx.doi.org/10.1053/j.gastro.2008.10.018
layer in the prevention of inflammatory bowel disease. Proc. Natl. Acad. Sci. Li, C., P. Yang, Y. Sun, T. Li, C. Wang, Z. Wang, Z. Zou, Y. Yan, W. Wang, C.
USA. 100:53245329. http://dx.doi.org/10.1073/pnas.0831037100 Wang, et al. 2012. IL-17 response mediates acute lung injury induced
Dilantika, C., E.R. Sedyaningsih, M.R. Kasper, M.Agtini, E. Listiyaningsih,T.M. by the 2009 pandemic influenza A (H1N1) virus. Cell Res. 22:528538.
Uyeki, T.H. Burgess, P.J. Blair, and S.D. Putnam. 2010. Influenza virus in- http://dx.doi.org/10.1038/cr.2011.165
fection among pediatric patients reporting diarrhea and influenza-like ill- Lupp, C., M.L. Robertson, M.E. Wickham, I. Sekirov, O.L. Champion, E.C.
ness. BMC Infect. Dis. 10:3. http://dx.doi.org/10.1186/1471-2334-10-3 Gaynor, and B.B. Finlay. 2007. Host-mediated inflammation disrupts the
Esplugues, E., S. Huber, N. Gagliani, A.E. Hauser, T. Town, Y.Y. Wan, W. intestinal microbiota and promotes the overgrowth of Enterobacteriaceae.
OConnor Jr., A. Rongvaux, N. Van Rooijen, A.M. Haberman, et al. Cell Host Microbe. 2:204. http://dx.doi.org/10.1016/j.chom.2007.08.002
2011. Control of TH17 cells occurs in the small intestine. Nature. 475: Maslowski, K.M., A.T.Vieira, A. Ng, J. Kranich, F. Sierro, D.Yu, H.C. Schilter,
514518. http://dx.doi.org/10.1038/nature10228 M.S. Rolph, F. Mackay, D. Artis, et al. 2009. Regulation of inflamma-
Ferretti, S., O. Bonneau, G.R. Dubois, C.E. Jones, and A. Trifilieff. 2003. IL-17, tory responses by gut microbiota and chemoattractant receptor GPR43.
produced by lymphocytes and neutrophils, is necessary for lipopolysaccha- Nature. 461:12821286. http://dx.doi.org/10.1038/nature08530
ride-induced airway neutrophilia: IL-15 as a possible trigger. J. Immunol. Mauad, T., L.A. Hajjar, G.D. Callegari, L.F. da Silva, D. Schout, F.R. Galas,V.A.
170:21062112. http://dx.doi.org/10.4049/jimmunol.170.4.2106 Alves, D.M. Malheiros, J.O. Auler Jr., A.F. Ferreira, et al. 2010. Lung pathol-
Gallichan, W.S., R.N. Woolstencroft, T. Guarasci, M.J. McCluskie, H.L. Davis, ogy in fatal novel human influenza A (H1N1) infection. Am. J. Respir. Crit.
and K.L. Rosenthal. 2001. Intranasal immunization with CpG oligode- Care Med. 181:7279. http://dx.doi.org/10.1164/rccm.200909-1420OC
oxynucleotides as an adjuvant dramatically increases IgA and protection Mazmanian, S.K., J.L. Round, and D.L. Kasper. 2008. A microbial symbio-
against herpes simplex virus-2 in the genital tract. J. Immunol. 166:3451 sis factor prevents intestinal inflammatory disease. Nature. 453:620625.
3457. http://dx.doi.org/10.4049/jimmunol.166.5.3451 http://dx.doi.org/10.1038/nature07008
Garrett, W.S., G.M. Lord, S. Punit, G. Lugo-Villarino, S.K. Mazmanian, McDermott, M.R., and J. Bienenstock. 1979. Evidence for a common muco-
S. Ito, J.N. Glickman, and L.H. Glimcher. 2007. Communicable ulcerative sal immunologic system. I. Migration of B immunoblasts into intestinal,
colitis induced by T-bet deficiency in the innate immune system. Cell. respiratory, and genital tissues. J. Immunol. 122:18921898.
131:3345. http://dx.doi.org/10.1016/j.cell.2007.08.017 McDermott, M.R., D.A. Clark, and J. Bienenstock. 1980. Evidence for a com-
Gill, N., M. Wlodarska, and B.B. Finlay. 2010. The future of mucosal im- mon mucosal immunologic system. II. Influence of the estrous cycle on
munology: studying an integrated system-wide organ. Nat. Immunol. 11: B immunoblast migration into genital and intestinal tissues. J. Immunol.
558560. http://dx.doi.org/10.1038/ni0710-558 124:25362539.
Holmgren, J., and C. Czerkinsky. 2005. Mucosal immunity and vaccines. Nat. Monto, A.S., S. Gravenstein, M. Elliott, M. Colopy, and J. Schweinle. 2000.
Med. 11:S45S53. http://dx.doi.org/10.1038/nm1213 Clinical signs and symptoms predicting influenza infection. Arch. Intern.
Hooper, L.V., and J.I. Gordon. 2001. Commensal host-bacterial relationships Med. 160:32433247. http://dx.doi.org/10.1001/archinte.160.21.3243
in the gut. Science. 292:11151118. http://dx.doi.org/10.1126/science Murray, J.A., and A. Rubio-Tapia. 2012. Diarrhoea due to small bowel diseases.
.1058709 Best Pract.Res.Clin.Gastroenterol. 26:581600.http://dx.doi.org/10.1016/
Hooper, L.V., and A.J. Macpherson. 2010. Immune adaptations that maintain j.bpg.2012.11.013
homeostasis with the intestinal microbiota. Nat. Rev. Immunol. 10:159 Neutra, M.R., and P.A. Kozlowski. 2006. Mucosal vaccines: the promise and
169. http://dx.doi.org/10.1038/nri2710 the challenge. Nat. Rev. Immunol. 6:148158. http://dx.doi.org/10.1038/
Ichinohe, T., I.K. Pang, Y. Kumamoto, D.R. Peaper, J.H. Ho, T.S. Murray, nri1777
and A. Iwasaki. 2011. Microbiota regulates immune defense against respi Nguyen, R.N., L.S. Taylor, M. Tauschek, and R.M. Robins-Browne. 2006.
ratory tract influenza A virus infection. Proc. Natl. Acad. Sci. USA. 108: Atypical enteropathogenic Escherichia coli infection and prolonged diarrhea
53545359. http://dx.doi.org/10.1073/pnas.1019378108 in children. Emerg. Infect. Dis. 12:597603. http://dx.doi.org/10.3201/
Ivanov, I.I., K. Atarashi, N. Manel, E.L. Brodie, T. Shima, U. Karaoz, D. Wei, eid1204.051112
K.C. Goldfarb, C.A. Santee, S.V. Lynch, et al. 2009. Induction of intes- OHara, A.M., and F. Shanahan. 2006. The gut flora as a forgotten organ.
tinal Th17 cells by segmented filamentous bacteria. Cell. 139:485498. EMBO Rep. 7:688693. http://dx.doi.org/10.1038/sj.embor.7400731
http://dx.doi.org/10.1016/j.cell.2009.09.033 Ochoa, T.J., and C.A. Contreras. 2011. Enteropathogenic Escherichia coli in-
Johnston, J.A., C.M. Bacon, D.S. Finbloom, R.C. Rees, D. Kaplan, K. Shibuya, fection in children. Curr. Opin. Infect. Dis. 24:478483. http://dx.doi
J.R. Ortaldo, S. Gupta, Y.Q. Chen, J.D. Giri, et al. 1995. Tyrosine phos- .org/10.1097/QCO.0b013e32834a8b8b
phorylation and activation of STAT5, STAT3, and Janus kinases by inter- Ohoka, Y., A. Yokota, H. Takeuchi, N. Maeda, and M. Iwata. 2011. Retinoic
leukins 2 and 15. Proc. Natl. Acad. Sci. USA. 92:87058709. http://dx.doi acid-induced CCR9 expression requires transient TCR stimulation and
.org/10.1073/pnas.92.19.8705 cooperativity between NFATc2 and the retinoic acid receptor/retinoid
Kamanaka, M., S.T. Kim,Y.Y. Wan, F.S. Sutterwala, M. Lara-Tejero, J.E. Galn, X receptor complex. J. Immunol. 186:733744. http://dx.doi.org/10
E. Harhaj, and R.A. Flavell. 2006. Expression of interleukin-10 in intestinal .4049/jimmunol.1000913
lymphocytes detected by an interleukin-10 reporter knockin tiger mouse. Pandiyan, P., X.P. Yang, S.S. Saravanamuthu, L. Zheng, S. Ishihara, J.J. OShea,
Immunity. 25:941952. http://dx.doi.org/10.1016/j.immuni.2006.09.013 and M.J. Lenardo. 2012. The role of IL-15 in activating STAT5 and

JEM Vol. 211, No. 12 2409


fine-tuning IL-17A production in CD4 T lymphocytes. J. Immunol. Stenstad, H., A. Ericsson, B. Johansson-Lindbom, M. Svensson, J. Marsal, M.
189:42374246. http://dx.doi.org/10.4049/jimmunol.1201476 Mack, D. Picarella, D. Soler, G. Marquez, M. Briskin, and W.W. Agace.
Papadakis, K.A., J. Prehn, V. Nelson, L. Cheng, S.W. Binder, P.D. Ponath, D.P. 2006. Gut-associated lymphoid tissue-primed CD4+ T cells display
Andrew, and S.R.Targan. 2000.The role of thymus-expressed chemokine CCR9-dependent and -independent homing to the small intestine. Blood.
and its receptor CCR9 on lymphocytes in the regional specialization of 107:34473454. http://dx.doi.org/10.1182/blood-2005-07-2860
the mucosal immune system. J. Immunol. 165:50695076. http://dx.doi Swidsinski, A., J. Weber, V. Loening-Baucke, L.P. Hale, and H. Lochs. 2005.
.org/10.4049/jimmunol.165.9.5069 Spatial organization and composition of the mucosal flora in patients with
Polakos, N.K., J.C. Cornejo, D.A. Murray, K.O. Wright, J.J. Treanor, I.N. inflammatory bowel disease. J. Clin. Microbiol. 43:33803389. http://dx
Crispe, D.J. Topham, and R.H. Pierce. 2006. Kupffer cell-dependent .doi.org/10.1128/JCM.43.7.3380-3389.2005
hepatitis occurs during influenza infection. Am. J. Pathol. 168:1169 Wurbel, M.A., M.G. McIntire, P. Dwyer, and E. Fiebiger. 2011. CCL25/CCR9
1178, quiz :14041405. http://dx.doi.org/10.2353/ajpath.2006.050875 interactions regulate large intestinal inflammation in a murine model of
Reid, A.H.,T.G. Fanning, J.V. Hultin, and J.K.Taubenberger. 1999. Origin and acute colitis. PLoS ONE. 6:e16442. http://dx.doi.org/10.1371/journal
evolution of the 1918 Spanish influenza virus hemagglutinin gene. Proc. .pone.0016442
Natl. Acad. Sci. USA. 96:16511656. http://dx.doi.org/10.1073/pnas Yokota, A., H. Takeuchi, N. Maeda, Y. Ohoka, C. Kato, S.Y. Song, and M.
.96.4.1651 Iwata. 2009. GM-CSF and IL-4 synergistically trigger dendritic cells
Sato, S., and H. Kiyono. 2012. The mucosal immune system of the respiratory to acquire retinoic acid-producing capacity. Int. Immunol. 21:361377.
tract. Curr. Opin. Virol. 2:225232. http://dx.doi.org/10.1016/j.coviro http://dx.doi.org/10.1093/intimm/dxp003
.2012.03.009 Zaki, M.H., K.L. Boyd, P. Vogel, M.B. Kastan, M. Lamkanfi, and T.D.
Schulthess, J., B. Meresse, E. Ramiro-Puig, N. Montcuquet, S. Darche, B. Kanneganti. 2010. The NLRP3 inflammasome protects against loss of
Bgue, F. Ruemmele, C. Combadire, J.P. Di Santo, D. Buzoni-Gatel, epithelial integrity and mortality during experimental colitis. Immunity.
and N. Cerf-Bensussan. 2012. Interleukin-15-dependent NKp46+ in- 32:379391. http://dx.doi.org/10.1016/j.immuni.2010.03.003
nate lymphoid cells control intestinal inflammation by recruiting inflam- Zhou, R., H. Wei, R. Sun, and Z. Tian. 2007a. Recognition of double-stranded
matory monocytes. Immunity. 37:108121. http://dx.doi.org/10.1016/ RNA by TLR3 induces severe small intestinal injury in mice. J. Immunol.
j.immuni.2012.05.013 178:45484556. http://dx.doi.org/10.4049/jimmunol.178.7.4548
Shinde, V., C.B. Bridges, T.M. Uyeki, B. Shu, A. Balish, X. Xu, S. Zhou, R., H.Wei, R. Sun, J. Zhang, and Z.Tian. 2007b. NKG2D recognition
Lindstrom, L.V. Gubareva, V. Deyde, R.J. Garten, et al. 2009. Triple- mediates Toll-like receptor 3 signaling-induced breakdown of epithelial
reassortant swine influenza A (H1) in humans in the United States, homeostasis in the small intestines of mice. Proc. Natl. Acad. Sci. USA.
2005-2009. N. Engl. J. Med. 360:26162625. http://dx.doi.org/10.1056/ 104:75127515. http://dx.doi.org/10.1073/pnas.0700822104
NEJMoa0903812 Ziolkowska, M., A. Koc, G. Luszczykiewicz, K. Ksiezopolska-Pietrzak, E.
Sobko, T., J. Schitt, A. Ehlin, J. Lundberg, S. Montgomery, and M. Norman. Klimczak, H. Chwalinska-Sadowska, and W. Maslinski. 2000. High levels
2010. Neonatal sepsis, antibiotic therapy and later risk of asthma and al- of IL-17 in rheumatoid arthritis patients: IL-15 triggers in vitro IL-17
lergy. Paediatr. Perinat. Epidemiol. 24:8892. http://dx.doi.org/10.1111/ production via cyclosporin A-sensitive mechanism. J. Immunol. 164:2832
j.1365-3016.2009.01080.x 2838. http://dx.doi.org/10.4049/jimmunol.164.5.2832

2410 Microbiota cause intestine injury during influenza | Wang et al.


Article

Dysfunctional CD8+ T cells in hepatitis


B and C are characterized by a lack
of antigen-specific T-bet induction
Peter D. Kurktschiev,1,2 Bijan Raziorrouh,1,2 Winfried Schraut,1,2
Markus Backmund,2,3 Martin Wchtler,4 Clemens-Martin Wendtner,4
Bertram Bengsch,5 Robert Thimme,5 Gerald Denk,2 Reinhart Zachoval,2
Andrea Dick,6 Michael Spannagl,6 Jrgen Haas,7 Helmut M. Diepolder,2
Maria-Christina Jung,8 and Norbert H. Gruener1,2
1Institutefor Immunology, Ludwig-Maximilians-University, 80539 Munich, Germany
2Department of Medicine II, University Hospital Munich, 80539 Munich, Germany
3PiT Praxis im Tal, 80331 Munich, Germany
4Department of Medicine, Klinikum Schwabing, 81925 Munich, Germany
5Department of Medicine II, University Hospital Freiburg, 79106 Freiburg, Germany
6Laboratory of Immunogenetics and Molecular Diagnostics, 80539 Munich, Germany
7Division of Infection and Pathway Medicine, University of Edinburgh, Edinburgh EH16 4SB, Scotland, UK
8Leberzentrum Mnchen, 80336 Munich, Germany

The transcription factor T-bet regulates the production of interferon- and cytotoxic
molecules in effector CD8 T cells, and its expression correlates with improved control of
chronic viral infections. However, the role of T-bet in infections with differential outcome
remains poorly defined. Here, we report that high expression of T-bet in virus-specific CD8
T cells during acute hepatitis B virus (HBV) and hepatitis C virus (HCV) infection was asso-
ciated with spontaneous resolution, whereas T-bet deficiency was more characteristic of
chronic evolving infection. T-bet strongly correlated with interferon- production and
proliferation of virus-specific CD8 T cells, and its induction by antigen and IL-2 stimulation
partially restored functionality in previously dysfunctional T-betdeficient CD8 T cells.
However, restoration of a strong interferon- response required additional stimulation with
IL-12, which selectively induced the phosphorylation of STAT4 in T-bet+ CD8 T cells. The
observation that T-bet expression rendered CD8 T cells responsive to IL-12 suggests a
stepwise mechanism of T cell activation in which T-bet facilitates the recruitment of
additional transcription factors in the presence of key cytokines. These findings support a
critical role of T-bet for viral clearance and suggest T-bet deficiency as an important
mechanism behind chronic infection.

CORRESPONDENCE
Peter Kurktschiev: The transcription factor T-bet (T-box expressed observed in dysfunctional CD8 T cells of chronic
peter.kurktschiev@ HIV patients and in the murine LCMV model
in T cells; Tbx21) is a crucial regulator of T cell
med.uni-muenchen.de
immunity. It mediates the differentiation of of chronic viral infection (Kao et al., 2011;
Abbreviations used: acHCV, CD4 T cells into Th1 cells and of CD8 T cells Ribeiro-dos-Santos et al., 2012). Furthermore,
acute chronic-evolving HCV; it has been shown that T-bet and the homolo-
arHBV, acute resolving into Tc1 cells (Szabo et al., 2000; Mullen et al.,
HBV; arHCV, acute resolving 2001; Sullivan et al., 2003). In effector CD8 gous transcription factor Eomesodermin (Eomes)
HCV; cHBV, chronic HBV; T cells, T-bet is an activator of interferon- define two distinct states of virus-specific CD8
cHCV, chronic HCV; EBV, T cells and their balance plays an important
Eppstein-Barr virus; Eomes, production and correlates with increased cyto-
role in the control of chronic viral infection
Eomesodermin; Flu, Influenza toxic activity (Szabo et al., 2000; Cruz-Guilloty
A virus; HBV, hepatitis B virus; (Paley et al., 2012). Interestingly, retroviral
HCV, hepatitis C virus; PBSE,
et al., 2009). A recent study has found that T-bet
Pacific Blue succinimidyl ester; is highly expressed in HIV-specific CD8 T cells 2014 Kurktschiev et al. This article is distributed under the terms of an Attribution
rHBV, long-term resolved HBV; of HIV elite controllers who control viral load NoncommercialShare AlikeNo Mirror Sites license for the first six months
rHCV, long-term resolved after the publication date (see http://www.rupress.org/terms). After six months
HCV; STAT4, signal transducer to very low levels without therapy (Hersperger it is available under a Creative Commons License (AttributionNoncommercial
Share Alike 3.0 Unported license, as described at http://creativecommons.org/
and activator of transcription 4. et al., 2011). Correspondingly, its loss has been licenses/by-nc-sa/3.0/).

The Rockefeller University Press $30.00


J. Exp. Med. 2014 Vol. 211 No. 10 2047-2059 2047
www.jem.org/cgi/doi/10.1084/jem.20131333
overexpression of T-bet prevented CD8 T cell exhaustion in RESULTS
chronic LCMV infection, demonstrating the therapeutic po- T-bet is highly expressed during
tential of T-bet modulation (Kao et al., 2011). However, the acute resolving HBV infection
role of T-bet in human viral infections with dichotomous out- Acute HBV infection resolved spontaneously in all enrolled
come remains to be determined. Because HIV and LCMV patients. Therefore, we would expect up-regulation of T-bet
clone13 establish chronic infection in all infected subjects, in HBV-specific CD8 T cells of these individuals, in case it
other pathogens would be more suitable to dissect the differ- plays an important role in viral clearance. Ex vivo expression
ences between successful versus failing immune response dur- of T-bet was determined by intracellular flow cytometry
ing acute infection. combined with MHC-I pentamers detecting HBV core 1827
Human hepatitis B virus (HBV) and hepatitis C virus (c18-27)specific CD8 T cells, which react with a major
(HCV) infection can both either resolve spontaneously or immunodominant epitope in MHC-I A0201 background.
establish chronic infection. Virus-specific CD8 T cells play a The frequencies of virus-specific CD8 T cells are shown
causal role in the clearance of both infections, as demonstrated in Table S1.
by in vivo CD8 T cell depletion in the chimpanzee model When we compared T-bet expression in patients with
where all subjects challenged with HBV or HCV developed acute resolving HBV (arHBV), chronic HBV (cHBV) and
chronic infection (Shoukry et al., 2003; Thimme et al., 2003). resolved HBV (rHBV) infection, we found a significantly
In chronic HBV and HCV infection, virus-specific CD8 higher mean percentage of T-bet+ c18-27specific CD8
T cells gradually lose their effector functions and become in- T cells during arHBV (57.9%) compared with cHBV (10%)
creasingly dysfunctional (Lechner et al., 2000a; Gruener et al., and rHBV (0.7%; Fig. 1, A and C). Our subanalysis of anti-
2001; Boni et al., 2007). One hallmark of severe dysfunction HBe+ and anti-HBe cHBV patients showed no significant
is the lack of antigen-specific interferon- production by difference in T-bet expression (unpublished data). In healthy
T cells (Lechner et al., 2000b). Although the molecular mech- controls T-bet+ CD8 T cells comprised 10.3% (mean) of
anisms behind T cell dysfunction are the focus of intensive total CD8 T cells (unpublished data). Elevated T-bet expres-
research (Bowen et al., 2004; von Hahn et al., 2007; Wherry, sion during arHBV was confirmed for 2 additional HBV-
2011) it is yet unknown how far impaired regulation of specific epitopes (HBV envelope 183191 and HBV polymerase
T-bet might be involved in the development of chronic HBV 573581; Fig. 1, D and E). To rule out nonspecific bystander
and HCV infection. up-regulation of T-bet in CD8 T cells, we determined its
In this study, we determined the expression of T-bet expression in non-HBV-specific CD8 T cells during acute
in virus-specific CD8 T cells during acute HBV and HCV HBV infection. We chose EBV-specific CD8 T cells because
infection and examined its correlation with the clinical they are broadly detectable. There was no significant increase
outcome. T-bet was highly expressed in spontaneously re- in T-bet+ EBV-specific CD8 T cells (mean 10.2%; unpub-
solving but deficient in chronic-evolving infection. When lished data). As a proof of principle, we determined antigen-
we further characterized the functional correlates behind specific T-bet up-regulation in EBV-specific CD8 T cells
these differential expression patterns, we found a strong as- (EBV BMLF-1 259267) during acute (aEBV) and latent
sociation of T-bet with antigen-specific proliferation and persisting (pEBV) EBV infection. 62.7% (mean) of EBV-
interferon- production by virus-specific CD8 T cells. In- specific CD8 T cells were T-bet+ in aEBV, whereas only
duction of T-bet by antigen or IL-2 recovered antigen- 10.2% were T-bet+ in pEBV. Additionally, we found a mean
specific proliferation but was not sufficient to restore of 7.5% T-bet+ CD8 T cells specific for Influenza A (Flu; In-
interferon- expression. However, restoration of a strong fluenza A MP 5866) in healthy controls with previously re-
interferon- response in previously dysfunctional CD8 solved infection, which served as control for a memory CD8
T cells was achieved by additional stimulation with IL-12, T cell response (Fig. 1, A and C).
which selectively induced phosphorylation of STAT4
(pSTAT4) in T-bet+ CD8 T cells. This is consistent with Strong T-bet expression in acute HCV infection
previous findings that T-bet and STAT4 cooperate in the correlates with spontaneous resolution
transcriptional control of interferon- (Thieu et al., 2008). Whereas acute HBV infection in adults is almost universally
The observation that T-bet rendered CD8 T cells suscep- cleared, acute HCV infection becomes chronic in the major-
tible to IL-12 suggests a stepwise mechanism of T cell acti- ity of cases (Wright and Lau, 1993; Lauer and Walker, 2001).
vation in which T-bet facilitates the recruitment of additional In our study, 50% of the patients with acute HCV developed
transcription factors in the presence of key cytokines, and chronic infection. HCV-specific CD8 T cells were detected
thus contributes to the adjustment of an appropriate by HCV-specific pentamers covering several epitopes with
T cell response. different MHC-I backgrounds. T-bet expression was deter-
These findings indicate a critical role of T-bet for a suc- mined during the earliest available time points of acute HCV
cessful CD8 T cell response against HBV and HCV infection infection that was either cleared (arHCV) or became chronic
and suggest that impaired induction of T-bet could be an im- (acHCV) later on. All analyzed patients were viremic at this
portant mechanism involved in CD8 T cell dysfunction dur- time. Consistent with our results found in arHBV infec-
ing chronic viral infections. tion, frequencies of T-bet+ HCV-specific CD8 T cells were

2048 Dysfunctional CD8+ T cells in HBV/HCV lack T-bet | Kurktschiev et al.


Ar ticle

Figure 1. HBV- and HCV-specific CD8


T cells express high amounts of T-bet dur-
ing acute spontaneously resolving infec-
tion. PBMCs were isolated from (A) patients
with arHBV, cHBV, rHBV (top), aEBV, pEBV, or
resolved Flu infection (bottom) and (B) pa-
tients with arHCV (top left), rHCV (top right),
acHCV (bottom left), or cHCV infection (bot-
tom right) and analyzed by flow cytometry.
The outlined areas indicate the population of
pentamer+ CD8 T cells and the numbers indi-
cate the percentage of T-bet+ (above) and
T-bet (below) cells among total pentamer+
CD8 T cells. (C) Quantification of ex vivo T-bet
expression in virus-specific CD8 T cells of the
subjects described in A and B. Bars represent
the mean percentage of T-bet+ pentamer+
CD8 T cells among total pentamer+ CD8 T cells
of patients with arHBV (n = 19), cHBV (n = 24),
rHBV (n = 5), arHCV (n = 7), acHCV (n = 7),
cHCV (n = 7), rHCV (n = 7), aEBV (n = 3), pEBV
(n = 6), and Flu (n = 6). Error bars indicate the
SEM. *, P < 0.05; ***, P < 0.001 (Mann-Whitney-
U test). (D) Representative ex-vivo flow
cytometry plots showing expression of T-bet
in HBV envelope (183191)specific (left) and
HBV polymerase (573581)specific CD8
T cells (right) of patients with arHBV. The
numbers indicate the percentage of T-bet+/
and pentamer+/ CD8 T cells among total CD8
T cells. (E) Scatter dot plot showing the percent-
age of T-bet+ envelope (183191)specific
(filled circles; n = 5) or polymerase (573581)-
specific (open circles; n = 5) CD8 T cells among
total pentamer+ CD8 T cells of patients with
arHBV. The data are representative of one
experiment due to limited patient material.

significantly higher in patients with spontaneous resolution T cells we performed longitudinal measurements of T-bet
of HCV infection (arHCV; mean, 66.3%) compared with expression in 5 patients with arHBV, 4 patients with acHCV,
patients with chronic-evolving infection (acHCV; mean, 14%; and 4 patients with arHCV. This kinetics profile contained
Fig. 1, B and C). The frequencies of T-bet+ HCV-specific data from 3 different time points within 6 mo after symptom
CD8 T cells in long-term chronic (cHCV; mean, 4.5%) and onset. The first time point (t1) was defined as the earliest
long-term resolved (rHCV; mean, 6.9%) HCV infection were available patient sample obtained within 1 mo after onset of
comparable to those found in cHBV and rHBV. acute symptoms. The following analyzed time points were
13 mo (t2) and 36 mo (t3) after symptom onset, respec-
T-bet deficiency in early acute HCV tively. In arHBV, we observed a high percentage of T-bet+
precedes chronic-evolving infection c18-27specific CD8 T cells at t1 (mean, 73.1%), which
To characterize the duration of T-bet up-regulation and gradually decreased to 40.3% at t2 and 21.8% at t3 (Fig. 2,
fluctuations in its expression levels in virus-specific CD8 left). In arHCV infection, we found a similar pattern as seen

JEM Vol. 211, No. 10 2049


Figure 2. T-bet expression is lost early in acute chronic-evolving HCV infection. Percentage of T-bet+ pentamer+ CD8 T cells among total pen-
tamer+ CD8 T cells of patients with arHBV (n = 5; left), arHCV (n = 4; middle), and acHCV (n = 4; right) was determined by ex vivo flow cytometry at the
indicated time points. Data show the mean percentages and are representative of one experiment due to limited patient material. Error bars indicate SEM.

in HBV-specific CD8 T cells during arHBV (mean t1 = 67.1%; acHCV (10.5%), and cHCV (8.7%). In contrast, T-bet+/
t2 = 54.5%; and t3 = 29.2%) with high initial expression of Eomes cells, which comprised the majority of virus-specific
T-bet in HCV-specific CD8 T cells, which slowly decreased CD8 T cells, were significantly more frequent during
over the 6 mo of follow up (Fig. 2, middle). In contrast, in arHBV (mean, 38.5%) and arHCV (54.6%) as compared
acHCV T-bet expression was low right from the beginning with cHBV (9.2%), acHCV (11.7%), and cHCV (13.4%).
(mean: t1 = 14.8%; t2 = 12.6%, and t3 = 11.9%; Fig. 2, right). Interestingly, we consistently observed a population of Eomes+/
The 4 patients with acHCV were further differentiated into T-bet+ cells, which were significantly increased in arHBV
partial (n = 3) versus no controllers of viremia (n = 1) accord- (19.4%, mean) and arHCV (11.7%) compared with cHBV
ing to the evolution of viral load between t1 and t3 (Table 1). (0.8%), acHCV (2.3%), and cHCV (1.1%).
No correlation was found between T-bet or Eomes expres-
sion and quality of viral control. However, studies on larger T-bet and PD-1 are coexpressed during acute
cohorts would be required to confirm this observation. but not in chronic HBV or HCV infection
PD-1 is the hallmark inhibitory receptor found on exhausted
Eomes and T-bet can be coexpressed and define CD8 T cells. As recent publications have shown that T-bet can
different subsets of virus-specific CD8 T cells suppress PD-1 we examined the coexpression of both factors in
As recent literature has reported that Eomes, which bears HBV and HCV infection ex vivo. T-bet and PD-1 are highly
strong homology with T-bet, can compensate for T-bet de- coexpressed in virus-specific CD8 T cells during resolving
ficiency in CD8 T cells of T-bet/ mice, we investigated arHBV (mean 51.2%) and arHCV (45%), whereas the frequen-
if Eomes is associated with a successful immune response in cies of T-bet+/PD-1+ virus-specific CD8 T cells were signifi-
human HBV and HCV infection as well (Intlekofer et al., cantly lower in cHBV (11%), acHCV (7.6%), and cHCV
2005). Furthermore, one study has demonstrated that the (0.7%; Fig. 3, B and E). In contrast, the mean percentage of
balance of two subsets of CD8 T cells, T-bet (high) and PD-1+/T-bet virus-specific CD8 T cells was highest in cHBV
Eomes (high), respectively, is important for the control of (65.7%) and cHCV (50.9%) as compared with acute arHBV
murine LCMV and chronic HCV infection (Paley et al., (37.7%), arHCV (19.2%), and acHCV (19%) infection.
2012). Therefore, we examined the co-expression of T-bet
and Eomes in virus-specific CD8 T cells under several con-
Mutually exclusive expression of T-bet and CD127
ditions (Fig. 3, A and D). We found that Eomes+/T-bet
In the murine LCMV infection model, it has been shown
CD8 T cells did not show any significant difference be-
that T-bet promotes the differentiation of effector and effector
tween arHBV (10%, mean), cHBV (8.9%), arHCV (3%),
memory CD8 T cells at the cost of centralmemory cells by
repression of IL-7R (CD127; Intlekofer et al., 2007). We
Table 1. Kinetics of viral load in acute chronic-evolving HCV investigated if T-bet can repress CD127 expression on virus-
infection specific CD8 T cells during HBV and HCV infection, as this
Patient t1 viral load [IU/ml] t3 viral load [IU/ml] could affect the generation of memory CD8 T cells. T-bet+/
P1 1,300 14
CD127+ virus-specific CD8 T cells were rare (means: arHBV,
P2 2,980 15
3.7%; cHBV, 7.7%; arHCV, 4.9%; acHCV, 3.1%; and cHCV,
5.9%). The T-bet+/CD127 population showed higher fre-
P3 110,000 150
quencies in resolving (means: arHBV, 61.8%; arHCV, 48.9%)
P4 66,000 9,350,000
as compared with chronic-evolving infections (cHBV, 5.8%;

2050 Dysfunctional CD8+ T cells in HBV/HCV lack T-bet | Kurktschiev et al.


Ar ticle

Figure 3. Expression of T-bet is associated with distinct phenotypes of virus-specific CD8 T cells. Ex vivo expression of T-bet, Eomes, PD-1, and
CD127 in PBMCs from patients with acute HBV or HCV infection was analyzed by flow cytometry on the earliest available samples obtained within 3 wk
of acute symptom onset. The data on cHBV and cHCV patients were obtained at any time point during chronic infection. (A) Contour plots show ex vivo
coexpression of T-bet and Eomes in virus-specific CD8 T cells of patients with arHBV, cHBV, arHCV, acHCV, and cHCV infection. The numbers indicate the
percentage of T-bet+/ and Eomes+/ CD8 T cells among pentamer+ CD8 T cells. (B) Representative contour plots of T-bet and PD-1 coexpression as
described in (A). (C) Representative contour plots of T-bet and CD127 coexpression as described in A. (D) Mean percentage of pentamer+ CD8 T cells with
a T-bet+/Eomes, T-bet/Eomes+, and T-bet+/Eomes+ phenotype as determined by flow cytometry. Data were obtained from arHBV (n = 19), cHBV (n = 24),
arHCV (n = 7), acHCV (n = 7), and cHCV (n = 7) patients. (E) Mean percentage of pentamer+ CD8 T cells with T-bet+/ PD-1+ and T-bet/ PD-1+ phenotype
found in the patients described in D. (F) Mean percentage of T-bet+/ CD127+, T-bet/ CD127+, and T-bet+/CD127 analogous to D. All error bars indicate
SEM. ***, P < 0.001 (Mann-Whitney-U test). Data are representative of one experiment due to limited patient material.

JEM Vol. 211, No. 10 2051


acHCV, 13.5%; cHCV, 10.8%). On the contrary, T-bet/ literature (Lighvani et al., 2001; Ylikoski et al., 2005) could in-
CD127+ cells were more frequent in chronic-evolving crease T-bet and interferon- expression in virus-specific CD8
(means: cHBV, 69.8%; acHCV, 29.5%; cHCV, 50.4%) than T cells of chronic HBV patients. We tested several cytokines in
in resolving infection (arHBV, 4.8%; arHCV, 10.4%; Fig. 3, different concentrations (see Materials and methods) for their
C and F). potential to induce T-bet in CD8 T cells (Fig. 4 B). The only
cytokine that significantly induced T-bet was IL-2. Therefore,
Induction of T-bet by IL-2 facilitates antigen- we wanted to examine if T-bet induction by IL-2 was directly
specific interferon- production in HBV-specific coupled with interferon- production. We found a significant
CD8 T cells in cooperation with IL-12 but weak increase of T-bet+/interferon-+ CD8 T cells after
To examine the effects of T-bet on CD8 T cell functionality, stimulation with IL-2+c18-27 (mean, 0.154%) compared with
we compared its coexpression with interferon- and perforin stimulation with c18-27 antigen alone (mean, 0.012%) or the
in CD8 T cells of healthy controls, which were nonspecifically control (mean, 0.008%). However, we observed that stimula-
activated by TCR-stimulation with CD3+CD28. The vast tion with IL-2+IL-12+c18-27 resulted in a much stronger
majority of interferon- and perforin-producing stimulated increase of T-bet+/interferon-+ CD8 T cells (mean 1.981%),
CD8 T cells were T-bet+ (unpublished data). Next, we ad- whereas IL-12+c18-27 was not able to induce the same effect
dressed the question of whether specific stimulation with anti- in the absence of IL-2 (0.014%). IL-2+IL-12 induced consider-
gen (c18-27) or cytokines described as T-bet inducers in able background stimulation (1.191%; Fig. 4, A and C).

Figure 4. Antigen-specific interferon- production correlates with T-bet and can be restored by IL-2+IL-12 co-stimulation. PBMCs of pa-
tients with cHBV (n = 11) were cultured for 3 d in culture medium (control) containing either HBV-c18-27 antigen (Ag), c18-27 antigen+IL-2 (Ag+IL-2),
c18-27 antigen+IL-12 (Ag+IL-12), IL-2+IL-12, c18-27 antigen+IL-2+IL-12 (Ag+IL-2+IL-12), or CD3+CD28. On day 3 antigen-treated groups were
restimulated with antigen and all groups were incubated for 6 h in the presence of Brefeldin A before intracellular flow cytometry was performed.
(A) Expression of T-bet and interferon- by CD8 T cells. The numbers indicate the percentage of T-bet+/ and interferon-+/ CD8 T cells among total CD8
T cells. (B) Mean induction of T-bet by treatment with the respective cytokines. Induction was defined as the percentage of T-bet+ CD8 T cells after stimu-
lation subtracted by the percentage of T-bet+ CD8 T cells in unstimulated controls. Error bars represent the SEM. (C) Mean percentage of T-bet+ interferon-+
CD8 T cells after stimulation. Error bars indicate the SEM. Data are representative of one experiment due to limited patient material. ***, P < 0.001 (Mann-
Whitney-U test).

2052 Dysfunctional CD8+ T cells in HBV/HCV lack T-bet | Kurktschiev et al.


Ar ticle

High expression of T-bet is associated importance of T-bet in HIV and LCMV clone 13 that univer-
with strong antigen-specific proliferation sally establish chronic infection, we assessed its role in human
Antigen-specific proliferation of CD8 T cells is key to the gen- HBV and HCV infection that allowed us the direct compari-
eration of sufficient amounts of effector cells for control of the son of successful versus failing CD8 T cell response against
virus.We analyzed the correlation between T-bet expression and the same pathogen (Hersperger et al., 2011; Kao et al., 2011;
expansion of epitope-specific CD8 T cells upon stimulation. Ribeiro-dos-Santos et al., 2012). Our most important finding
PBMCs of patients with chronic HBV were stimulated with is that expression of T-bet in virus-specific CD8 T cells was
either antigen c18-27 and/or cytokines IL-2 and IL-12 for 7 d strongly associated with clearance of acute HCV infection.
as described in the Materials and methods. The expansion of HCV-specific CD8 T cells in acute chronic-evolving HCV
c18-27specific CD8 T cells and the correlation between T-bet infection, though broadly detectable, failed to clear HCV in-
expression and proliferation was determined by flow cytometry fection and were deficient in T-bet expression. In acute HBV
with the proliferation marker Pacific Blue succinimidyl ester infection, increased expression levels of T-bet were observed
(PBSE).The strongest expansion of c18-27specific CD8 T cells in patients who later resolved the disease, whereas expression
was observed in the groups stimulated with antigen c18-27 was lost in dysfunctional CD8 T cells in spite of viral persis-
(mean, 0.7% of total CD8 T cells), IL-2+c18-27 (mean, 1.83%), tence during long-term chronic infection. As acute HBV in-
and IL-2+IL-12+c18-27 (mean, 1.93%), which was a significant fection rarely takes chronic course in adults, we were not able
induction compared with controls (mean, 0.1%). Of note, IL-12 to confirm T-bet deficiency in HBV-specific CD8 T cells of
did not significantly increase proliferation when added to such patients.Therefore, further studies will be required to es-
IL-2+Ag. PBMCs stimulated with IL-2 (mean, 0.1%), IL-12 tablish a definitive association between T-bet expression and
(mean, 0.09%), or IL-2+IL-12 (mean, 0.15%) showed no increased clinical course of HBV infection. All patients who later con-
frequencies of virus-specific CD8 T cells (Fig. 5,A and B). Next, trolled viral replication had high expression of T-bet in virus-
we divided proliferating PBSE- and c18-27specific CD8 specific CD8 T cells, and we confirmed the same pattern in
T cells in a T-bet+ and a T-bet subset to determine if T-bet was acute and latent persisting EBV infection. These data provide
preferentially expressed in proliferating cells. We found signifi- further evidence for a central role of T-bet for a successful
cantly higher frequencies of T-bet+ CD8 T cells in the groups virus-specific CD8 T cell response in self-limiting human
with strong proliferation, whereas T-bet CD8 T cells showed viral infections whereas T-bet deficiency was associated with
low frequencies (Ag mean: T-bet+, 0.29%; T-bet, 0.09%; chronic infection. Several studies have provided insights into
IL-2+Ag T-bet+, 1.12%; T-bet, 0.07%; and IL-2+IL12+Ag the mechanisms behind T-bet deficiency in chronic infec-
T-bet+, 1.04%;T-bet, 0.05%; Fig. 5, C and D). Interestingly, we tions. In the murine LCMV infection model, antigen persis-
observed strong induction of T-bet+ virus-specific CD8 T cells tence and high viral load lead to reduced T-bet levels in CD8
in the groups stimulated with antigen c18-27 (mean, 82.6% of T cells, possibly by impaired T cell receptor signaling (Kao
specific cells) or IL-2 (mean, 56.2%) compared with IL-12 et al., 2011). Direct inhibition of CD8 T cell signaling by in-
(mean, 12.5%), and control (mean, 8.5%; unpublished data). teraction with viral proteins can occur as well. For example,
HCV core protein inhibits proliferation and interferon- pro-
IL-12 selectively induces phosphorylation duction of HCV-specific T cells by blocking their C1q com-
of STAT4 in T-bet+ CD8 T cells plement receptor (Kittlesen et al., 2000). Generation of escape
We sought to determine the mechanism behind the much mutations can also lead to reduced T cell receptor stimulation
stronger induction of interferon- after additional stimulation (Chang et al., 1997). Interestingly, we did not observe ex vivo
with IL-12. STAT4 is of crucial importance for signal trans- expression of T-bet and Eomes on HCV-specific CD4 T cells
duction by the IL-12 receptor and is known to cooperate with of patients with acute HCV infection, which warrants further
T-bet in the regulation of the interferon- gene (Thieu et al., investigation (Fig. S1). Of note, T-bet was readily inducible in
2008).Therefore, we investigated if phosphorylation of STAT4 CD4 T cells under Th1 culture conditions.
might be a possible explanation. PBMCs were either pretreated Our longitudinal measurements demonstrate that T-bet
with IL-2 to induce T-bet or left unstimulated as control. On expression levels are stable over longer time spans and are not
day 3, cells were restimulated with either IL-12 which should affected by rapid fluctuations. In acute resolving HBV and
induce STAT4 phosphorylation or IL-2 as negative control. HCV infection, the highest expression of T-bet was observed
We found that IL-12 preferentially induced pSTAT4 in T-bet+ at the earliest available time points and remained elevated
CD8 T cells (mean, 2.6%) as compared with T-bet cells compared with controls for at least 46 mo. In acute chronic-
(0.38%; Fig. 6, A and B). IL-2 restimulation minimally induced evolving HCV infection, however, T-bet remained low at all
pSTAT4 in T-bet+ (0.3%) and T-bet CD8 T cells (0.34%) analyzed time points. Although very early loss of previously
compared with controls (T-bet+, 0.03%; T-bet, 0.05%). induced T-bet might be one explanation, our observations of
T-bet kinetics in resolving infection suggest that this elevation
DISCUSSION should be detectable for several months. As this was not the
In this study, we investigated in how far T-bet is involved in case, an alternative explanation could be that T-bet is only
the early events during acute infection, which are critical for weakly induced and thus lost at earlier time points or is not
viral clearance. Although previous studies have discovered the induced at all.

JEM Vol. 211, No. 10 2053


Figure 5. Induction of T-bet by IL-2 and antigen is associated with antigen-specific proliferation. PBMCs of patients with cHBV (n = 10) were
labeled with the proliferation marker PBSE and cultured for 7 d in culture medium in the presence or absence of HBV-c18-27 antigen (Ag), IL-2, or IL-12.
Antigen c18-27 was added on day 0, and cytokines were administered on day 4 in the designated groups. Negative controls were cultured in medium
without further supplements, while positive controls were stimulated with CD3+CD28. On day 7, cells were stained for flow cytometry. (A) Frequencies of
pentamer+ CD8 T cells among total CD8 T cells and their PBSE labeling intensity. (B) Expression of T-bet and the frequency of pentamer+ CD8 T cells after
stimulation. (C) Mean percentage of c18-27specific CD8 T cells among total CD8 T cells. (D) Mean percentage of PBSE/T-bet (white) and mean per-
centage of PBSE/T-bet+ specific CD8 T cells (black) among total CD8 T cells. Error bars represent the SEM. Data are representative of one experiment due
to limited patient material. *, P < 0.05; **, P < 0.01; ***, P < 0.001 (Mann-Whitney-U test).

Eomes can trigger antigen-specific interferon- pro- that play an important role in the control of chronic viral infec-
duction in CD8+ T cells of T-bet/ mice and, together with tion (Pearce et al., 2003; Paley et al., 2012). As Eomes might
T-bet, it defines different subsets of virus-specific CD8 T cells compensate for the lack of T-bet in HBV and HCV infection,

2054 Dysfunctional CD8+ T cells in HBV/HCV lack T-bet | Kurktschiev et al.


Ar ticle

Figure 6. IL-12 selectively induces STAT4


phosphorylation in T-bet+ CD8 T cells. PBMCs
of patients with cHBV (n = 7) were cultured for 3 d
with either IL-2 or medium as control. On day 3
cells were restimulated for 20 min with either
IL-2 or IL-12. Controls were left unstimulated.
Cells were then analyzed by flow cytometry.
(A) Coexpression of T-bet and pSTAT4 in CD8
T cells of an unstimulated control (left), PBMCs
cultured with IL-2 and restimulated with IL-2
(middle), and PBMCs cultured in IL-2 and restim-
ulated with IL-12 (right). Representative plots
demonstrate the percentage of T-bet+/ and
pSTAT4+/ CD8 T cells among total CD8 T cells.
(B) Mean frequency of T-bet pSTAT4+ (white)
and mean frequency of T-bet+ pSTAT4+ (black)
CD8 T cells among total CD8 T cells after stimula-
tion with the respective cytokines. Error bars rep-
resent the SEM. Data are representative of one
experiment due to limited patient material.
**, P < 0.01 (Mann-Whitney-U test).

we further analyzed its coexpression with T-bet in virus-specific However, as T-bet was not expressed at later stages of infec-
CD8 T cells under different conditions. In contrast to the pre- tion in our analyzed samples we could not assess possible sup-
dominant T-bet+ Eomes population, which was significantly pressive effects on PD-1 ex vivo.
more frequent in acute resolving versus chronic-evolving in- A successful immune response also depends on the bal-
fection and correlated with viral clearance, we did not observe anced differentiation of CD8 T cells into terminally differen-
any up-regulation of Eomes+ T-bet CD8 T cells in acute tiated effector CD8 T cells and self-renewing centralmemory
resolving infection. Of note, this was true for patients with CD8 T cells. Former studies demonstrated that T-bet drives
cHBV and cHCV as well, which differs from previous studies effector CD8 T cell differentiation at the expense of central
that have suggested up-regulation of Eomes in exhausted cells memory cells. Our finding that T-bet and CD127 are ex-
of patients with chronic infection (Paley et al., 2012). This pressed in a mutually exclusive way fits into this hypothesis.
might be explained by the fact that we focused on HBV- It remains to be determined how far T-bet expression during
and HCV-specific peripheral blood lymphocytes, whereas the acute infection could impair the differentiation of memory
cited study analyzed HCV-specific intrahepatic lymphocytes CD8 T cells. Of note, T-bet was down-regulated in mem-
or LCMV-specific lymphocytes in murine infection. Further- ory CD8 T cells of patients with resolved HBV, HCV, and
more, a subset of T-bet+ Eomes+ cells was consistently detect- Flu infection. This could be explained by a lack of TCR
able and showed significantly elevated frequencies in resolving stimulation after antigen elimination. Another possible expla-
versus chronic-evolving infection, as described for the T-bet+ nation is that T-bet+ CD8 T cells are short-lived effector cells
Eomes subset. The finding that Eomes was preferentially ex- and are lost over time (Intlekofer et al., 2005; Joshi et al.,
pressed in T-bet+ CD8 T cells supports the notion of a domi- 2007). In summary, the factor that shows the strongest corre-
nant role of T-bet for a functional CD8 T cell response during lation with viral clearance is expression of T-bet. Although
acute infection. significant, the correlation of T-bet+Eomes+ or T-bet+PD-1+
One recent study has shown that the expression of PD-1, HCV-specific CD8 T cells and viral clearance was less pro-
the hallmark inhibitory receptor of exhausted CD8 T cells, nounced. Interestingly, CD127 expression on virus-specific
can be repressed by T-bet in chronic murine LCMV infec- CD8 T cells during acute HCV infection has shown a nega-
tion. Retroviral overexpression of T-bet in CD8 T cells led tive correlation with viral clearance.
to a sustained antiviral CD8 T cell response and down-regulation Because our data suggested that the association of T-bet
of several inhibitory surface receptors (Kao et al., 2011). In with clearance of infection is not just a secondary phenomenon
another study, induction of T-bet by third signal cytokines but causally involved in the mechanisms behind a successful
decreases PD-1 levels in chronic HBV infection and restored immune response, we further investigated the effects of T-bet on
interferon- production (Schurich et al., 2013). We found proliferation and interferon- production of virus-specific CD8
that T-bet and PD-1 are highly coexpressed in acute infec- T cells.We observed that virtually all interferon-producing
tion but later on T-bet is lost while PD-1 levels remain high. CD8 T cells expressed T-bet as well, whereas T-betexpressing
The lack of T-bet might facilitate the high expression of CD8 T cells did not necessarily produce interferon-. We
PD-1 on dysfunctional CD8 T cells during chronic infection. tested several cytokines and cytokine combinations, including

JEM Vol. 211, No. 10 2055


interferon-, interferon-, IL-2, and IL-12, which have been result from impairment at different levels of this regulatory net-
used for induction of T-bet in recent literature and observed work during its adjustment. Loss of HCV-specific CD4 T cell
the most striking effect on T-bet induction when stimulating responses and IL-2 production as consistently observed in
with IL-2 (Lighvani et al., 2001; Ylikoski et al., 2005). Sub patients developing chronic infection is one mechanism that
sequent experiments revealed a similar effect by stimulation with could interfere with proper induction of T-bet in CD8 T cells
antigen, which had an additive effect when combined with (Diepolder et al., 1995; Gerlach et al., 1999; Urbani et al.,
IL-2. It was surprising that antigen-stimulated proliferation in 2006). Furthermore, several studies have reported that HCV
exhausted CD8 T cells. However, it has been previously core protein disturbs APC maturation during acute HCV infec-
shown that exhausted T cells can proliferate extensively upon tion, leading to decreased levels of IL-12, which could contrib-
antigen exposure without producing cytokines (Shin et al., ute to loss of interferon- production (Auffermann-Gretzinger
2007). This fits with our experience that T cells require anti- et al., 2001; Eisen-Vandervelde et al., 2004).
gen for in vitro expansion. Although IL-2 plays an important Our data provide for the first time evidence for the cen-
role for in vitro expansion, our results suggest that antigen itself tral role of T-bet in self-limiting human viral infections and
can trigger proliferation to some extent, too. We used low for deficient T-bet induction in virus-specific CD8 T cells as
doses of antigen because higher doses can induce anergy. In a mechanism associated with viral persistence. T-bet induc-
addition, antigen concentration was kept constant. This is dif- tion by IL-2 and co-stimulation with IL-12 restored function
ferent from the in vivo setting, where antigen levels are fluc in previously exhausted virus-specific CD8 T cells and could
tuating and can reach much higher levels as used in our be a potential target for future therapies.
experiments. Induction of T-bet by IL-2 and/or antigen
strongly induced proliferation of HBV-specific CD8 T cells MATERIALS AND METHODS
but did not induce interferon-. Restoration of a strong inter- Study subjects. Peripheral blood was obtained from patients and controls at
the University Hospital Munich. We examined HBV-specific CD8 T cell
feron- response required additional stimulation with IL-12,
responses in patients with MHC-I A0201 background and arHBV, cHBV, or
which could be explained by the finding that T-bet can induce rHBV infection. The HCV-specific CD8 T cell responses were analyzed in
the IL-12R2 receptor, and thus make T cells more suscepti- patients with arHCV, acHCV, cHCV, and rHCV HCV infection. HCV pa-
ble to IL-12 (Liao et al., 2011). We sought to clarify the mech- tients had MHC-I backgrounds A0101, A0201, A0301, B0701, or B3501. All
anism behind the enhanced IFN- induction in IL-12stimulated HCV genotypes were included. Patients with acute EBV infection and
CD8 T cells and examined if STAT4, which is known as spe- healthy subjects served as controls.The patient characteristics are summarized
in Table 2.
cific signal transducer of the IL-12 receptor, might be involved.
Interestingly, we found that IL-12 selectively induced STAT4 Ethics statement. This study was conducted in conformity with the ethical
phosphorylation in T-bet+ CD8 T cells, which is consistent guidelines of the Declaration of Helsinki. Written informed consent was
with previous studies that describe cooperation of T-bet and obtained from all patients. Approval for this study was obtained from the In-
pSTAT4 in the induction of IFN- (Thieu et al., 2008). IL-12 stitutional Review Board of the medical faculty of the Ludwig-Maximilians-
single treatment induced neither T-bet up-regulation nor University (Munich).
interferon- secretion or proliferation. We observed background
Diagnostic criteria. Acute hepatitis was defined as acute onset of nonspe-
stimulation of CD8 T cells in the group stimulated with
cific Flu-like symptoms and jaundice in previously healthy persons with peak
IL-2+IL-12 but without antigen, which can be explained in part GPT elevation 10 times above the upper limit of normal. Acute HBV was
by TCR-independent reactivation of CD8 T cells by cyto- confirmed by concomitant detection of HBsAg, HBV-DNA, or anti-HBc-
kines, as described for memory CD8 T cells (Rau et al., IgM and acute HCV by detection of HCV-RNA or seroconversion of anti-
2013). One recent study has found that IL-12 induces T-bet HCV. Other possible causes of acute hepatitis, like autoimmune hepatitis,
and interferon- in CD8 T cells of patients with chronic alcoholic liver disease, or toxins were excluded. Resolution of acute hepatitis
B was confirmed by seroconversion of anti-HBs. Acute-resolving HCV was
HBV infection (Schurich et al., 2013). However, as IL-2 was
defined as spontaneous loss of initially detectable HCV-RNA, which re-
universally added to cell culture medium in the cited study, mained negative for at least 12 mo. In chronic-evolving acute HCV infec-
considering our observations, we suggest that the observed tion, HCV-RNA remained detectable for longer than 6 mo after symptom
effects occur only under combined treatment with both cyto- onset. Resolved HCV was defined as a previously cleared HCV infection
kines. This is important, as the synergistic effects of both cy- without detection of HCV-RNA for at least 12 mo before enrollment. Di-
tokines allowed us a 200-fold reduction of the effective IL-12 agnosis of chronic HCV infection was based on elevated serum GPT levels
for at least 6 mo and the consistent detection of HCV-RNA. Chronic HBV
dose, thus making it more approachable for potential clinical was defined by detection of HBV-DNA or HBsAg for more than 6 mo.
trials. Although previous trials with single cytokine treat- Acute EBV was diagnosed by acute clinical symptoms, typical hematologic
ment of chronic HBV and HCV infection found no signifi- findings, and detection of EBV-DNA.
cant improvement of the clinical outcome (Pardo et al., 1997;
Artillo et al., 1998; Zeuzem et al., 1999; Carreo et al., 2000), Isolation of PBMCs. Human PBMCs were isolated from heparinized
a combination treatment with reduced cytokine doses might blood by Ficoll-Paque density-gradient centrifugation as described earlier
and were either analyzed directly or cryopreserved (Perlmann et al., 1976).
be more promising.
Viral clearance requires a balanced interplay of virus- HLA typing. DNA was extracted from PBMCs with the QIAamp DNA
specific CD4 T cells, CD8 T cells and APC. The various pat- Blood Mini kit (QIAGEN) following the manufacturers instructions. HLA
terns of viral replication seen during acute infection could typing was performed as described previously (Witt et al., 2002).

2056 Dysfunctional CD8+ T cells in HBV/HCV lack T-bet | Kurktschiev et al.


Ar ticle

Table 2. Patient characteristics


Group n Female /male Age (mean) GPT(U/l; mean) Viral load (mean) HCV GT1 Anti-HBe n.d.
arHBV 19 6/13 38.4 2,567 19.6 cop/ml
106 - 8 6 5
cHBV 24 7/17 44.6 59.5 18.3 103 cop/ml - 13 5 6
aHCV 14 6/8 53.5 984 6.9 106 IU/ml 9 -
cHCV 7 3/4 44.8 81.9 0.8 106 IU/ml n.d. -
rHBV 5 0/5 47.4 normal negative - -
rHCV 7 3/4 53.3 normal negative n.d. -
aEBV 3 2/1 44.3 n.d. negative - -
healthy 9 8/1 39.1 normal negative - -

Antibody reagents and viability dyes. The following antibodies were cells were enriched by anti-PE MACS-beads, directed against PE-labeled
used for flow cytometry: FITC antiT-bet (clone 4B10), Pacific Blue anti- pentamers. Calculation of pentamer+ CD8 T cells was performed as pre
CD45RA (HI100), APC-H7 antiinterferon- (4S.B3), and Pacific Blue viously described (Lucas et al., 2007). In brief, samples were divided in a
anti-CD57 (HCD57; BioLegend); APC anti-Eomes(WD1928) and eFluor780 preenrichment probe (10% of sample cells), which was used to determine the
anti-CD127 (eBioRDR5; eBioscience); APC-H7 anti-CD27 (M-T271), input number of CD8 T cells and a post-enrichment probe (90% of sample
APC antiPD-1 (MIH4), Alexa Fluor 647 anti-STAT4 (pY693; 38/pStat4), cells), which was run through miniMACS MS separation columns (Miltenyi
PerCP anti-CD3 (SK7), V500 anti-CD8 (SK1), V450 anti-CD8 (RPA-T8), Biotec). Frequencies of pentamer+ CD8 T cells were calculated by dividing
PerCP anti-CD14 (MP9), and PerCP anti-CD19 (SJ25C1; BD). 7-AAD the absolute number of pentamer+ CD8 T cells from the postenrichment
(BD) was used as viability dye in ex vivo stainings, whereas FVD eFluor450 probe by the number of CD8 T cells in the preenrichment probe 9. Sam-
(eBioscience) was used for fixed cells. ples were acquired on a FACSCanto II flow cytometer (BD). Data were ana-
lyzed with FlowJo 9.6.1. software (Tree Star). Gating strategy excluded
Synthetic peptides, pentamers, and cytokines. The following MHC-I monocytes (CD14+), B-lymphocytes (CD19+), and dead cells (7-AAD+) by
pentamers were used for the detection of epitope-specific CD8 T cells: HBV a dump channel.
core antigen 1827 FLPSDFFPSV, HBV polymerase 573581 FLLSLGIHL,
HBV envelope 183191 FLLTRILTI, HCV-NS3 14061415 KLVALGINAV, Detection of phosphorylated STAT4 by intracellular phospho-flow
EBV BMLF-1 259267 GLCTLVAML, and Flu MP 5866 GILGFVFTL (all cytometry. 2 106 PBMCs per well were incubated for 3 d with either
with MHC-I A0201 background); HCV-NS3 14361444 ATDALMTGF IL-2 or medium as control. On day 3, cells were stimulated for 20 min with
(HLA-A0101); HCV-NS5 25882596 RVCEKMALY (HLA-A0301); HCV either IL-2 or IL-12. After fixation and permeabilization with Perm buffer
core 4149 GPRLGVRAT (HLA-B0701) and HCV-NS3 13591367 HPNIE- III (BD) cells were stained with anti-pSTAT4 and antiT-bet before analysis
EVAL (HLA-B3501). All MHC-I pentamers were PE-labeled and obtained by flow cytometry.
from ProImmune. All corresponding peptides were synthesized by EMC micro-
collections and used for the in vitro stimulation of specific CD8 T cells. For Statistical analysis. Statistical analysis was performed with Prism 5.0 soft-
stimulation experiments, recombinant human IL-2 (R&D Systems) and IL-12 ware (GraphPad) and included 2-sided Wilcoxon matched pairs test and
p70 (eBioscience) were used.The following cytokines were screened for induc- Mann-Whitney-U test for unpaired samples. Statistical significance was de-
tion of T-bet in CD8 T cells: IL-18 (10 ng/ml), IL-21 (25 ng/ml), IL-23 (50 ng/ml), fined as P < 0.05.
interferon- (2,000 IU/ml), interferon- (2,000 IU/ml; eBioscience).
Online supplemental material. Table S1 shows the ex vivo frequencies of
PBMC stimulation. PBMCs were cultured for 3 d in RPMI 1640 medium pentamer+ CD8 T cells of the patients described in Fig. 1. Fig. S1 demon-
(Invitrogen) containing 2 mM l-glutamine, 1 mM sodium pyruvate, strates flow cytometry plots and gating procedure of CD4 tetramer stainings
100 U/ml of penicillin, 100 g of streptomycin/ml and 5% human AB of 2 patients with acute HCV (central plots). Online supplemental material
serum. In the cytokine-stimulated groups, IL-2 (20 IU/ml) and/or IL-12 http://www.jem.org/cgi/content/full/jem.20131333/DC1.
(50 pg/ml) were added on day 0. Antigen-stimulated groups received 5 g/ml
antigen on day 0 and were restimulated with the same dose of antigen on day 3. We thank Michaela Zankl for excellent technical assistance and Jutta Drrmann for
On day 3, cells were incubated for 6 h under stimulating conditions in the her continuous support.
presence of Brefeldin A (eBioscience). After stimulation, cells were prepared This work was supported by the Research Network Host and viral
for flow cytometry by intracellular cytokine staining. determinants for susceptibility and resistance to hepatitis C virus infection from
the German Federal Ministry of Research, and the European Research Council
funded Research Network HepaCute: Host and viral factors in acute hepatitis C.
PBSE proliferation assay. 500,000 PBMCs per condition were stained in
The authors have no competing financial interests.
120 M PBSE (Life Technologies). Cells were distributed on a 96-well
round-bottomed cell culture plate in 100 l of culture medium and were
Author contributions: P.D. Kurktschiev, B. Raziorrouh, W. Schraut, H.M. Diepolder,
cultured in the presence or absence of antigen (5 g/ml) for 7 d. On day 4,
and N.H. Gruener designed the experiments and analyzed the data; M. Spannagl
IL-2 (20 IU/ml) and/or IL-12 (50pg/ml) were added. On day 7, cells were
and A. Dick did the HLA typing; B. Bengsch and R. Thimme performed analysis of
collected and stained for flow cytometry. cHCV patients and critically reviewed the manuscript; M. Wchtler, M. Backmund,
R. Zachoval, and G. Denk provided patient samples and clinical information;
Flow cytometry. 23 106 PBMCs were stained with MHC-I pentamers M.C. Jung and J. Haas provided patient samples and clinical information and
according to the manufacturers instructions. After staining with viability dyes critically reviewed the manuscript; and P.D. Kurktschiev and N.H. Gruener wrote
and antibodies specific for surface markers, cells were fixed with intracellular the manuscript.
fixation buffer and permeabilized with permeabilization buffer (both from
eBioscience). After the permeabilization step, cells were stained with intracel- Submitted: 26 June 2013
lular markers (T-bet, Eomes, and interferon-). For ex vivo staining, pentamer+ Accepted: 25 July 2014

JEM Vol. 211, No. 10 2057


REFERENCES PD-1 and sustains virus-specific CD8+ T cell responses during chronic
Artillo, S., G. Pastore, A. Alberti, M. Milella, T. Santantonio, G. Fattovich, G. infection. Nat. Immunol. 12:663671. http://dx.doi.org/10.1038/ni.2046
Giustina, J.C. Ryff, M. Chaneac, J. Bartolom, and V. Carreo. 1998. Kittlesen, D.J., K.A. Chianese-Bullock, Z.Q. Yao, T.J. Braciale, and Y.S.
Double-blind, randomized controlled trial of interleukin-2 treatment of Hahn. 2000. Interaction between complement receptor gC1qR and
chronic hepatitis B. J. Med.Virol. 54:167172. http://dx.doi.org/10.1002/ hepatitis C virus core protein inhibits T-lymphocyte proliferation.
(SICI)1096-9071(199803)54:3<167::AID-JMV4>3.0.CO;2-3 J. Clin. Invest. 106:12391249. http://dx.doi.org/10.1172/JCI10323
Auffermann-Gretzinger, S., E.B. Keeffe, and S. Levy. 2001. Impaired dendritic cell Lauer, G.M., and B.D. Walker. 2001. Hepatitis C virus infection. N. Engl. J.
maturation in patients with chronic, but not resolved, hepatitis C virus infec- Med. 345:4152. http://dx.doi.org/10.1056/NEJM200107053450107
tion. Blood. 97:31713176. http://dx.doi.org/10.1182/blood.V97.10.3171 Lechner, F., N.H. Gruener, S. Urbani, J. Uggeri,T. Santantonio, A.R. Kammer,
Boni, C., P. Fisicaro, C. Valdatta, B. Amadei, P. Di Vincenzo, T. Giuberti, D. A. Cerny, R. Phillips, C. Ferrari, G.R. Pape, and P. Klenerman. 2000a.
Laccabue,A. Zerbini,A. Cavalli, G. Missale, et al. 2007. Characterization of CD8+ T lymphocyte responses are induced during acute hepatitis C virus
hepatitis B virus (HBV)-specific T-cell dysfunction in chronic HBV infec- infection but are not sustained. Eur. J. Immunol. 30:24792487. http://dx
tion. J.Virol. 81:42154225. http://dx.doi.org/10.1128/JVI.02844-06 .doi.org/10.1002/1521-4141(200009)30:9<2479::AID-IMMU2479>3
Bowen, D.G., M. Zen, L. Holz, T. Davis, G.W. McCaughan, and P. Bertolino. .0.CO;2-B
2004.The site of primary T cell activation is a determinant of the balance Lechner, F., D.K.Wong, P.R. Dunbar, R. Chapman, R.T. Chung, P. Dohrenwend,
between intrahepatic tolerance and immunity. J. Clin. Invest. 114:701 G. Robbins, R. Phillips, P. Klenerman, and B.D. Walker. 2000b. Analysis of
712. http://dx.doi.org/10.1172/JCI200421593 successful immune responses in persons infected with hepatitis C virus. J.
Carreo,V., S. Zeuzem, U. Hopf, P. Marcellin,W.G. Cooksley, J. Fevery, M. Diago, Exp. Med. 191:14991512. http://dx.doi.org/10.1084/jem.191.9.1499
R. Reddy, M. Peters, K. Rittweger, et al. 2000.A phase I/II study of recombi- Liao,W., J.X. Lin, L.Wang, P. Li, and W.J. Leonard. 2011. Modulation of cytokine
nant human interleukin-12 in patients with chronic hepatitis B. J. Hepatol. receptors by IL-2 broadly regulates differentiation into helper T cell lin-
32:317324. http://dx.doi.org/10.1016/S0168-8278(00)80078-1 eages. Nat. Immunol. 12:551559. http://dx.doi.org/10.1038/ni.2030
Chang, K.M., B. Rehermann, J.G. McHutchison, C. Pasquinelli, S. Southwood, Lighvani,A.A., D.M. Frucht, D. Jankovic, H.Yamane, J.Aliberti, B.D. Hissong, B.V.
A. Sette, and F.V. Chisari. 1997. Immunological significance of cytotoxicT lym Nguyen, M. Gadina, A. Sher,W.E. Paul, and J.J. OShea. 2001.T-bet is rapidly
phocyte epitope variants in patients chronically infected by the hepatitis C induced by interferon-gamma in lymphoid and myeloid cells. Proc. Natl.Acad.
virus. J. Clin. Invest. 100:23762385. http://dx.doi.org/10.1172/JCI119778 Sci. USA. 98:1513715142. http://dx.doi.org/10.1073/pnas.261570598
Cruz-Guilloty, F., M.E. Pipkin, I.M. Djuretic, D. Levanon, J. Lotem, M.G. Lucas, M., A. Ulsenheimer, K. Pfafferot, M.H. Heeg, S. Gaudieri, N. Grner, A.
Lichtenheld,Y. Groner, and A. Rao. 2009. Runx3 and T-box proteins co- Rauch, J.T. Gerlach, M.C. Jung, R. Zachoval, et al. 2007. Tracking virus-
operate to establish the transcriptional program of effector CTLs. J. Exp. specific CD4+ T cells during and after acute hepatitis C virus infection.
Med. 206:5159. http://dx.doi.org/10.1084/jem.20081242 PLoS ONE. 2:e649. http://dx.doi.org/10.1371/journal.pone.0000649
Diepolder, H.M., R. Zachoval, R.M. Hoffmann, E.A. Wierenga, T. Mullen, A.C., F.A. High, A.S. Hutchins, H.W. Lee, A.V. Villarino, D.M.
Santantonio, M.C. Jung, D. Eichenlaub, and G.R. Pape. 1995. Possible Livingston, A.L. Kung, N. Cereb, T.P. Yao, S.Y. Yang, and S.L. Reiner.
mechanism involving T-lymphocyte response to non-structural protein 2001. Role of T-bet in commitment of TH1 cells before IL-12-
3 in viral clearance in acute hepatitis C virus infection. Lancet. 346:1006 dependent selection. Science. 292:19071910. http://dx.doi.org/10.1126/
1007. http://dx.doi.org/10.1016/S0140-6736(95)91691-1 science.1059835
Eisen-Vandervelde, A.L., S.N. Waggoner, Z.Q. Yao, E.M. Cale, C.S. Hahn, and Paley, M.A., D.C. Kroy, P.M. Odorizzi, J.B. Johnnidis, D.V. Dolfi, B.E. Barnett,
Y.S. Hahn. 2004. Hepatitis C virus core selectively suppresses interleukin-12 E.K. Bikoff, E.J. Robertson, G.M. Lauer, S.L. Reiner, and E.J.Wherry. 2012.
synthesis in human macrophages by interfering with AP-1 activation. J. Biol. Progenitor and terminal subsets of CD8+T cells cooperate to contain chronic
Chem. 279:4347943486. http://dx.doi.org/10.1074/jbc.M407640200 viral infection. Science. 338:12201225. http://dx.doi.org/10.1126/
Gerlach, J.T., H.M. Diepolder, M.C. Jung, N.H. Gruener, W.W. Schraut, R. science.1229620
Zachoval, R. Hoffmann, C.A. Schirren, T. Santantonio, and G.R. Pape. Pardo, M., I. Castillo, H. Oliva, A. Fernndez-Flores, R. Brcena, M.A. de
1999. Recurrence of hepatitis C virus after loss of virus-specific CD4(+) Peuter, and V. Carreo. 1997. A pilot study of recombinant interleukin-2
T-cell response in acute hepatitis C. Gastroenterology. 117:933941. for treatment of chronic hepatitis C. Hepatology. 26:13181321.
http://dx.doi.org/10.1016/S0016-5085(99)70353-7 Pearce, E.L., A.C. Mullen, G.A. Martins, C.M. Krawczyk, A.S. Hutchins,
Gruener, N.H., F. Lechner, M.C. Jung, H. Diepolder, T. Gerlach, G. Lauer, V.P. Zediak, M. Banica, C.B. DiCioccio, D.A. Gross, C.A. Mao, et al.
B. Walker, J. Sullivan, R. Phillips, G.R. Pape, and P. Klenerman. 2001. 2003. Control of effector CD8+ T cell function by the transcription
Sustained dysfunction of antiviral CD8+ T lymphocytes after infec- factor Eomesodermin. Science. 302:10411043. http://dx.doi.org/10
tion with hepatitis C virus. J. Virol. 75:55505558. http://dx.doi.org/ .1126/science.1090148
10.1128/JVI.75.12.5550-5558.2001 Perlmann, H., P. Perlmann, G.R. Pape, and G. Halldn. 1976. Purification,
Hersperger, A.R., J.N. Martin, L.Y. Shin, P.M. Sheth, C.M. Kovacs, G.L. fractionation and assay of antibody-dependent lymphocytic effector cells
Cosma, G. Makedonas, F. Pereyra, B.D. Walker, R. Kaul, et al. 2011. (K cells) in human blood. Scand. J. Immunol. 5:5768. http://dx.doi.org/
Increased HIV-specific CD8+ T-cell cytotoxic potential in HIV elite 10.1111/j.1365-3083.1976.tb03856.x
controllers is associated with T-bet expression. Blood. 117:37993808. Rau, H.P., C. Beadling, J. Haun, and M.K. Slifka. 2013. Cytokine-mediated
http://dx.doi.org/10.1182/blood-2010-12-322727 programmed proliferation of virus-specific CD8(+) memory T cells. Immunity.
Intlekofer, A.M., N. Takemoto, E.J. Wherry, S.A. Longworth, J.T. Northrup,V.R. 38:131139. http://dx.doi.org/10.1016/j.immuni.2012.09.019
Palanivel, A.C. Mullen, C.R. Gasink, S.M. Kaech, J.D. Miller, et al. 2005. Ribeiro-dos-Santos, P., E.L. Turnbull, M. Monteiro, A. Legrand, K. Conrod, J.
Effector and memory CD8+ T cell fate coupled by T-bet and eomesoder- Baalwa, P. Pellegrino, G.M. Shaw, I.Williams, P. Borrow, and B. Rocha. 2012.
min. Nat. Immunol. 6:12361244. http://dx.doi.org/10.1038/ni1268 Chronic HIV infection affects the expression of the 2 transcription fac-
Intlekofer, A.M., N. Takemoto, C. Kao, A. Banerjee, F. Schambach, J.K. tors required for CD8 T-cell differentiation into cytolytic effectors. Blood.
Northrop, H. Shen, E.J. Wherry, and S.L. Reiner. 2007. Requirement for 119:49284938. http://dx.doi.org/10.1182/blood-2011-12-395186
T-bet in the aberrant differentiation of unhelped memory CD8+ T cells. Schurich, A., L.J. Pallett, M. Lubowiecki, H.D. Singh, U.S. Gill, P.T. Kennedy, E.
J. Exp. Med. 204:20152021. http://dx.doi.org/10.1084/jem.20070841 Nastouli, S. Tanwar, W. Rosenberg, and M.K. Maini. 2013. The third signal
Joshi, N.S., W. Cui, A. Chandele, H.K. Lee, D.R. Urso, J. Hagman, L. cytokine IL-12 rescues the anti-viral function of exhausted HBV-specific
Gapin, and S.M. Kaech. 2007. Inflammation directs memory precur- CD8 T cells. PLoS Pathog. 9:e1003208. http://dx.doi.org/10.1371/journal
sor and short-lived effector CD8(+) T cell fates via the graded expres- .ppat.1003208
sion of T-bet transcription factor. Immunity. 27:281295. http://dx.doi Shin, H., S.D. Blackburn, J.N. Blattman, and E.J.Wherry. 2007.Viral antigen and
.org/10.1016/j.immuni.2007.07.010 extensive division maintain virus-specific CD8 T cells during chronic infec-
Kao, C., K.J. Oestreich, M.A. Paley,A. Crawford, J.M.Angelosanto, M.A.Ali,A.M. tion. J. Exp. Med. 204:941949. http://dx.doi.org/10.1084/jem.20061937
Intlekofer, J.M. Boss, S.L. Reiner, A.S. Weinmann, and E.J. Wherry. 2011. Shoukry, N.H., A. Grakoui, M. Houghton, D.Y. Chien, J. Ghrayeb, K.A.
Transcription factor T-bet represses expression of the inhibitory receptor Reimann, and C.M. Walker. 2003. Memory CD8+ T cells are required

2058 Dysfunctional CD8+ T cells in HBV/HCV lack T-bet | Kurktschiev et al.


Ar ticle

for protection from persistent hepatitis C virus infection. J. Exp. Med. von Hahn, T., J.C. Yoon, H. Alter, C.M. Rice, B. Rehermann, P. Balfe, and J.A.
197:16451655. http://dx.doi.org/10.1084/jem.20030239 McKeating. 2007. Hepatitis C virus continuously escapes from neutralizing
Sullivan, B.M., A. Juedes, S.J. Szabo, M. von Herrath, and L.H. Glimcher. antibody andT-cell responses during chronic infection in vivo.Gastroenterology.
2003. Antigen-driven effector CD8 T cell function regulated by T-bet. 132:667678. http://dx.doi.org/10.1053/j.gastro.2006.12.008
Proc. Natl. Acad. Sci. USA. 100:1581815823. http://dx.doi.org/10 Wherry, E.J. 2011. T cell exhaustion. Nat. Immunol. 12:492499. http://dx
.1073/pnas.2636938100 .doi.org/10.1038/ni.2035
Szabo, S.J., S.T. Kim, G.L. Costa, X. Zhang, C.G. Fathman, and L.H. Glimcher. Witt, C.S., P. Price, G. Kaur, K. Cheong, U. Kanga, D. Sayer, F. Christiansen,
2000. A novel transcription factor,T-bet, directs Th1 lineage commitment. and N.K. Mehra. 2002. Common HLA-B8-DR3 haplotype in Northern
Cell. 100:655669. http://dx.doi.org/10.1016/S0092-8674(00)80702-3
India is different from that found in Europe. Tissue Antigens. 60:474480.
Thieu,V.T., Q.Yu, H.C. Chang, N.Yeh, E.T. Nguyen, S. Sehra, and M.H. Kaplan.
http://dx.doi.org/10.1034/j.1399-0039.2002.600602.x
2008. Signal transducer and activator of transcription 4 is required for the
Wright,T.L., and J.Y. Lau. 1993. Clinical aspects of hepatitis B virus infection. Lancet.
transcription factor T-bet to promote T helper 1 cell-fate determination.
Immunity. 29:679690. http://dx.doi.org/10.1016/j.immuni.2008.08.017 342:13401344. http://dx.doi.org/10.1016/0140-6736(93)92250-W
Thimme, R., S. Wieland, C. Steiger, J. Ghrayeb, K.A. Reimann, R.H. Purcell, Ylikoski, E., R. Lund, M. Kylniemi, S. Filn, M. Kilpelinen, J. Savolainen,
and F.V. Chisari. 2003. CD8(+) T cells mediate viral clearance and disease and R. Lahesmaa. 2005. IL-12 up-regulates T-bet independently of IFN-
pathogenesis during acute hepatitis B virus infection. J. Virol. 77:6876. gamma in human CD4+ T cells. Eur. J. Immunol. 35:32973306. http://
http://dx.doi.org/10.1128/JVI.77.1.68-76.2003 dx.doi.org/10.1002/eji.200526101
Urbani, S., B. Amadei, P. Fisicaro, D. Tola, A. Orlandini, L. Sacchelli, C. Mori, Zeuzem, S., U. Hopf,V. Carreno, M. Diago, M. Shiffman, S. Grne, F.J. Dudley,
G. Missale, and C. Ferrari. 2006. Outcome of acute hepatitis C is related A. Rakhit, K. Rittweger, S.H. Yap, et al. 1999. A phase I/II study of re-
to virus-specific CD4 function and maturation of antiviral memory CD8 combinant human interleukin-12 in patients with chronic hepatitis C.
responses. Hepatology. 44:126139. http://dx.doi.org/10.1002/hep.21242 Hepatology. 29:12801287. http://dx.doi.org/10.1002/hep.510290429

JEM Vol. 211, No. 10 2059


Article

Interplay between regulatory T cells


and PD-1 in modulating T cell
exhaustion and viral control
during chronic LCMV infection
Pablo Penaloza-MacMaster,1 Alice O. Kamphorst,1 Andreas Wieland,1
Koichi Araki,1 Smita S. Iyer,1 Erin E. West,1 Leigh OMara,1 Shu Yang,1,2
Bogumila T. Konieczny,1 Arlene H. Sharpe,3 Gordon J. Freeman,4
Alexander Y. Rudensky,5,6,7 and Rafi Ahmed1
1Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
2XiangyaSchool of Medicine, Central South University, Changsha, Hunan Province, 410013, China
3Department of Microbiology and Immunology, and 4Department of Medical Oncology and Dana Farber Cancer Institute,
Department of Medicine, Harvard Medical School, Boston, MA 02115
5Howard Hughes Medical Institute, 6Immunology Program, Sloan-Kettering Institute for Cancer Research, and 7Ludwig Center

at Memorial Sloan-Kettering Cancer Center, New York, NY 10065

Regulatory T (T reg) cells are critical for preventing autoimmunity mediated by self-reactive
T cells, but their role in modulating immune responses during chronic viral infection is not
well defined. To address this question and to investigate a role for T reg cells in exhaustion
of virus-specific CD8 T cells, we depleted T reg cells in mice chronically infected with
lymphocytic choriomeningitis virus (LCMV). T reg cell ablation resulted in 10100-fold
expansion of functional LCMV-specific CD8 T cells. Rescue of exhausted CD8 T cells was
dependent on cognate antigen, B7 costimulation, and conventional CD4 T cells. Despite the
striking recovery of LCMV-specific CD8 T cell responses, T reg cell depletion failed to
diminish viral load. Interestingly, T reg cell ablation triggered up-regulation of the molecule
programmed cell death ligand-1 (PD-L1), which upon binding PD-1 on T cells delivers
inhibitory signals. Increased PD-L1 expression was observed especially on LCMV-infected
cells, and combining T reg cell depletion with PD-L1 blockade resulted in a significant
reduction in viral titers, which was more pronounced than that upon PD-L1 blockade alone.
These results suggest that T reg cells effectively maintain CD8 T cell exhaustion, but block-
ade of the PD-1 inhibitory pathway is critical for elimination of infected cells.

CORRESPONDENCE Regulatory T cells expressing transcription fac- and facilitate disease progression due to inhibi-
Rafi Ahmed: tor Foxp3 are indispensable for preventing im- tion of T cell responses (Zou, 2006; Li et al.,
rahmed@emory.edu
OR
mune responses to self, and their absence results 2008; Belkaid and Tarbell, 2009; Dietze et al.,
Alexander Rudensky: in multi-organ autoreactivity and death (Kim 2011; Punkosdy et al., 2011).
rudenska@mskcc.org et al., 2007; Sakaguchi et al., 2008). In addition In cancer and persistent infections, chronic
to their major role in maintaining peripheral antigenic stimulation causes deterioration of
Abbreviations used: Arm, Arm-
strong; cl-13, clone 13; DT, tolerance, T reg cells also control immune re- T cell responses.T cell exhaustion is manifested
diphtheria toxin; LCMV, lym- sponses to infections. During acute infection, by progressive loss of proliferative potential,
phocytic choriomeningitis virus; T reg cells can promote migration of effector cytokine production, and for CD8 T cells, killing
MFI, mean fluorescence inten-
sity; PD-1, programmed cell
immune cells to infection sites by modulating capability (Zajac et al., 1998;Wherry, 2003, 2011).
death-1; PD-L1, programmed chemokine production (Lund et al., 2008), and This progressive T cell dysfunction is associated
cell death ligand-1. prevent the activation of low avidity CD8 with expression of programmed cell death-1
T cells (Pace et al., 2012). However, in cancer
2014 Penaloza-MacMaster et al. This article is distributed under the terms of an
and persistent infections, T reg cells may expand AttributionNoncommercialShare AlikeNo Mirror Sites license for the first six months
after the publication date (see http://www.rupress.org/terms). After six months
it is available under a Creative Commons License (AttributionNoncommercial
Pablo Penaloza-MacMaster and Alice O. Kamphorst contrib- Share Alike 3.0 Unported license, as described at http://creativecommons.org/
uted equally to this paper. licenses/by-nc-sa/3.0/).

The Rockefeller University Press $30.00


J. Exp. Med. 2014 Vol. 211 No. 9 1905-1918 1905
www.jem.org/cgi/doi/10.1084/jem.20132577
Figure 1. T reg cells have an activated phenotype during chronic viral infection. Analysis was done on splenocytes from naive uninfected mice,
mice infected with LCMV Arm, and analyzed at least 100 d after acute infection and LCMV cl-13 chronically infected mice (mice transiently depleted of CD4
T cells and infected with LCMV cl-13, with analysis done at least 40 d after infection). (A) Percentage of T reg cells (Foxp3+) among CD4 T cells. (B) Absolute
number of T reg cells in spleen. (C) Frequency of V5+ cells among T reg cells. Mice infected with LCMV cl-13 and analyzed 21 d later were included as posi-
tive controls (2240% V5+ cells). (D) Graph shows MFI of CD25 expression on T reg cells. (E) Histograms show representative expression of different
markers by T reg cells and numbers represent MFI. Data are a compilation of 4 experiments (A and B) or show representative results from 3 experiments
(CE; n = 34 mice per group). Error bars indicate SEM. Non-parametric Mann Whitney test, where *, P < 0.05; **, P < 0.01; NS = not significant.

(PD-1) and other inhibitory receptors such as Tim-3 and LAG-3 Foxp3DTR knock-in mice in which Foxp3+ T reg cells express
(Barber et al., 2006; Blackburn et al., 2009; Jin et al., 2010). Im- the human diphtheria toxin (DT) receptor under control of
portantly, proliferation and function of exhausted T cells can be the endogenous Foxp3 locus and can be efficiently and spe-
rescued by blockade of inhibitory pathways, which can result in cifically deleted by administration of DT (Kim et al., 2007).
restoration of effective immune responses that control infec- Using this approach, we found that T reg cell ablation in
tions and tumors (Barber et al., 2006; Fourcade et al., 2010; chronically infected mice leads to a striking rescue of exhausted
Sakuishi et al., 2010; Butler et al., 2012; Topalian et al., 2012). viral-specific CD8 T cells. Restoration of antiviral CD8 T cell
Multiple pathways contribute to T cell dysfunction. Be- responses was dependent on cognate antigen, B7 costimula-
sides expression of inhibitory receptors by T cells, extrinsic tion, and conventional CD4 T cells. Interestingly, viral control
factors such as cytokines also play a fundamental role in T cell was not achieved unless T reg cell depletion was combined to
exhaustion (Wherry, 2011). In addition, lack of CD4 help blockade of the PD-1 pathway. Thus, we propose that even
exacerbates CD8 T cell exhaustion (Matloubian et al., 1994; though T reg cells maintain CD8 T cells in an exhausted state
Zajac et al., 1998; Lichterfeld et al., 2004), and its restoration during persistent infections, the PD-1 inhibitory pathway
via adoptive transfer of CD4 T cells can reinvigorate virus- further operates by inhibiting the cytotoxic activity of res-
specific responses in mice chronically infected with lympho- cued CD8 T cells toward target cells expressing high levels of
cytic choriomeningitis virus (LCMV; Aubert et al., 2011). programmed cell death ligand-1 (PD-L1).
IL-21 produced by CD4 T cells plays an important role in
sustaining CD8 T cells during chronic infection (Elsaesser RESULTS
et al., 2009; Frhlich et al., 2009; Yi et al., 2009), and it was Regulatory T cells modulate exhausted
recently reported that IL-21 may also aid CD8 T cells by CD8 T cells during chronic viral infection
restricting T reg cell expansion (Schmitz et al., 2013). Thus, To establish a lifelong viral infection of multiple organs ac-
conventional CD4 T cells have a positive impact on modu- companied by exhaustion of virus-specific CD8 T cells, we in
lating CD8 T cell function during persistent antigenic stimu- fected mice with LCMV cl-13 after antibody-mediated transient
lation. In contrast, it has been described that T reg cells are depletion of CD4 T cells (Matloubian et al., 1994). By day 45
detrimental to virus-specific T cell responses during persis- after infection, CD4 T cells bounced back to normal numbers
tent infection in mice (Dittmer et al., 2004; Dietze et al., in infected mice. When compared with naive or LCMV Arm-
2011; Schmitz et al., 2013); nevertheless, the role of T reg cells strong (Arm)infected mice that had cleared virus, LCMV
in maintaining T cell exhaustion has not been well character- chronically infected mice had increased frequency of T reg
ized or fully explored as a therapeutic approach. cells (Fig. 1 A). However, due to decreased splenic cellularity,
To analyze the effects of T reg cells on exhausted virus- the absolute numbers of T reg cells were actually reduced in
specific CD8 T cells, we used LCMV clone 13 (cl-13) infected chronically infected mice (Fig. 1 B). Punkosdy et al. (2011)

1906 T reg cells and PD-1 modulate T cell exhaustion | Penaloza-MacMaster et al.
Ar ticle

Figure 2. Exhausted CD8 T cells expand and undergo phenotypic changes upon T reg cell ablation. (A) Experimental outline. (B) Total numbers
of activated T cells in the spleen of PBS-treated or T reg celldepleted (DT) mice chronically infected with LCMV. (C) Number of LCMV-specific (Db GP276)
CD8 T cells among PBMCs before and after DT treatment. (D and E) Absolute numbers (D) and frequency (E) of LCMV-specific (Db GP276) CD8 T cells
within total CD8 T cell population. (F) Absolute numbers of LCMV DbGP276-specific CD8 T cells in the spleen at different days after T reg cell ablation.
(G) Proliferative activity of LCMV-specific (Db-GP276) CD8 T cells. Numbers show MFI of Ki67 expression. (H) Phenotype of LCMV-specific (Db GP276) CD8
T cells in spleen. Numbers show MFI of different CD8 T cell markers. Data are a compilation of 5 independent experiments with 35 mice per group. Error
bars indicate SEM. Non-parametric Mann Whitney test, where *, P < 0.05; ***, P < 0.001; NS = not significant.

have reported a transient expansion of V5+ T reg cells recogniz- and at least 45 d after infection, T reg cells were depleted by
ing Mtv-9 endogenous retrovirus that peaks at day 17 of chronic DT administration (Fig. 2 A). T reg cell ablation in LCMV
LCMV infection in B6 mice. Hence, the frequency of V5+ T reg chronically infected mice led to a marked increase in absolute
cells was not significantly increased in LCMV chronically infected numbers of activated T cells (Fig. 2 B) and LCMV-specific
mice analyzed at 40 or more days after infection (Fig. 1 C). CD8 T cells (Fig. 2, CE). There was a >50-fold increase in
To further characterize the T reg cell population in LCMV Db GP276 LCMV-specific CD8 T cells in blood (Fig. 2 C),
10-fold increase in the spleen, and up to 100-fold increase in
chronically infected mice, we assessed the expression of sev-
multiple nonlymphoid tissues (Fig. 2, D and E). This striking
eral cell surface molecules. CD25 expression was not signifi-
reversal of CD8 T cell exhaustion occurred between day 5 and 7
cantly different between T reg cells of chronically infected
after T reg cell depletion (Fig. 2 F) and was accompanied by
mice, naive mice, or LCMV Arminfected mice that had restoration of proliferative capacity assessed by the expression
cleared virus (Fig. 1 D). However, based on the expression of the cell cycle progression marker Ki67 (Fig. 2 G).These re-
levels of several other markers, such as CD69, CD44, CD62L, sults show that during chronic infection, T reg cell depletion
ICOS, GITR, and CTLA-4, T reg cells from chronically in- enables expansion of previously exhausted T cells.
fected mice appeared more activated (Fig. 1 E). In addition to expansion, the surface phenotype of ex-
To examine the role of T reg cells during chronic infec- hausted LCMV-specific CD8 T cells was also altered by T reg
tion, we infected Foxp3DTR knock-in mice with LCMV cl-13 cell ablation manifested by up-regulation of IL-7 receptor
after antibody-mediated transient depletion of CD4 T cells, (CD127), and increased CD44 and granzyme B expression

JEM Vol. 211, No. 9 1907


Figure 3. LCMV-specific CD8 T cells re-
gain effector function upon T reg cell ab-
lation in chronically infected mice. LCMV
chronically infected Foxp3DTR knock-in mice
were depleted of T reg cells for 10 d by DT
administration as in Fig. 2. (A) Absolute num-
bers of CD8 T cells in spleen producing IFN-.
(B) MFI of IFN- production by CD8 T cells
after in vitro restimulation with various LCMV
peptides. (C) Percentage among DbGP276-
specific cells that express IFN- in spleen.
(D) Frequency of CD8 T cells producing both
IFN- and TNF after in vitro restimulation with
LCMV peptides. (E) Degranulation and surface
expression of CD107 a/b in CD8 T cells after
in vitro restimulation with GP276 LCMV peptide.
(F) Granzyme B expression on LCMV DbG276-
specific CD8 T cells in spleen. (G) Ex vivo cyto-
toxic activity of splenic CD8 T cells measured
by 51Cr release from MC57 target cells un-
pulsed (control) or pulsed with a mix of LCMV
peptides (GP33, GP276, and NP396). Data are
a compilation of 5 independent experiments
with 35 mice per group. Error bars indicate
SEM. Non-parametric Mann Whitney test,
where *, P < 0.05; ***, P < 0.001.

(Fig. 2 H). CD127 is expressed on naive and memory cells to induces CD127 expression on exhausted CD8 T cells during
promote their long-term survival but is down-regulated on chronic LCMV infection (West et al., 2013). Hence, it is pos-
exhausted T cells (Kaech et al., 2003; Wherry and Ahmed, sible that an increase in IL-2 availability upon T reg cell abla-
2004). Interestingly, a recent report described that IL-2 therapy tion triggers CD127 up-regulation on exhausted cells.

Figure 4. Rescue of LCMV-specific CD8 T cell


responses by T reg cell ablation is greater than
by blockade of the PD-1 pathway. LCMV chron-
ically infected Foxp3DTR knock-in mice were de-
pleted of T reg cells for 10 d by DT administration
as in Fig. 2 or mice received 3 doses of PD-L1
blocking antibody, every 3 d. (A and B) Dot plots
(A) show frequency in the same mouse and graphs
(B) show number of LCMV-specific (Db GP276)
CD8 T cells among PBMCs before and after treat-
ment. (C and D) Dot plots show frequency (C) and
graphs show number (D) of LCMV-specific (Db
GP33) CD8 T cells in different organs 11 d after
treatment. Data are a compilation of 3 indepen-
dent experiments with 35 mice per group. Error
bars indicate SEM. Non-parametric Mann Whitney
test, where *, P < 0.05; **, P < 0.01; ***, P < 0.001;
NS = not significant.

1908 T reg cells and PD-1 modulate T cell exhaustion | Penaloza-MacMaster et al.
Ar ticle

Figure 5. Cognate antigen is necessary for activation of antiviral T cells after T reg cell depletion. (A) Experimental outline. (B) Total numbers
of activated T cells in the spleen of PBS-treated or T reg celldepleted (DT) mice 100 d after infection with LCMV Arm. (C) Phenotype of total CD8 T cells
in spleen. (D) Absolute numbers of LCMV-specific (Db GP276) CD8 T cells. (E) Absolute numbers of cells in the spleen that produce IFN- after in vitro
restimulation with various LCMV peptides. (F) Phenotype of splenic LCMV-specific (Db GP276) CD8 T cells. A representative of 3 independent experiments
is shown, with 56 mice per group. Error bars indicate SEM. Non-parametric Mann Whitney test, where ***, P < 0.001; NS = not significant.

Next, we wanted to ascertain whether T reg cell ablation For example, blockade of the PD-1 inhibitory pathway in-
in chronically infected mice restored functionality of exhausted creased the frequency of LCMV-specific CD8 T cells in blood
T cells. We observed a marked increase in frequency and ab- by an average of fivefold, whereas T reg depletion led to an
solute numbers of virus-specific CD8 T cells that could pro- increase of almost 100-fold (Fig. 4, A and B). Similarly, the
duce IFN- upon stimulation with LCMV peptides (Fig. 3 A). frequency and total number of LCMV-specific CD8 T cells in
T reg cell depletion not only affected the number of LCMV- spleen, lung, and liver achieved by T reg cell depletion was
specific functional T cells but also substantially increased the higher than by blockade of the PD-1 inhibitory pathway in
amount of IFN- produced per cell as indicated by the mean LCMV chronically infected mice (Fig. 4, C and D).Thus,T reg
fluorescence intensity (MFI) of intracellular IFN- staining cells play a prominent role in the maintenance of CD8 T cell
(Fig. 3 B). After T reg cell depletion, >75% of Db GP276 LCMV- exhaustion during chronic viral infection.
specific T cells became functionally competent and acquired
ability to produce IFN- (Fig. 3 C).We also observed increased Cognate antigen is necessary for activation
TNF production to all LCMV epitopes tested and even se- of antiviral T cells after T reg cell depletion
verely exhausted CD8 T cells specific for the NP396 LCMV Our results so far revealed that T reg cell ablation leads to a
epitope were rescued upon T reg cell ablation (Fig. 3 D). striking rescue of virus-specific CD8 T cells during chronic
Furthermore, after T reg cell depletion, LCMV-specific LCMV infection.To explore if cognate antigen was necessary
CD8 T cells showed heightened ability to degranulate upon for T cell activation that ensues after T reg cell depletion, we
cognate peptide stimulation as measured by surface expression examined antiviral CD8 T cells after acute infection when
of the lysosomal marker CD107a/b (Fig. 3 E). As much as 73% antigen has been cleared. We infected Foxp3DTR knock-in
of all CD8 T cells expressed granzyme B (Fig. 3 F), and res- mice with LCMV Arm, and T reg cells were depleted by DT
cued LCMV-specific T cells were capable of in vitro killing of administration at least 100 d after infection (Fig. 5 A). LCMV
antigen-bearing target cells (Fig. 3 G). Hence,T reg cell ablation Arm causes an acute infection that is cleared within 810 d,
in LCMV chronically infected mice rescued virus-specific and generates virus-specific long-lived memory CD8 T cells
CD8 T cells to proliferate and recover function. that persist in the absence of cognate antigen (Lau et al., 1994;
The impressive increase in LCMV-specific responses after Murali-Krishna et al., 1999;Wherry and Ahmed, 2004). Simi-
T reg cell depletion was of a much higher magnitude than the lar to mice chronically infected with LCMV, T reg cell abla-
one caused by other means of rescue of exhausted CD8 T cells. tion triggered expansion of total activated CD4 and CD8

JEM Vol. 211, No. 9 1909


Figure 6. Co-stimulation and CD4 T cells are
required for the rescue of exhausted CD8 T cells
upon T reg cell depletion. Foxp3DTR knock-in mice
chronically infected with LCMV cl-13 received DT for 10 d
(as in Fig. 2). CTLA-4 Ig was administered every other
day during T reg cell depletion starting at day 2,
and CD4 T cells were depleted upon administration of
CD4 antibody on days 2 and 1 of T reg cell abla-
tion. (A) Representative dot plots show expression of
B7.1 and B7.2 on splenic DCs after 8 d of T reg cell
ablation. (B and C) Frequency of splenic CD8 T cells
that co-express IFN- and TNF (B) and absolute
numbers of CD8 T cells that produce IFN- (C) after
in vitro restimulation with LCMV peptides. (D) Frequency
of LCMV-specific (Db-GP276) and CD127-expressing
splenic CD8 T cells. (E) Graphs show B7.1 and B7.2 MFI
on CD11b+ splenic DCs after 8 d of T reg cell ablation
preceded or not by conventional CD4 T cell depletion.
The data are representative of 3 independent experi-
ments with 35 mice per group. Error bars indicate
SEM. Non-parametric Mann Whitney test, where
*, P < 0.05; **, P < 0.01; ***, P < 0.001.

T cells (Fig. 5 B). After T reg cell depletion, CD8 T cells showed antigen. Our results show that bystander effects upon T reg
extensive phenotypic changes associated with activation such cell ablation do not impact memory T cells once the antigen
as down-regulation of CD62L and CD127 (Fig. 5 C). However, has been cleared and indicate that T reg cells mostly suppress
despite massive activation of the bulk CD4 and CD8 T cell antigen-driven proliferation of T cells.
subsets upon T reg cell ablation, the absolute numbers of LCMV-
specific CD8 T cells remained unchanged in the spleen and Rescue of exhausted CD8 T cells by T reg ablation is dependent
nonlymphoid organs in mice that had cleared LCMV (Fig. 5 D). on B7 costimulation and conventional CD4 T cells
Analysis of T cell responses to multiple LCMV epitopes also To further explore potential mechanisms of T reg cell
showed no alterations in the functionality of LCMV-specific dependent enforcement of an exhausted state of T cells, we
memory CD8 T cells after T reg cell depletion (Fig. 5 E). In looked at DC activation status. Under steady-state conditions,
addition, the phenotype associated with long-lived memory T reg cells control expression of costimulatory molecules on
CD8 T cells was unaltered by depletion of T reg cells (Fig. 5 F). DCs (Kim et al., 2007; Wing et al., 2008; Schildknecht et al.,
Thus, T reg cells do not play a significant role in regulating 2010; Qureshi et al., 2011). Consistent with previous reports,
the homeostasis of memory T cells in the absence of cognate we noticed increased expression of costimulatory molecules

1910 T reg cells and PD-1 modulate T cell exhaustion | Penaloza-MacMaster et al.
Ar ticle

Figure 7. Inability to clear virus coincides with PD-L1 up-regulation. Foxp3DTR knock-in mice chronically infected with LCMV received DT for 10 d
(as in Fig. 2). (A) Viral load after 10 d of T reg cell depletion. (B) Dot plots show PD-1 expression on DbGP276-specific CD8 T cells. (C) PD-L1 expression on
splenic DCs after 7 d of T reg cell ablation. (D) Graph shows IFN- in serum before and at day 5 after initiation of DT treatment. Dashed line indicates the
limit of detection of this assay. (E) Representative dot plots show intracellular staining of LCMV nucleoprotein (NP) and PD-L1 expression on splenic DCs.
Staining of DCs in uninfected mice is shown as a control for specificity of NP staining and for basal PD-L1 expression. PD-L1 expression is shown as MFI
on infected (NP positive) or noninfected (NP negative) DCs. Data are a compilation of 3 experiments (A) or show representative results of 23 experiments
(BE) with 36 mice per group. Error bars indicate SEM. (A) Non-parametric Mann Whitney test, where NS = not significant. (D) Wilcoxon matched pairs
test, where *, P < 0.05. (E) Students t test, where **, P < 0.01; ***, P < 0.001.

B7-1 (CD80) and B7-2 (CD86) on CD11b+ and CD8+ CD4 T cells in our model, we depleted total CD4 cells in
DC subsets after T reg cell depletion (Fig. 6 A). To explore concert with T reg cell depletion. We observed that CD4
whether increased costimulation was necessary to rescue T cell depletion reduced DC activation, especially of the
virus-specific CD8+ T cells, we treated T reg cell ablated CD11b+ DCs (Fig. 6 E), a DC subset which efficiently pres-
mice with CTLA-4 Ig, a fusion protein which binds B7-1 ents antigens to CD4 T cells (Dudziak et al., 2007). Impor-
and B7-2 and thus blocks the B7/CD28 costimulatory path- tantly, CD4 T cell depletion completely abrogated the rescue
way. In mice subjected to T reg cell depletion in combina of LCMV-specific CD8 T cells observed upon T reg cell abla-
tion with CTLA4-Igmediated B7 blockade, the number of tion in chronically infected mice (Fig. 6, BD).These findings
functionally competent LCMV-specific CD8 T cells (Fig. 6, suggest that in chronically infected mice,T reg cells modulate
B and C) and their phenotype (Fig. 6 D) remained similar to DC activity and conventional CD4 T cells to maintain an
that of untreated mice. Thus, B7 costimulation blockade pre- exhausted state of virus-specific CD8 T cells. In the chronic
vented the increase in the response of virus-specific CD8 viral infection model used in our studies, CD4 T cells were
T cells that ensues upon T reg cell ablation in chronically in- transiently depleted before infection with LCMV cl-13 and
fected mice. These results imply a critical role for costimula- even after restoration of CD4 T cell numbers, LCMV-specific
tion in the rescue of exhausted T cell responses after T reg cell CD4 T cells were not detected (unpublished data), as it was
depletion and suggest an important role of DC activation in expected due to the infection of the thymus and negative se-
this process. lection (Matloubian et al., 1994). Therefore, it is highly un-
It was previously proposed that conventional CD4 T cells likely that cognate CD4 T cell help is involved in the rescue
mediate the DC activation that ensues after T reg cell deple- of CD8 T cells upon T reg cell depletion, at least in this model
tion (Kim et al., 2007). To address the role of conventional of chronic LCMV infection.

JEM Vol. 211, No. 9 1911


Figure 8. Essential role for PD-L1 block-
ade in viral control after T reg cell deple-
tion. Foxp3DTR knock-in mice chronically
infected with LCMV received DT for 10 d (as in
Fig. 2) in combination with PD-L1 blocking
antibody. PD-L1 antibody was administered
on days 1, 4, and 7 of DT treatment. (A) Viral
titers in serum before and after treatment.
(B) Fold reduction in serum viral titer after
treatment. (C) Absolute numbers of splenic
CD8 T cells that produce IFN- after in vitro
restimulation with LCMV peptides. (D) Abso-
lute numbers of CD8 T cells co-expressing
IFN- and TNF after in vitro restimulation
with GP276 LCMV peptide. (E) Representative
dot plots showing frequency of CD8 T cells
producing cytokines as in D. Data are a com-
pilation (A and B) or show representative
results of 3 independent experiments, with
46 mice per group. Error bars indicate SEM.
Non-parametric Mann Whitney test, where
*, P < 0.05; ***, P < 0.001; NS = not significant.

Failure to control virus by T reg cell ablation, despite (Fig. 7 B). PD-1 is triggered by TCR signaling; thus, as long
striking rescue of LCMV-specific CD8 T cell responses, as virus persists, LCMV-specific CD8 T cells maintain PD-1
may be due to PD-L1 up-regulation on infected cells expression (Blattman et al., 2009). Furthermore, T reg cell
We then tested whether the functional rescue of LCMV- ablation also led to increased expression of PD-L1 in both
specific CD8 T cells was associated with viral control. Unex- CD11b+ and CD8+ DCs (Fig. 7 C). To gain an insight into
pectedly, we did not observe a significant reduction in viral potential reasons for PD-L1 up-regulation, we assessed in-
load in chronically infected mice (Fig. 7 A) despite the im- flammatory cytokine levels in the serum of LCMV chroni-
pressive rescue of LCMV-specific CD8 T cell responses in- cally infected mice 5 d after T reg cell depletion. Most notably,
duced by T reg cell depletion. Antiviral T cells are known to we found increased levels of serum IFN- (Fig. 7 D), which
control virus by direct killing of infected cells and by produc- has been described to modulate PD-L1 expression (Dong
tion of cytokines (Kaech et al., 2002). Because T reg cell et al., 2002). Interestingly, we found higher PD-L1 expression
depletion resulted in a substantial increase in the number of on infected, LCMV nucleoprotein-positive DCs in compari-
LCMV-specific functional CD8 T cells (Fig. 3), we reasoned son with their uninfected counterparts (Fig. 7 E). In addition
that the lack of viral control might be due to an inhibitory to DCs, LCMV cl-13 replicates extensively in nonhemato-
process that limits killing of infected cells in vivo. poietic cells, such as fibroblastic reticular cells, endothelial
Although LCMV-specific T cells were rescued by T reg cells, and diverse parenchymal cell types, and PD-L1 expres-
cell ablation and regained function, they still remained PD-1+ sion in these cells is known to restrict viral control (Mueller

1912 T reg cells and PD-1 modulate T cell exhaustion | Penaloza-MacMaster et al.
Ar ticle

Figure 9. Transient T reg cell depletion


improves CD8 T cell rescue and viral con-
trol when combined with PD-L1 blockade
without causing overt disease in LCMV
chronically infected mice. (A) Foxp3DTR
knock-in mice chronically infected with LCMV
received continuous DT (on days 0, 1, 4, and 7)
and/or PD-L1 antibody (on days 1, 4, and 7),
as in Fig. 8. Mice weight after 11 d of treat-
ment, relative to initial weight before treat-
ment. (B) As in A, but mice received transient
DT treatment (on days 0, 1, and 4). Mice
weight after 12 d of treatment. (C) Represen-
tative dot plots show frequency of T reg cells
in PBMC before and after continuous or tran-
sient DT treatment. (D) Absolute numbers of
LCMV-specific (Db GP276) CD8 T cells within
the total splenic CD8 T cell population.
(E and F) Frequency of CD8 T cells producing
both IFN- and TNF (E) and absolute numbers
of CD8 T cells in the spleen that produce IFN-
after in vitro restimulation with LCMV peptide
GP276 (F). (G) Viral titers in serum before and
after treatment. (H) Fold reduction in serum
viral titer after treatment. A representative of
3 independent experiments is shown, with
6 mice per group. Error bars indicate SEM.
Non-parametric Mann Whitney test, where
*, P < 0.05; **, P < 0.01; ***, P < 0.001;
NS = not significant.

et al., 2007, 2010; Frebel et al., 2012). Thus, we reasoned that rescue. Because T reg cell depletion resulted in a greater num-
PD-1 interactions with the increased levels of PD-L1 ex- ber of functional virus-specific T cells when compared with
pressed by infected cells might oppose viral clearance by func- PD-1 blockade alone (Fig. 8, CE), and the combined ther-
tionally competent CD8 T cells in T reg celldepleted mice. apy further improved viral control (Fig. 8, A and B), our data
support the use of combination therapies based on modula-
Combining T reg cell depletion to blockade tion of T reg cell function and the PD-1 pathway to rescue
of the PD-1 pathway results in viral control exhausted T cell responses. Moreover, our data indicate that
To test the idea that PD-L1 expression could be protecting the magnitude of virus-specific CD8 T cell responses and de-
LCMV-infected cells from cytotoxic T cells, we administered crease in viral load are not always directly correlated. These
PD-L1 blocking antibodies to LCMV chronically infected results show that target cell elimination is affected both by in-
Foxp3DTR knock-in mice in conjunction with T reg cell de- trinsic cytotoxic potential of antigen-specific CD8 T cells and
pletion. Combining blockade of the PD-1 pathway with by inhibitory ligands such as PD-L1 displayed by target cells
T reg cell ablation resulted in a significant reduction of viral that may limit cytotoxicity.
load (Fig. 8 A), which was more pronounced than that upon
blockade of the PD-1 pathway alone (Fig. 8 B). These results Transient T reg cell depletion improves antiviral T cell
demonstrated an essential role for the PD-1PD-L1 pathway responses when combined to blockade the PD-1 pathway
in limiting viral control after CD8 T cell rescue. Importantly, In adult healthy mice, chronic T reg cell ablation triggers
the overall magnitude of multiple LCMV-specific CD8 T cell autoimmunity as early as 10 d after the start of DT administra-
responses observed after T reg cell depletion alone or in com- tion, and all mice succumb to overt disease by 3 wk of sustained
bination with the PD-1 pathway blockade was similar (Fig. 8, absence of T reg cells (Kim et al., 2007). Not unexpectedly,
CE), with the exception of an additive increase in the fre- sustained T reg cell ablation in LCMV chronically infected
quency of IFN- and TNF double-producing cells (Fig. 8 E). mice also induced immune-mediated wasting disease, which
Thus, these results demonstrated an essential role for the was further exacerbated by blockade of the PD-1 pathway
PD-1PD-L1 pathway in limiting viral control after CD8 T cell (Fig. 9 A). To establish the therapeutic utility of our approach,

JEM Vol. 211, No. 9 1913


we subjected chronically infected mice to a transient T reg cell In this study, we uncovered a major role of T reg cells in
depletion regimen where mice received three DT doses instead T cell exhaustion. Interestingly,T reg cells from LCMV chroni-
of four doses. This regimen of DT administration did not cally infected mice were phenotypically different from mice
cause an overt disease since these mice showed only minimal that had cleared LCMV infection or naive mice. During chronic
weight loss (Fig. 9 B). Although continuous DT treatment re- infection, T reg cells displayed an activated phenotype, as pre
sulted in nearly complete absence of T reg cells in Foxp3DTR viously reported (Punkosdy et al., 2011), with lower CD62L
knock-in mice, upon transient depletion the T reg cell subset and higher CD44 and CD69 expression, as well as an increase
had mostly recovered by the time of analysis (day 12; Fig. 9 C). in the inhibitory molecule PD-1. T reg cells from persistently
To ensure that wasting disease and mortality were associated infected animals also had increased expression of CD103,
with the chronic T reg cell depletion rather than nonspecific an important molecule for lymphocyte retention in skin and
DT toxicity, we treated LCMV chronically infected B6 mice other epithelial rich sites (Suffia et al., 2005). Furthermore, we
with DT as a control and did not observe significant weight observed increased expression of molecules that have been
loss or mortality. associated with T reg suppressive activity, such as ICOS (Chen
Transient T reg cell ablation in LCMV chronically infected et al., 2012), granzyme B (Gondek et al., 2005; Cao et al.,
mice did not significantly improve antiviral responses as a sin- 2007), and CD39, an ectoenzyme which catabolizes proin-
gle therapy, but improved T cell rescue and viral control when flammatory extracellular ATP (Borsellino et al., 2007).
combined to blockade of the PD-1 pathway (Fig. 9, DH). Most notably,T reg cells in chronically infected mice showed
Most importantly, combination treatment was more effective a twofold increase in the levels of CTLA-4, an inhibitory
for viremia control than antiPD-L1 treatment alone (Fig. 9, protein implicated in T reg cellmediated suppressive func-
G and H). Additionally, even partial (fivefold lower DT dose) tion (Wing et al., 2008). It has been shown that CTLA-4 in-
and transient T reg cell depletion was able to improve LCMV- teracts with and internalizes B7 molecules by trans-endocytosis
specific T cell responses obtained by PD-L1 blockade, and mice from the surface of the interacting cells, which results in APCs
remained healthy (unpublished data).Thus, careful optimization with lower stimulatory capacity for effector T cells (Qureshi
of the dose and timing of immunotherapy based on a combina- et al., 2011). In our model,T reg cell depletion leads to increased
tion of T reg cell and PD-1 signaling pathway can maximize B7 expression on DCs and we demonstrate that B7 stimula-
the desired T cell responses while minimizing adverse events. tion is essential for rescue of exhausted LCMV-specific CD8
T cells. T reg cells may affect DC activation status directly
DISCUSSION (possibly through CTLA-4 interactions with B7 molecules)
In this study, we show that T reg cells play a major role in and also indirectly (possibly through conventional CD4
T cell exhaustion during chronic viral infection. T reg cell T cells). It is important to point out that during chronic
ablation in LCMV chronically infected mice led to a striking LCMV infection CTLA-4 blockade does not rescue ex-
rescue of exhausted CD8 T cells. Virus-specific CD8 T cells hausted CD8 T cells (Barber et al., 2006), suggesting that
expanded 10100-fold after depletion of T reg cells, and res- CTLA-4 does not constitute a major nonredundant suppres-
cued CD8 T cells were able to degranulate, produce IFN-, sive mechanism in this model.
TNF, and granzyme B, and also kill LCMV peptide-pulsed We found that T reg cell control of CD8 T cell exhaustion
cells. Notably, the anti-LCMV CD8 T cell response obtained operates by restraining activation of DCs and conventional
upon T reg cell ablation was of much higher magnitude than CD4 T cells. After depletion of T reg cells we observed gener-
the rescue obtained in LCMV chronically infected mice by alized activation of CD4 T cells but no LCMV-specific CD4
PD-1PD-L1 blockade (Barber et al., 2006) or even combi- T cells that could provide cognate help to virus-specific CD8
nation of PD-1 pathway blockade with IL-2 therapy (West T cells. Most likely, when autoreactive CD4 T cells are released
et al., 2013). In spite of this, the antiviral CD8 T cell responses from T reg cell inhibition, they become activated and then
that ensued after T reg cell depletion were ineffective to re- trigger activation of DCs presenting autoantigens, as it has been
duce viral burden. When we tried to understand this unex- previously suggested (Kim et al., 2007). Consistent with the
pected lack of viral control, we detected up-regulation of the concept that CD4 T cells activate DCs, we found that deple-
inhibitory molecule PD-L1 after T reg cell ablation, especially tion of conventional CD4 T cells abrogated activation of splenic
in infected cells. Blockade of the PD-1 pathway in combina- CD11b+ DCs triggered by T reg cell ablation in LCMV chron-
tion with T reg cell depletion resulted in significant control of ically infected mice. Nonetheless, T reg cells might also affect
viral load, despite minimal improvement of CD8 T cell rescue DC activation status by other mechanisms as we observed that
when compared with T reg cell ablation alone. We propose depletion of conventional CD4 T cells could not completely
that the increased PD-L1 on target cells binds PD-1 on the abrogate activation of splenic CD8+ DCs after T reg cell abla-
rescued exhausted CD8 T cells and inhibits cytotoxic activity. tion. There might be important differences between DC sub-
These results support the hypothesis that T reg cells modulate sets with regard to interaction with T reg cells and how distinct
T cell exhaustion, but the PD-1PD-L1 pathway operates to DC subsets respond to the lack of T reg cells.
prevent destruction of target cells. Our data show that rescue Even though our results suggest that DC activation plays
of effective T cell responses can be improved by combining an important role in the rescue of exhausted CD8 T cells, it is
blockade of the PD-1 pathway to T reg cell depletion. important to take into account that the presence of conventional

1914 T reg cells and PD-1 modulate T cell exhaustion | Penaloza-MacMaster et al.
Ar ticle

CD4 T cells was required for the rescue of exhausted CD8 Thus, the presence of inflammatory cytokines in the serum is
T cells after T reg cell depletion. CD4 T cells can directly provide most likely the result of autoreactivity unleashed by T reg cell
factors for the rescue of exhausted CD8 T cells, such as IL-2 depletion and not a measure of antiviral T cell responses.Thus,
(Blattman et al., 2003; West et al., 2013) and IL-21 (Elsaesser it is likely that LCMV-specific CD8 T cells rescued by T reg
et al., 2009; Frhlich et al., 2009;Yi et al., 2009). Activation of cell ablation might be quite different from CD8 T cells rescued
CD4 T cells and DCs constitute a very intricate relationship by blockade of the PD-1 pathway, and better understanding of
and interfering with one will affect the other, thus, in our these differences might help shed light into potential molecu-
study we could not determine exactly the factors necessary lar mechanisms involved in rescue of T cell exhaustion.
for the rescue of exhausted CD8 T cells. Our data highlight Importantly, our study shows that even a 10100-fold in-
that manipulation of costimulatory pathways and identifica- crease in the number of functional viral-specific CD8+ T cells
tion of activating signals from CD4+ T cells would provide does not necessarily correlate with a decline in viral load. We
pivotal knowledge for treating chronic infections and cancer. show that the PD-1 pathway plays a fundamental role in viral
During chronic LCMV infection, antigen is abundant, yet clearance and we propose that PD-L1 expression in virus-
virus-specific CD8 T cells are suppressed and maintained by infected cells is a potent inhibitor of target cell elimination.We
minimal proliferation. Our data indicate that in chronic viral observed an increase in B7 and PD-L1 expression on DCs
infection T reg cell ablation promotes T cell activation but does after T reg cell depletion.We propose that LCMV-specific CD8
not elicit de novo T cell responses. Consistent with this notion, T cells are rescued because there is a net positive balance of
T reg cell depletion did not generate LCMV-specific CD8 signals from APCs presenting viral antigens to exhausted CD8
T cell responses in carrier mice infected with LCMV at birth T cells. However, the bulk viral burden in chronically infected
in which there are no LCMV-specific T cells in the periphery mice is largely due to LCMV infection of nonhematopoietic
due to negative selection in the thymus (Pircher et al., 1989; cells that express PD-L1 but no B7 molecules (Mueller et al.,
King et al., 1992). After 11 d of sustained T reg cell depletion 2007, 2010). Thus, even though LCMV-specific CD8 T cells
in LCMV carrier Foxp3DTR knock-in mice (>10,000 PFU/ml regain function after T reg cell depletion, there is no improve-
in serum), despite prominent T cell activation (85.73%, SD = ment in viral control because rescued CD8 T cells maintain
2.55 CD44hi CD8 T cells in DT treated vs. 34.4%, SD = PD-1 expression and their cytotoxic activity is inhibited by
2.81 in untreated mice), we were unable to detect any PD-L1expressing infected cells (Mueller et al., 2010; Frebel
LCMV-specific CD8 T cells (DbGP33+ or DbGP276+). This et al., 2012). Notably, PD-L1 also protects tumor cells from
important observation implies that antigen-specific T cells must CD8 T cellmediated killing (Iwai et al., 2002). When T reg
be present to elicit T cell responses by T reg cell manipulation. cell depletion was combined with PD-L1 blocking antibod-
In addition, our data suggest that the T cell activation that ies, rescue of exhausted T cells was only marginally improved
occurs upon T reg cell ablation is antigen-driven. In contrast as reflected by higher frequency of LCMV-specific CD8
to the impressive rescue of virus-specific CD8 T cells in LCMV T cells coproducing IFN- and TNF. However, T reg cell de-
chronically infected mice, T reg cell depletion in mice that pletion in combination with blockade of the PD-1 pathway
had cleared the infection did not affect the numbers and the resulted in marked improvement of viral control. In summary,
activation state of virus-specific CD8 T cells.This observation our data show that the magnitude of T cell responses and
reveals that T reg cells do not control maintenance of mem- effective control of infected cells (and possibly tumors) are not
ory cells and suggests that T reg cell manipulation would not always coupled together.
affect homeostasis of memory cells generated by vaccination PD-L1 is ubiquitously expressed and its expression is fur-
or acute infection. ther increased by several cytokines, including IFN- (Dong
In this study, we compared the effect of T reg cell deple- et al., 2002; Keir et al., 2008).We showed that T reg cell deple-
tion and blockade of the PD-1 pathway on the antiviral re- tion in LCMV chronically infected mice causes a systemic
sponse in chronically infected mice. T reg cell depletion in increase in IFN- as early as day 5 after treatment, which may
LCMV chronically infected mice resulted in an unprecedented modulate PD-L1 expression. In addition, functional rescue of
rescue of antiviral CD8 T cells. Distinctly, after T reg cell abla- CD8 T cells might directly induce PD-L1 up-regulation, es-
tion, LCMV-specific CD8 T cells re-expressed CD127, which pecially on infected cells presenting LCMV antigens due to
may be indicative of increased IL-2 signals (West et al., cognate interactions with T cells and local IFN- production.
2013). T cells integrate signals received from APCs and the It is important to point out that LCMV cl-13 and other viruses
environment to determine their differentiation program. Be- that cause chronic infections are relatively resistant to IFNs
sides increased systemic IFN-, we also observed an increase (Moskophidis et al., 1994) and IFNs may even have a detri-
in TNF (10140 pg/ml), MCP-1 (45360 pg/ml), and IL-6 mental effect on the course of the infection (Teijaro et al.,
(655 pg/ml), as it has been previously reported after T reg 2013; Wilson et al., 2013). Thus, therapies that promote cyto-
cell depletion in naive mice (Chinen et al., 2010). Interest- toxic T cell responses still need to ensure that further inhibi-
ingly, we could not detect any significant amount of inflam- tory mechanisms will not hamper destruction of target cells.
matory cytokines and chemokines (IFN-,TNF, MCP-1, and Reversal of T cell exhaustion is the ultimate goal of im-
IL-6) in the serum of LCMV chronically infected mice munotherapies aiming to treat chronically infected patients as
treated with antiPD-L1 blocking antibodies for 5 or 8 d. well as cancer patients.There have been many recent advances

JEM Vol. 211, No. 9 1915


in this field, and although single therapies such as blockade responses were generated by i.p. injection with 2 105 PFU of LCMV Arm,
of the PD-1 pathway and antiCTLA-4 antibody treatment which results in an acute infection that is cleared within 8 d. Chronic infec-
tions with exhausted T cell responses and lifelong viremia were induced by
have achieved success in clinical trials with advanced cancer
antibody-mediated transient depletion of CD4 T cells with 2 doses of 500 g
patients, most studies suggest that a combination of therapies GK1.5 (Bio X Cell) i.p., followed by i.v. injection with 2 106 PFU LCMV
may achieve better results. Rational combination of therapies cl-13 as described previously (Matloubian et al., 1994). To analyze memory
that operate through different mechanisms will probably im- T cell responses, we waited at least 100 d after LCMV Arm infection, and for
prove synergy to achieve better response rates as well as lon- exhausted T cell responses, we waited at least 45 d after LCMV cl-13 infec-
gevity of the immune response. Our study uncovers a potential tion. Titration of virus was performed on Vero cell monolayers as described
mechanism for synergistic effects of T reg cell depletion and previously (Ahmed et al., 1984). All mouse experiments were performed ac-
cording to institutional guidelines and were approved by the Emory Univer-
blockade of the PD-1 pathway.
sity Institutional Animal Care and Use Committee.
Recent evidence suggests that antiCTLA-4 may function
by depletion of T reg cells at the tumor site (Bulliard et al.,
Antibody treatments and T reg cell depletion. 500 g CTLA-4 Ig (gift
2013; Selby et al., 2013; Simpson et al., 2013). Importantly, it
from the Ford and Larsen laboratory, Emory University, Atlanta, GA) was
was recently shown that combining antiCTLA-4 to PD-1 injected i.p., on days 2, 0, 2, 4, 6, 8, and 10 of T reg cell ablation. CD4+
blockade resulted in >40% objective responses in advanced T cell depletion was achieved by i.p. injection of 500 g GK1.5 on days 2
melanoma patients (Wolchok et al., 2013). In this study, among and 1 of T reg cell ablation. 200 g PD-L1 blocking antibody (10F.9G2)
responders, most patients had >80% tumor regression, which was administered i.p. on days 1, 4, and 7 after T reg cell ablation. Each
exceeds the rate of responses reported previously in monother- mouse received 1 g DT (Sigma-Aldrich) i.p. (50 g/kg).
apy trials. Thus, our results provide important concepts that
may have direct relevance to cancer treatment. Nevertheless, ad- Cytotoxicity activity by 51 chromium-release assay. MC57 mouse fi-
ditional studies need to be done to determine which chronic broblast cell targets were coated with a mix of GP33, GP276, and NP396
peptides or no peptide, and labeled with 350 Ci 51-Cr. Target cells were
infections and cancers would be responsive to this combina-
incubated for 6 h with different amounts of effector splenic CD8+ T cells
torial immunotherapy involving T reg cell depletion and enriched using mouse CD8 beads (Miltenyi Biotec). Absolute numbers of
blockade of the PD-1 pathway because there may be hetero- viral-specific CD8+ T cells was retroactively calculated and plotted. E:T = ef-
geneity in the response to such therapies (Bos et al., 2013). fector to target ratio. MC57 cells with 1% Triton X-100 were used as positive
In LCMV chronically infected mice, virus is widespread and control (total release), and MC57 cells without effectors were used to calcu-
there is a delicate balance between an effective immune response late spontaneous release.
that controls viral load and overt immunopathology. Using
Antibodies and flow cytometry. Single cell suspensions were obtained
Foxp3DTR knock-in mice, we were able to achieve very efficient
from blood, spleen, lungs, liver, kidney, and gut as previously described
depletion of T reg cells.Although the complete absence of T reg (Masopust et al., 2001). For analysis of dendritic cells, spleens were digested
cells may increase T cell rescue, serious adverse effects may limit with 0.4 U/ml collagenase D (Roche) for 30 min at 37C. Single cell sus-
its application in a clinical setting. We show that transient T reg pensions were stained with anti-CD8 (536.7), -CD4 (RM4-5), -CD25
cell depletion, as a single therapy, failed to rescue antiviral responses, (PC61), CD40 (3/23), CD80 (16-10A1), CD86 (GL1), -CD107a (1D4B),
but when combined to PD-1 pathway blockade, transient T reg -CD107b (ABL-93),V 5.1 5.2 (MR9-4), and Ki67 (B56; from BD); -ICOS
cell depletion improved both CD8 T cell responses and viral con- (7E.17G9), PD-L1 (MIH5), -CD11c (N418), -CD11b (M1/70), -CD127
(A7R34), -MHCI (AF6-88.5.5.3), and -Foxp3 (FJK-16s; from eBioscience);
trol.Thus, milder T reg cell depletion regimens, which could be
-CD44 (IM7) and PD-1 (RMP1-30; from BioLegend); and granzyme B
achieved in patients with antibody-based depletion strategies, (MHGB04; Invitrogen). Anti-LCMV NP antibody was a gift from the
might improve immune responses induced by other therapies Buchmeier Laboratory (University of California, Irvine, Irvine, CA). Dead
without overt adverse effects. Alternative approaches that would cells were excluded by gating out cells positive for Live/Dead fixable dead cell
preferentially affect a subset of T reg cells, such as CCR4+ stain (Invitrogen). LCMV MHC class I tetramers were prepared and used as
T reg cells which are predominant at the tumor site (Sugiyama previously described (Wherry et al., 2003). The LCMV MHC class II tetra-
et al., 2013) or Nrp1+ T reg cells which are recruited to VEGF- mer (I-Ab GP61-80) was obtained from the National Institutes of Health
tetramer facility and stains were performed at 37C for 3 h, gently mixing
producing tumors (Hansen et al., 2012), have also been pro- cells every 30 min. LCMV-specific responses were assessed by restimulating
posed as potential strategies that would evoke immune responses splenocytes with 0.1 g/ml GP33, NP396, GP276, NP118, or NP235 LCMV
while minimizing autoimmunity. peptides in the presence of brefeldin and monensin for 5 h at 37C. Intracel-
In summary, our studies demonstrate that T reg cells control lular staining for IFN-, TNF, Ki67, and granzyme B were performed with
chronically stimulated exhausted CD8 T cells during persistent the Cytofix/Cytoperm kit (BD). Intracellular staining of Foxp3 was per-
viral infection.We show that this T reg cellmediated control of formed according to manufacturers instructions (eBioscience). Serum cyto-
exhausted CD8 T cells involves restraint of B7 costimulation kines were measured by cytometric bead array according to manufacturers
instructions (BD). Samples were acquired with a FACSCanto or LSRII (BD)
and of help from conventional CD4 T cells. The findings pre- and analyzed using FlowJo (Tree Star).
sented here also underscore a critical role of PD-L1 in deter-
mining viral control after T cell rescue from an exhausted state.
Statistical analysis. Statistical analysis was performed on Prism software
MATERIALS AND METHODS (GraphPad Software).
Mice and infections. 48-wk-old homozygous Foxp3DTR knock-in mice
on a C57BL/6 background (Kim et al., 2007) or C57BL/6 (The Jackson We thank Dr. Rouse, Dr. Pulendran, and members of the Ahmed laboratory for
Laboratory) were infected with LCMV Arm or LCMV cl-13. Memory T cell helpful discussion.

1916 T reg cells and PD-1 modulate T cell exhaustion | Penaloza-MacMaster et al.
Ar ticle

This work was supported by National Institutes of Health grants R01 AI3004 differential ICOS expression. J. Immunol. 188:16981707. http://dx.doi
(R. Ahmed), P01 AI080192 (R. Ahmed and G.J. Freeman), P01 AI056299 (R. Ahmed, .org/10.4049/jimmunol.1102448
G.J. Freeman, and A.H. Sharpe), and R37 AI034206 (A.Y. Rudensky), by the Ludwig Chinen, T., P.Y.Volchkov, A.V. Chervonsky, and A.Y. Rudensky. 2010. A criti-
Cancer Research (A.Y. Rudensky), and by the Cancer Research Institutes Irvington cal role for regulatory T cellmediated control of inflammation in the
Institute Fellowship Program (A.O. Kamphorst). A.Y. Rudensky is an investigator with absence of commensal microbiota. J. Exp. Med. 207:23232330. http://
the Howard Hughes Medical Institute. dx.doi.org/10.1084/jem.20101235
R. Ahmed, A.H. Sharpe, and G.J. Freeman hold patents and receive patent royal Dietze, K.K., G. Zelinskyy, K. Gibbert, S. Schimmer, S. Francois, L. Myers, T.
ties related to the PD-1 inhibitory pathway. A.H. Sharpe and G.J. Freeman are scien Sparwasser, K.J. Hasenkrug, and U. Dittmer. 2011. Transient depletion
tific founders of Costim Pharmaceuticals. R. Ahmed, A.H. Sharpe, and G.J. Freeman of regulatory T cells in transgenic mice reactivates virus-specific CD8+
declare no additional financial interests. The remaining authors declare no T cells and reduces chronic retroviral set points. Proc. Natl. Acad. Sci. USA.
competing financial interests. 108:24202425. http://dx.doi.org/10.1073/pnas.1015148108
Dittmer, U., H. He, R.J. Messer, S. Schimmer, A.R. Olbrich, C. Ohlen,
Submitted: 13 December 2013 P.D. Greenberg, I.M. Stromnes, M. Iwashiro, S. Sakaguchi, et al. 2004.
Accepted: 10 July 2014 Functional impairment of CD8+ T cells by regulatory T cells during
persistent retroviral infection. Immunity. 20:293303. http://dx.doi.org/
10.1016/S1074-7613(04)00054-8
REFERENCES Dong, H., S.E. Strome, D.R. Salomao, H. Tamura, F. Hirano, D.B. Flies, P.C.
Ahmed, R., A. Salmi, L.D. Butler, J.M. Chiller, and M.B. Oldstone. 1984. Roche, J. Lu, G. Zhu, K. Tamada, et al. 2002. Tumor-associated B7-H1
Selection of genetic variants of lymphocytic choriomeningitis virus in promotes T-cell apoptosis: a potential mechanism of immune evasion.
spleens of persistently infected mice. Role in suppression of cytotoxic Nat. Med. 8:793800. http://dx.doi.org/10.1038/nm730
T lymphocyte response and viral persistence. J. Exp. Med. 160:521540. Dudziak, D.,A.O. Kamphorst, G.F. Heidkamp,V.R. Buchholz, C.Trumpfheller,
http://dx.doi.org/10.1084/jem.160.2.521 S. Yamazaki, C. Cheong, K. Liu, H.W. Lee, C.G. Park, et al. 2007.
Aubert, R.D., A.O. Kamphorst, S. Sarkar, V. Vezys, S.J. Ha, D.L. Barber, L. Ye, Differential antigen processing by dendritic cell subsets in vivo. Science.
A.H. Sharpe, G.J. Freeman, and R. Ahmed. 2011. Antigen-specific CD4
315:107111. http://dx.doi.org/10.1126/science.1136080
T-cell help rescues exhausted CD8 T cells during chronic viral infec-
Elsaesser, H., K. Sauer, and D.G. Brooks. 2009. IL-21 is required to control
tion. Proc. Natl. Acad. Sci. USA. 108:2118221187. http://dx.doi.org/
chronic viral infection. Science. 324:15691572. http://dx.doi.org/10
10.1073/pnas.1118450109
.1126/science.1174182
Barber, D.L., E.J. Wherry, D. Masopust, B. Zhu, J.P. Allison, A.H. Sharpe, G.J.
Fourcade, J., Z. Sun, M. Benallaoua, P. Guillaume, I.F. Luescher, C. Sander,
Freeman, and R. Ahmed. 2006. Restoring function in exhausted CD8
J.M. Kirkwood, V. Kuchroo, and H.M. Zarour. 2010. Upregulation of
T cells during chronic viral infection. Nature. 439:682687. http://dx.doi
Tim-3 and PD-1 expression is associated with tumor antigen-specific
.org/10.1038/nature04444
CD8+ T cell dysfunction in melanoma patients. J. Exp. Med. 207:2175
Belkaid,Y., and K.Tarbell. 2009. Regulatory T cells in the control of host-mi-
2186. http://dx.doi.org/10.1084/jem.20100637
croorganism interactions (*). Annu. Rev. Immunol. 27:551589. http://
Frebel, H.,V. Nindl, R.A. Schuepbach, T. Braunschweiler, K. Richter, J.Vogel,
dx.doi.org/10.1146/annurev.immunol.021908.132723
C.A. Wagner, D. Loffing-Cueni, M. Kurrer, B. Ludewig, and A. Oxenius.
Blackburn, S.D., H. Shin,W.N. Haining,T. Zou, C.J.Workman, A. Polley, M.R.
Betts, G.J. Freeman, D.A.Vignali, and E.J. Wherry. 2009. Coregulation of 2012. Programmed death 1 protects from fatal circulatory failure during
CD8+T cell exhaustion by multiple inhibitory receptors during chronic viral systemic virus infection of mice. J. Exp. Med. 209:24852499. http://
infection. Nat. Immunol. 10:2937. http://dx.doi.org/10.1038/ni.1679 dx.doi.org/10.1084/jem.20121015
Blattman, J.N., J.M. Grayson, E.J. Wherry, S.M. Kaech, K.A. Smith, and R. Frhlich, A., J. Kisielow, I. Schmitz, S. Freigang, A.T. Shamshiev, J. Weber, B.J.
Ahmed. 2003.Therapeutic use of IL-2 to enhance antiviralT-cell responses Marsland, A. Oxenius, and M. Kopf. 2009. IL-21R on T cells is critical
in vivo. Nat. Med. 9:540547. http://dx.doi.org/10.1038/nm866 for sustained functionality and control of chronic viral infection. Science.
Blattman, J.N., E.J. Wherry, S.J. Ha, R.G. van der Most, and R. Ahmed. 2009. 324:15761580. http://dx.doi.org/10.1126/science.1172815
Impact of epitope escape on PD-1 expression and CD8 T-cell exhaus- Gondek, D.C., L.F. Lu, S.A. Quezada, S. Sakaguchi, and R.J. Noelle. 2005. Cutting
tion during chronic infection. J.Virol. 83:43864394. http://dx.doi.org/ edge: contact-mediated suppression by CD4+CD25+ regulatory cells involves
10.1128/JVI.02524-08 a granzyme B-dependent, perforin-independent mechanism. J. Immunol.
Borsellino, G., M. Kleinewietfeld, D. Di Mitri, A. Sternjak, A. Diamantini, R. 174:17831786. http://dx.doi.org/10.4049/jimmunol.174.4.1783
Giometto, S. Hpner, D. Centonze, G. Bernardi, M.L. DellAcqua, et al. Hansen, W., M. Hutzler, S. Abel, C. Alter, C. Stockmann, S. Kliche, J. Albert,
2007. Expression of ectonucleotidase CD39 by Foxp3+ Treg cells: hy- T. Sparwasser, S. Sakaguchi, A.M. Westendorf, et al. 2012. Neuropilin
drolysis of extracellular ATP and immune suppression. Blood. 110:1225 1 deficiency on CD4+Foxp3+ regulatory T cells impairs mouse mela-
1232. http://dx.doi.org/10.1182/blood-2006-12-064527 noma growth. J. Exp. Med. 209:20012016. http://dx.doi.org/10
Bos, P.D., G. Plitas, D. Rudra, S.Y. Lee, and A.Y. Rudensky. 2013. Transient .1084/jem.20111497
regulatory T cell ablation deters oncogene-driven breast cancer and en- Iwai, Y., M. Ishida, Y. Tanaka, T. Okazaki, T. Honjo, and N. Minato. 2002.
hances radiotherapy. J. Exp. Med. 210:24352466. http://dx.doi.org/10 Involvement of PD-L1 on tumor cells in the escape from host immune
.1084/jem.20130762 system and tumor immunotherapy by PD-L1 blockade. Proc. Natl.Acad. Sci.
Bulliard, Y., R. Jolicoeur, M. Windman, S.M. Rue, S. Ettenberg, D.A. Knee, USA. 99:1229312297. http://dx.doi.org/10.1073/pnas.192461099
N.S.Wilson, G. Dranoff, and J.L. Brogdon. 2013.Activating Fc receptors Jin, H.T., A.C. Anderson, W.G. Tan, E.E. West, S.J. Ha, K. Araki, G.J. Freeman,
contribute to the antitumor activities of immunoregulatory receptor- V.K. Kuchroo, and R. Ahmed. 2010. Cooperation of Tim-3 and PD-1 in
targeting antibodies. J. Exp. Med. 210:16851693. http://dx.doi.org/10 CD8 T-cell exhaustion during chronic viral infection. Proc. Natl.Acad. Sci.
.1084/jem.20130573 USA. 107:1473314738. http://dx.doi.org/10.1073/pnas.1009731107
Butler, N.S., J. Moebius, L.L. Pewe, B. Traore, O.K. Doumbo, L.T. Tygrett, T.J. Kaech, S.M., E.J. Wherry, and R. Ahmed. 2002. Effector and memory T-cell
Waldschmidt, P.D. Crompton, and J.T. Harty. 2012. Therapeutic blockade differentiation: implications for vaccine development. Nat. Rev. Immunol.
of PD-L1 and LAG-3 rapidly clears established blood-stage Plasmodium in- 2:251262. http://dx.doi.org/10.1038/nri778
fection. Nat. Immunol. 13:188195. http://dx.doi.org/10.1038/ni.2180 Kaech, S.M., J.T. Tan, E.J. Wherry, B.T. Konieczny, C.D. Surh, and R. Ahmed.
Cao, X., S.F. Cai, T.A. Fehniger, J. Song, L.I. Collins, D.R. Piwnica-Worms, 2003. Selective expression of the interleukin 7 receptor identifies effec-
and T.J. Ley. 2007. Granzyme B and perforin are important for regulatory tor CD8 T cells that give rise to long-lived memory cells. Nat. Immunol.
T cell-mediated suppression of tumor clearance. Immunity. 27:635646. 4:11911198.
http://dx.doi.org/10.1016/j.immuni.2007.08.014 Keir, M.E., M.J. Butte, G.J. Freeman, and A.H. Sharpe. 2008. PD-1 and its
Chen,Y., S. Shen, B.K. Gorentla, J. Gao, and X.P. Zhong. 2012. Murine regu- ligands in tolerance and immunity. Annu. Rev. Immunol. 26:677704.
latory T cells contain hyperproliferative and death-prone subsets with http://dx.doi.org/10.1146/annurev.immunol.26.021607.090331

JEM Vol. 211, No. 9 1917


Kim, J.M., J.P. Rasmussen, and A.Y. Rudensky. 2007. Regulatory T cells pre- contribute to peripheral CD8+ T-cell tolerance induced by steady-state
vent catastrophic autoimmunity throughout the lifespan of mice. Nat. dendritic cells. Proc. Natl. Acad. Sci. USA. 107:199203. http://dx.doi
Immunol. 8:191197. http://dx.doi.org/10.1038/ni1428 .org/10.1073/pnas.0910620107
King, C.C., B.D. Jamieson, K. Reddy, N. Bali, R.J. Concepcion, and R. Schmitz, I., C. Schneider, A. Frhlich, H. Frebel, D. Christ, W.J. Leonard, T.
Ahmed. 1992.Viral infection of the thymus. J.Virol. 66:31553160. Sparwasser, A. Oxenius, S. Freigang, and M. Kopf. 2013. IL-21 restricts
Lau, L.L., B.D. Jamieson, T. Somasundaram, and R. Ahmed. 1994. Cytotoxic virus-driven Treg cell expansion in chronic LCMV infection. PLoS
T-cell memory without antigen. Nature. 369:648652. http://dx.doi.org/ Pathog. 9:e1003362. http://dx.doi.org/10.1371/journal.ppat.1003362
10.1038/369648a0 Selby, M.J., J.J. Engelhardt, M. Quigley, K.A. Henning,T. Chen, M. Srinivasan,
Li, S., E.J. Gowans, C. Chougnet, M. Plebanski, and U. Dittmer. 2008. Natural and A.J. Korman. 2013. Anti-CTLA-4 antibodies of IgG2a isotype en-
regulatory T cells and persistent viral infection. J.Virol. 82:2130. http:// hance antitumor activity through reduction of intratumoral regulatory
dx.doi.org/10.1128/JVI.01768-07 T cells. Cancer Immunol Res. 1:3242. http://dx.doi.org/10.1158/2326-
Lichterfeld, M., D.E. Kaufmann, X.G. Yu, S.K. Mui, M.M. Addo, M.N. 6066.CIR-13-0013
Johnston, D. Cohen, G.K. Robbins, E. Pae, G. Alter, et al. 2004. Loss of Simpson, T.R., F. Li, W. Montalvo-Ortiz, M.A. Sepulveda, K. Bergerhoff,
HIV-1specific CD8+ T cell proliferation after acute HIV-1 infection F. Arce, C. Roddie, J.Y. Henry, H. Yagita, J.D. Wolchok, et al. 2013.
and restoration by vaccine-induced HIV-1specific CD4+ T cells. J. Exp. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines
Med. 200:701712. http://dx.doi.org/10.1084/jem.20041270 the efficacy of antiCTLA-4 therapy against melanoma. J. Exp. Med.
Lund, J.M., L. Hsing, T.T. Pham, and A.Y. Rudensky. 2008. Coordination of 210:16951710. http://dx.doi.org/10.1084/jem.20130579
early protective immunity to viral infection by regulatory T cells. Science. Suffia, I., S.K. Reckling, G. Salay, and Y. Belkaid. 2005. A role for CD103 in the
320:12201224. http://dx.doi.org/10.1126/science.1155209 retention of CD4+CD25+ Treg and control of Leishmania major infec-
Masopust, D., V. Vezys, A.L. Marzo, and L. Lefranois. 2001. Preferential lo- tion. J. Immunol. 174:54445455. http://dx.doi.org/10.4049/jimmunol
calization of effector memory cells in nonlymphoid tissue. Science. .174.9.5444
291:24132417. http://dx.doi.org/10.1126/science.1058867 Sugiyama, D., H. Nishikawa, Y. Maeda, M. Nishioka, A. Tanemura, I. Katayama,
Matloubian, M., R.J. Concepcion, and R. Ahmed. 1994. CD4+ T cells are S. Ezoe,Y. Kanakura, E. Sato,Y. Fukumori, et al. 2013. Anti-CCR4 mAb
required to sustain CD8+ cytotoxic T-cell responses during chronic viral selectively depletes effector-type FoxP3+CD4+ regulatory T cells, evok-
infection. J.Virol. 68:80568063. ing antitumor immune responses in humans. Proc. Natl. Acad. Sci. USA.
Moskophidis, D., M. Battegay, M.A. Bruendler, E. Laine, I. Gresser, and R.M. 110:1794517950. http://dx.doi.org/10.1073/pnas.1316796110
Zinkernagel. 1994. Resistance of lymphocytic choriomeningitis virus to Teijaro, J.R., C. Ng, A.M. Lee, B.M. Sullivan, K.C. Sheehan, M. Welch, R.D.
/ interferon and to interferon. J.Virol. 68:19511955. Schreiber, J.C. de la Torre, and M.B. Oldstone. 2013. Persistent LCMV
Mueller, S.N., M. Matloubian, D.M. Clemens, A.H. Sharpe, G.J. Freeman,
infection is controlled by blockade of type I interferon signaling. Science.
S. Gangappa, C.P. Larsen, and R. Ahmed. 2007. Viral targeting of fibro-
340:207211. http://dx.doi.org/10.1126/science.1235214
blastic reticular cells contributes to immunosuppression and persistence
Topalian, S.L., C.G. Drake, and D.M. Pardoll. 2012. Targeting the PD-1/
during chronic infection. Proc. Natl. Acad. Sci. USA. 104:1543015435.
B7-H1(PD-L1) pathway to activate anti-tumor immunity. Curr. Opin.
http://dx.doi.org/10.1073/pnas.0702579104
Immunol. 24:207212. http://dx.doi.org/10.1016/j.coi.2011.12.009
Mueller, S.N., V.K. Vanguri, S.J. Ha, E.E. West, M.E. Keir, J.N. Glickman,
West, E.E., H.T. Jin, A.U. Rasheed, P. Penaloza-Macmaster, S.J. Ha, W.G. Tan,
A.H.Sharpe,and R.Ahmed.2010.PD-L1 has distinct functions in hemato
B. Youngblood, G.J. Freeman, K.A. Smith, and R. Ahmed. 2013. PD-L1
poietic and nonhematopoietic cells in regulating T cell responses during
blockade synergizes with IL-2 therapy in reinvigorating exhausted T cells.
chronic infection in mice. J. Clin. Invest. 120:25082515. http://dx.doi
J. Clin. Invest. 123:26042615. http://dx.doi.org/10.1172/JCI67008
.org/10.1172/JCI40040
Wherry, E.J. 2011. T cell exhaustion. Nat. Immunol. 12:492499. http://
Murali-Krishna, K., L.L. Lau, S. Sambhara, F. Lemonnier, J.Altman, and R.Ahmed.
dx.doi.org/10.1038/ni.2035
1999. Persistence of memory CD8 T cells in MHC class I-deficient mice.
Science. 286:13771381. http://dx.doi.org/10.1126/science.286.5443.1377 Wherry, E.J., and R. Ahmed. 2004. Memory CD8 T-cell differentiation
Pace, L., A. Tempez, C. Arnold-Schrauf, F. Lemaitre, P. Bousso, L. Fetler, T. during viral infection. J. Virol. 78:55355545. http://dx.doi.org/10
Sparwasser, and S. Amigorena. 2012. Regulatory T cells increase the .1128/JVI.78.11.5535-5545.2004
avidity of primary CD8+ T cell responses and promote memory. Science. Wherry, E.J., J.N. Blattman, K. Murali-Krishna, R. van der Most, and R. Ahmed.
338:532536. http://dx.doi.org/10.1126/science.1227049 2003. Viral persistence alters CD8 T-cell immunodominance and tissue
Pircher, H., K. Brki, R. Lang, H. Hengartner, and R.M. Zinkernagel. distribution and results in distinct stages of functional impairment. J.Virol.
1989. Tolerance induction in double specific T-cell receptor transgenic 77:49114927. http://dx.doi.org/10.1128/JVI.77.8.4911-4927.2003
mice varies with antigen. Nature. 342:559561. http://dx.doi.org/10 Wilson, E.B., D.H. Yamada, H. Elsaesser, J. Herskovitz, J. Deng, G. Cheng,
.1038/342559a0 B.J. Aronow, C.L. Karp, and D.G. Brooks. 2013. Blockade of chronic
Punkosdy, G.A., M. Blain, D.D. Glass, M.M. Lozano, L. OMara, J.P. Dudley, type I interferon signaling to control persistent LCMV infection. Science.
R. Ahmed, and E.M. Shevach. 2011. Regulatory T-cell expansion dur- 340:202207. http://dx.doi.org/10.1126/science.1235208
ing chronic viral infection is dependent on endogenous retroviral super Wing, K.,Y. Onishi, P. Prieto-Martin, T.Yamaguchi, M. Miyara, Z. Fehervari,
antigens. Proc. Natl. Acad. Sci. USA. 108:36773682. http://dx.doi.org/ T. Nomura, and S. Sakaguchi. 2008. CTLA-4 control over Foxp3+ regu-
10.1073/pnas.1100213108 latory T cell function. Science. 322:271275. http://dx.doi.org/10.1126/
Qureshi, O.S., Y. Zheng, K. Nakamura, K. Attridge, C. Manzotti, E.M. science.1160062
Schmidt, J. Baker, L.E. Jeffery, S. Kaur, Z. Briggs, et al. 2011. Trans- Wolchok, J.D., H. Kluger, M.K. Callahan, M.A. Postow, N.A. Rizvi, A.M.
endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic Lesokhin, N.H. Segal, C.E. Ariyan, R.A. Gordon, K. Reed, et al. 2013.
function of CTLA-4. Science. 332:600603. http://dx.doi.org/10.1126/ Nivolumab plus ipilimumab in advanced melanoma. N. Engl. J. Med.
science.1202947 369:122133. http://dx.doi.org/10.1056/NEJMoa1302369
Sakaguchi, S., T. Yamaguchi, T. Nomura, and M. Ono. 2008. Regulatory Yi, J.S., M. Du, and A.J. Zajac. 2009.A vital role for interleukin-21 in the control
T cells and immune tolerance. Cell. 133:775787. http://dx.doi.org/ of a chronic viral infection. Science. 324:15721576. http://dx.doi.org/
10.1016/j.cell.2008.05.009 10.1126/science.1175194
Sakuishi, K., L. Apetoh, J.M. Sullivan, B.R. Blazar, V.K. Kuchroo, and A.C. Zajac, A.J., J.N. Blattman, K. Murali-Krishna, D.J. Sourdive, M. Suresh, J.D.
Anderson. 2010. Targeting Tim-3 and PD-1 pathways to reverse T cell Altman, and R. Ahmed. 1998. Viral immune evasion due to persistence
exhaustion and restore anti-tumor immunity. J. Exp. Med. 207:2187 of activated T cells without effector function. J. Exp. Med. 188:2205
2194. http://dx.doi.org/10.1084/jem.20100643 2213. http://dx.doi.org/10.1084/jem.188.12.2205
Schildknecht, A., S. Brauer, C. Brenner, K. Lahl, H. Schild, T. Sparwasser, H.C. Zou, W. 2006. Regulatory T cells, tumour immunity and immunotherapy.
Probst, and M. van den Broek. 2010. FoxP3+ regulatory T cells essentially Nat. Rev. Immunol. 6:295307. http://dx.doi.org/10.1038/nri1806

1918 T reg cells and PD-1 modulate T cell exhaustion | Penaloza-MacMaster et al.
INSIGHTS

CCR4 dr ives ATLL jail break


Adult T cell leukemia/lymphoma (ATLL), caused by human T cell lymphotropic virus 1 (HTLV-1), is
an aggressive cancer that is refractory to current therapies. The long latency and low overall penetrance of
ATLL in HTLV-1infected individuals infers the need for cooperating events, which include somatic
JAK3, NOTCH1, and FAS mutations. While overexpression of CCR4 is a hallmark of ATLL, it is not clear
whether dysregulation of CCR4 function contributes to disease pathogenesis. In this issue, Nakagawa et
al. report recurrent somatic mutations in the CCR4 chemokine receptor in ~25% of ATLL cases, impli-
cating these mutations in ATLL pathogenesis.
The authors performed RNA transcriptome analysis of two ATLL cases and targeted sequencing of addi- Insight from
tional ATLL patient samples and cell lines. Remarkably, the CCR4 mutations found in primary ATLL speci- Kevin Shannon
mens were heterozygous and introduced missense or truncating mutations in a conserved carboxy-terminal
domain of CCR4 involved in negative regulation. Together, these findings suggested a dominant gain-of-function mechansim of
action. Indeed, elegant functional studies revealed defective internalization of these mutant CCR4 proteins as well as enhanced
migration and chemotaxis in response to chemokines. The authors also demonstrated hyperactive PI3 kinase/Akt signaling in
ATLL cells expressing CCR4 mutant proteins.
So, how do CCR4 mutations promote malignant growth? The authors provide two logical and nonexclusive explana-
tions. First, these mutations might enable ATLL cells to migrate to and colonize niches in tissues such as skin and lymph nodes
that are favorable for cancer cell survival and proliferation. If this
idea is correct, it is possible that patients with and without CCR4
mutations will exhibit specific patterns of tissue involvement and
disease evolution. Second, dysregulated PI3K signaling down-
stream of mutant CCR4 might be the key biochemical driver con-
tributing to clonal selection of ATLL cells. Given this underlying
biology, it is reasonable to speculate that seed and soil both
contribute to the aberrant growth of ATLL tumors with somatic
CCR4 mutations. For example, because CCR4 mutations render
PI3K signaling hypersensitive to chemokine stimulation, specific
tissue microenvironments likely favor ATLL growth through para-
crine mechanisms.
An exciting aspect of these new mechanistic insights is their
potential for clinical translation. A first generation anti-CCR4
antibody called KW-0761 is showing promise in early phase clinical
trials. It will be interesting to determine whether mutant CCR4 is
Schematic of CCR4 mutant isoforms in ATLL a predictive biomarker of sensitivity to this and other anti-CCR4
agents. Deep genomic analysis of tumors with CCR4 mutations
that relapse after an initial response will provide additional insights. PI3 kinase inhibitorsboth alone and in combination with
other agentsare another potential therapeutic strategy for improving outcomes in this relentless cancer.
Nakagawa, M., et al. 2014. J. Exp. Med. http://dx.doi.org/10.1084/jem.20140987.

Kevin M. Shannon, University of California, San Francisco: shannonk@peds.ucsf.edu


Brief Definitive Report

Gain-of-function CCR4 mutations in adult


T cell leukemia/lymphoma
Masao Nakagawa,* Roland Schmitz,* Wenming Xiao, Carolyn K. Goldman,
Weihong Xu, Yandan Yang, Xin Yu, Thomas A. Waldmann,**
and Louis M. Staudt**

Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health,
Bethesda, MD 20892

Adult T cell leukemia/lymphoma (ATLL) is an aggressive malignancy caused by human T cell


lymphotropic virus type-I (HTLV-I) without curative treatment at present. To illuminate the
pathogenesis of ATLL we performed whole transcriptome sequencing of purified ATLL
patient samples and discovered recurrent somatic mutations in CCR4, encoding CC chemo-
kine receptor 4. CCR4 mutations were detected in 14/53 ATLL samples (26%) and con-
sisted exclusively of nonsense or frameshift mutations that truncated the coding region at
C329, Q330, or Y331 in the carboxy terminus. Functionally, the CCR4-Q330 nonsense
isoform was gain-of-function because it increased cell migration toward the CCR4 ligands
CCL17 and CCL22, in part by impairing receptor internalization. This mutant enhanced
PI(3) kinase/AKT activation after receptor engagement by CCL22 in ATLL cells and con-
ferred a growth advantage in long-term in vitro cultures. These findings implicate somatic
gain-of-function CCR4 mutations in the pathogenesis of ATLL and suggest that inhibition
of CCR4 signaling might have therapeutic potential in this refractory malignancy.

CORRESPONDENCE Adult T cell leukemia/lymphoma (ATLL) is (Ishida et al., 2003). Interestingly, the most
Louis M. Staudt: one of the most aggressive forms of peripheral frequent sites of ATLL involvement are lymph
lstaudt@mail.nih.gov
T cell lymphoma, with a median survival of nodes and skin (Campo et al., 2011), where den-
Abbreviations used: ATLL, <1 yr with current therapy, which consists pri- dritic cells, M2-phenotype macrophages, Lang-
adult T cell leukemia/ marily of cytotoxic chemotherapy (Campo et al., erhans cells, and cutaneous venules can produce
lymphoma; CCR4, CC 2011). Molecular analyses of ATLL cells re- CCL17 and/or CCL22 (Campbell et al., 1999;
chemokine receptor 4; GPCR,
G proteincoupled receptor; vealed that high expression of CC chemokine Vissers et al., 2001; Vulcano et al., 2001; Chong
huKO, human Kusabira- receptor 4 (CCR4) is a hallmark of this disease et al., 2004). These observations suggest that
Orange; PI3K, PI(3) kinase; (Yoshie et al., 2002; Ishida et al., 2003; Iqbal CCR4 could have a role in ATLL biology, but it
PTX, pertussis toxin; SNV,
single nucleotide variant.
et al., 2010). Clinical trials in ATLL of a thera- is still unclear whether dysregulation of CCR4
peutic monoclonal antibody directed against function contributes to ATLL pathogenesis.
CCR4 (KW-0761) are ongoing, and promising Human T cell lymphotropic virus type-I
early results have been reported (Yamamoto et al., (HTLV-I) is believed to be the causative agent
2010; Ishida et al., 2012). for ATLL (Matsuoka and Jeang, 2007; Campo
CCR4 is a chemokine receptor that has a et al., 2011). However, only a small proportion
critical role in immune cell trafficking.T-helper of HTLV-I carriers (27%) develop ATLL with
type 2 cells (Th2), regulatory T cells (Treg), a long latency (4050 yr; Arisawa et al., 2000;
interluekin-17producing T-helper cells (Th17), Campo et al., 2011). Thus, acquisition of so-
and skin-homing memory T cells express CCR4 matic mutations in cellular genes is likely to be
on their surface and migrate toward the che- crucial for the development of ATLL. Identi-
mokines CCL17 and CCL22 (Imai et al., 1997, fying such somatic mutations is essential not
1998; Yoshie, 2005). The leukemic cells in 90% only for understanding ATLL pathogenesis but
of ATLL cases express CCR4 on their surface also for defining molecular targets for therapy.

This article is distributed under the terms of an AttributionNoncommercial


*M. Nakagawa and R. Schmitz contributed equally to this paper. Share AlikeNo Mirror Sites license for the first six months after the publication
date (see http://www.rupress.org/terms). After six months it is available under a
**T.A. Waldmann and L.M. Staudt contributed equally to Creative Commons License (AttributionNoncommercialShare Alike 3.0 Unported
this paper. license, as described at http://creativecommons.org/licenses/by-nc-sa/3.0/).

The Rockefeller University Press $30.00


J. Exp. Med. 2014 Vol. 211 No. 13 24972505 2497
www.jem.org/cgi/doi/10.1084/jem.20140987
Somatic mutations in p53, NOTCH1, JAK3, and FAS have that these four lines were established from the same patient
been reported in ATLL (Elliott et al., 2011; Yamagishi and at different clinical time points.
Watanabe, 2012), but our knowledge of genetic aberrations in In cancer genome studies cataloged in the COSMIC data
this malignancy is nonetheless incomplete. base (Catalogue of somatic mutations in cancer) or con-
In the present study, we used whole transcriptome analy- ducted by The Cancer Genome Atlas (TCGA) initiative, we
sis (RNA-seq) to discover activating mutations in CCR4, found only three primary human cancer samples with CCR4
which we found to be a frequent genetic event in this malig- nonsense mutations, and these mutations did not target the
nancy. Functional analysis clarified the gain-of function nature carboxy-terminal cytoplasmic domain. Thus, truncation of
of these mutations, suggesting that dysregulation of CCR4 the CCR4 cytoplasmic domain by somatic mutation appears
function is key to the pathogenesis of ATLL. to be a specific and frequent genetic event to ATLL.

RESULTS AND DISCUSSION Mutant CCR4 enhances chemotaxis toward CCR4 ligands
Frequent mutation of CCR4 in ATLL Chemokine receptors, including CCR4, belong to the seven-
We performed RNA-seq of peripheral blood leukemia sam- transmembrane G proteincoupled receptor (GPCR) family.
ples from two ATLL patients, TW36R and TW51R, which All CCR4 mutations in ATLL encode truncated receptors that
allowed us to identify 85 and 127 genes with potential coding lack most of the carboxy-terminal cytoplasmic domain, which
region mutations in these two samples, respectively. These typically serves a regulatory role in GPCRs (Fig. 1, A and B;
candidates included two genes that were mutated in both Luttrell and Lefkowitz, 2002). The loss of this region of CCR4
samples: CCR4 and MICALL1. High expression of CCR4 is might deregulate its activity, potentially altering the migration
a well-known hallmark of ATLL, whereas MICALL1 has not of ATLL cells in response to CCR4 ligands. The CCR4 muta-
been implicated in this disease. Importantly, both ATLL sam- tions in ATLL are reminiscent of mutations affecting the
ples had the same nonsense CCR4 mutation affecting the chemokine receptor CXCR4 in WHIM syndrome, a human
Y331 codon (Y331*), suggesting that CCR4 mutations might immunodeficiency disease (Diaz, 2005). Most CXCR4 muta-
play a critical role in ATLL pathogenesis. The percentage of tions in WHIM syndrome are nonsense or frameshift mu-
mutant CCR4 sequencing reads was 39% in TW36R and tations that truncate the carboxy-terminal cytoplasmic domain
55% in TW51R. Because both blood samples had a high pro- of the protein, conferring a gain-of-function phenotype
portion of malignant cells (TW36R: 90%, TW51R: 84%), the with respect to chemotaxis toward the CXCR4 ligand SDF1
CCR4 mutations are likely to be heterozygous and poten- (Balabanian et al., 2005; Diaz, 2005; Kawai et al., 2005).
tially might exert a dominant functional effect. We therefore hypothesized that mutant CCR4 isoforms
By Sanger sequencing of genomic DNA, we confirmed might enhance chemotaxis of the affected cells to CCR4 li-
the heterozygous nature of the CCR4 nonsense mutations in gands. To study this, we infected a mouse myeloid cell line,
TW36R and TW51R. We extended this analysis to an addi- 32D, with retroviral vectors expressing WT CCR4 (CCR4-
tional cohort of ATLL primary patient samples (n = 41) WT) or CCR4-Q330* coding regions and tested the migra-
and ATLL cell lines (n = 12). CCR4 mutations were de- tion of the transduced cells toward CCL22, the most potent
tected in 26.4% (14/53) of ATLL samples (Fig. 1, A and B; CCR4 ligand (Fig. 2 A; DAmbrosio et al., 2002). Surface CCR4
and Table S1). In five cases for which paired normal DNA levels were similar between CCR4-WT and CCR4-Q330*
was available, three different CCR4 mutations were detected transduced 32D cells, whereas mock vectortransduced 32D
only in the ATLL cells (Q330*, Q330 frameshift, and Y331*), cells did not have detectable CCR4 expression (Fig. 2 B). Mock
demonstrating that they were acquired somatically during vectorinfected 32D cells did not migrate toward CCL22
malignant transformation or progression (Fig. 1 C). CCR4 (Fig. 2, A and B). CCR4-WTtransduced 32D cells mi-
mutations were identified in both primary ATLL patient samples grated with a typical bell-shaped doseresponse curve (Fig. 2,
(24%, 10/41) and in ATLL cell lines (33.3%, 4/12). Muta- A and B). Compared with CCR4-WT, CCR4-Q330*
tions were heterozygous in all samples except one cell line transduced 32D cells migrated to a significantly greater ex-
(ATL42T+). We also confirmed that mutant CCR4 mRNAs tent (Fig. 2, A and B). These results support the view that the
were heterozygously expressed in six primary ATLL sam- CCR4 mutants in ATLL are gain-of-function with respect to
ples (Table S1). These findings indicated that CCR4 muta- ligand-directed chemotaxis.
tions in ATLL might be either gain-of-function or potentially Next we evaluated the ability of CCR4 isoforms to me-
dominant negative. diate chemotaxis in ATLL cell lines (Fig. 2 C). Most ATLL
All CCR4 mutations were either nonsense or frameshift cell lines express CCR4 on their surface (not depicted), re-
in nature and affected the codons for four nearby amino acids flecting the high frequency (90%) of CCR4 expression in
(F326, C329, Q330, and Y331) that are located in an evolu- primary ATLL cases (Ishida et al., 2003). To evaluate CCR4-
tionarily conserved region in the carboxy terminus of the WT and CCR4-Q330* in ATLL cells, we first knocked
protein (Fig. 1, A and B). The most frequent nonsense muta- down the endogenous expression of CCR4 in ED40515(+)
tion affected codon Y331 (Y331*; 7/53, 13%). Four cell lines cells using an shRNA targeting the 3 untranslated region
(ED40515(+), ED40515(), ED41214(+), and ED41214 C()) (UTR) of the CCR4 mRNA. This resulted in a >90% reduc-
had Q330* nonsense mutations, which is understandable given tion of surface CCR4 expression (Fig. 2 D) and substantially

2498 Gain-of-function CCR4 mutations in ATLL | Nakagawa et al.


Br ief Definitive Repor t

Figure 1. CCR4 mutations in ATLL. (A) Amino acid residues in the carboxy-terminal region of CCR4. (B) Schematic of CCR4 mutant isoforms in ATLL.
(C) DNA sequence of the CCR4-Y331* mutation in the TW14R ATLL biopsy sample by Sanger sequencing (bottom). The WT sequence of normal DNA obtained
from the same patient is shown in the top.

decreased chemotaxis of the cells (Fig. 2 C). Cells were then Mutant CCR4 impairs receptor internalization
transduced with retroviruses expressing CCR4-WT or after CCL22 binding
CCR4-Q330* coding regions, resulting in cell populations The carboxy-terminal region of CCR4 that is truncated by
with equivalent expression of CCR4 on the cell surface mutations in ATLL contains a serine- and threonine-rich
(Fig. 2 D). In chemotaxis assays with CCL17, CCR4-WT motif that is shared by many GPCRs (Fig. 1 A, amino acid
reconstituted cells exhibited greater dose-dependent migra- positions 342351). These serine and threonine residues
tion than mock vectorreconstituted cells, but migration of become rapidly phosphorylated in response to ligand, which
CCR4-Q330*reconstituted cells was significantly greater results in receptor internalization and contributes to desen-
than either of the other populations (Fig. 2 C, left). In re- sitization of the cells to ligand (Luttrell and Lefkowitz,
sponse to CCL22, the most potent CCR4 ligand (DAmbrosio 2002). Carboxy-terminal truncation of CXCR4 in WHIM
et al., 2002), CCR4-Q330*reconstituted ED40515(+) cells syndrome impairs receptor internalization, contributing to
again displayed significantly greater chemotaxis than CCR4- the enhanced migration of these cells in response to ligand
WTreconstituted cells and responded better to lower con- (Balabanian et al., 2005; Kawai et al., 2005).
centrations of the ligand (Fig. 2 C, right). Because CCR4 We therefore studied the change in surface CCR4 levels
mutations in ATLL samples were heterozygous, we also after CCL22 exposure in CCR4-WT and CCR4-Q330*
analyzed whether these mutations could enhance chemotaxis reconstituted ED40515(+) cells. In CCR4-WTreconstituted
in the presence of WT CCR4. To this end, we transduced cells, surface CCR4 levels declined rapidly, with a 58% re-
the shCCR4-ED40515(+) line with one retroviral vector duction at 5 min after CCL22 exposure and a maximum re-
that coexpresses CCR4-WT and the human Kusabira-Orange duction of 75% at 20 min (Fig. 2 G). In comparison, CCR4
(huKO) fluorescent protein and another vector that co internalization in CCR4-Q330*reconstituted cells was
expresses CCR4-Q330 and GFP. This strategy allowed us to significantly impaired, with a 27% of reduction at 5 min and
compare the phenotype of GFP/huKO double-positive cells reaching only a 54% reduction at 20 min (Fig. 2 G). These
that expressed CCR4-WT and Q330* with GFP or huKO results suggest that the ATLL CCR4 mutants impair desensi-
single-positive cells that only ectopically expressed one CCR4 tization by ligand, which likely contributes to the enhanced
isoform (Fig. 2, E and F). Cells expressing both CCR4-WT chemotaxis of cells bearing these mutants.
and CCR4-Q330* showed greater CCL22-directed mi-
gration than cells expressing CCR4-WT alone, which was
similar to the phenotype of cells expressing CCR4-Q330* Mutant CCR4 enhances PI(3) kinase (PI3K)/AKT signaling
alone (Fig. 2 F). These results suggest that ATLL cells may in response to ligand
acquire CCR4 mutations to migrate more effectively toward We explored the influence of the ATLL CCR4 mutants on
their ligands. PI3Kdependent activation of AKT because it has been

JEM Vol. 211, No. 13 2499


Figure 2. CCR4 mutant isoforms enhance chemotaxis and impair receptor internalization. (A) CCL22-mediated chemotaxis of mouse 32D cells
ectopically expressing CCR4-WT or CCR4-Q330*. In these Transwell assays, the lower chamber contained the indicated amount of CCL22. After a 2-h incuba-
tion, the number of cells migrating from the upper to lower chamber was determined and plotted as a percentage of the input cell number. (B) Surface CCR4
expression levels of 32D cells ectopically expressing CCR4-WT or CCR4-Q330* analyzed by FACS. (C) Chemotactic ability of CCR4-WT or CCR4-Q330*
reconstituted ED40515(+) ATLL cells toward CCL17 and CCL22. (D) Surface CCR4 expression levels in CCR4-WT or CCR4-Q330*reconstituted ED40515(+)
ATLL cells analyzed by FACS. (E) Surface CCR4 expression levels analyzed by FACS in ED40515(+) ATLL cells ectopically expressing CCR4-WT and/or CCR4-Q330*.
(B, D, and E) Isotype control IgG staining is indicated in gray. (F) CCL22-induced chemotaxis of ED40515(+) ATLL cells ectopically expressing CCR4-WT and/or
CCR4-Q330*. (G) Time course of surface CCR4 levels after CCL22 exposure in CCR4-WT or CCR4-Q330*reconstituted ED40515(+) ATLL cells. Surface CCR4
levels were analyzed by FACS and normalized to the levels at time 0. Data in all panels are presented as mean SEM of technical duplicates representative of
at least two biological replicates. *, P < 0.05; **, P < 0.01; ***, P < 0.001 for a comparison between CCR4-WT and CCR4-Q330*.

reported that binding of CCL22 to CCR4 activates AKT in endogenous CCR4 reduced AKT activation by CCL22 com-
CEM leukemic T cells and in human Th2 cells (Cronshaw pared with mock-infected cells, confirming that AKT activation
et al., 2004). We first studied two ATLL cell lines: ED40515(+), was dependent on CCR4. Ectopic provision of CCR4-WT
which bears a CCR4-Q330* mutant allele, and KOB, in did not restore AKT activation despite expression of CCR4
which CCR4 is WT. Immunoblot analysis revealed that at a higher level than in mock-transduced ED40515(+)
baseline levels of phospho-AKT (P-AKT), a measure of AKT cells (Fig. 2 D), presumably because the endogenous CCR4
activation, were much lower in ED40515(+) than in KOB locus in ED40515(+) encodes the CCR4-Q330* isoform.
(Fig. 3 A, lane 2 vs. lane 6). However, ED40515(+) showed In contrast, cells reconstituted with CCR4-Q330* restored
stronger induction of AKT phosphorylation at 10 min after robust AKT activation in response to CCL22 (Fig. 3 B). In
CCL22 exposure than KOB (Fig. 3 A, lane 3 vs. lane 7). The KOB cells, reconstitution with CCR4-WT modestly in-
activation of AKT was transient in both cell lines, decreasing creased P-AKT activation in response to CCL22, but cells
by 30 min after stimulation (Fig. 3 A, lanes 4 and 8). These reconstituted with CCR4-Q330* again responded with a
findings indicated that AKT activation is one of the signal- significantly greater rise in P-AKT levels (Fig. 3, C and D).
ing pathways downstream of CCR4 in ATLL cells and sug- The effect of heterozygous mutation of CCR4 on AKT
gested that CCR4 mutations might affect the magnitude activation was analyzed using the dual fluorescence strat-
of AKT activation. To accurately evaluate the relative ability egy described above for experiments depicted in Fig. 2
of CCR4 isoforms to activate AKT, we studied ED40515(+) (E and F). After CCL22 treatment, ED40515(+) cells ex-
cells that were transduced with a CCR4 shRNA to knock pressing both CCR4-WT and CCR4-Q330* or CCR4-
down endogenous CCR4 expression and were reconstituted Q330* alone had elevated p-AKT levels compared with
with CCR4-WT or CCR4-Q330*. Cells were treated with cells expressing only CCR4-WT (Fig. 3 E). Together, these
CCL22, and P-AKT levels were measured by FACS at vari- data demonstrate that CCR4 mutations in ATLL enhance
ous time points after exposure (Fig. 3 B). Knockdown of AKT activation in response to ligand engagement.

2500 Gain-of-function CCR4 mutations in ATLL | Nakagawa et al.


Br ief Definitive Repor t

Figure 3. Enhanced PI3K/AKT activation by mutated


CCR4. (A) Immunoblot analysis for P-AKT levels in
ED40515(+) and KOB ATLL cells after CCL22 exposure
(50 ng/ml). Blots using P-AKT (S473) antibody are shown
with short and long exposure times. Relative scanning den-
sitometry estimates of P-AKT levels are depicted under the
panel showing the short exposure. The pan-PI3K inhibitor
BKM120 was used at 1 M. (B and C) Time course experi-
ment of AKT activation after CCL22 exposure (50 ng/ml)
using CCR4-reconstituted ED40515(+) cells (B) and KOB
cells (C), transduced as indicated with shRNA and cDNA
expression vectors. P-AKT (S473) levels were analyzed by
FACS at the indicated times after CCL22 exposure. MFI,
mean fluorescence intensity. (D) Surface CCR4 expression
levels in CCR4-WT or CCR4-Q330*reconstituted KOB cells.
Isotype control IgG staining is indicated in gray. (E) Time
course experiment of AKT activation after CCL22 expo-
sure (50 ng/ml) in ED40515(+) ATLL cells ectopically ex-
pressing CCR4-WT and/or CCR4-Q330*. (F) P-AKT (S473)
levels of ED40515(+) treated with inhibitors. Cells were
pretreated with 100 ng/ml PTX for 16 h or the indicated
amount of the pan-PI3K inhibitor BKM120 for 1 h and then
incubated with 50 ng/ml CCL22 for 5 min. P-AKT (S473) was
analyzed by FACS. (G) Chemotaxis of ED40515(+) cells
treated with inhibitors. Cells were pretreated with PTX or
BKM120 as in F and then used in a CCL22-mediated chemo-
taxis assay. Data in all panels are presented as mean SEM
of technical duplicates representative of at least two bio-
logical replicates. *, P < 0.05; **, P < 0.01 for a comparison
between CCR4-WT and CCR4-Q330*.

We next investigated whether AKT activation is involved Mutant CCR4 promotes ATLL expansion
in CCR4-mediated chemotaxis in ATLL. The pan-PI3K in- in the presence of ligand
hibitor BKM120 abrogated CCR4-mediated AKT activation Lastly, we tested whether the acquisition of CCR4 mutations
in ED40515(+) cells in a dose-dependent manner, indicating by ATLL cells imparts a selective growth advantage relative
that PI3K signaling contributes to AKT activation (Fig. 3 F). to cells with WT CCR4. After shRNA-mediated knockdown
However, BKM120 treatment only partially inhibited CCR4- of endogenous CCR4 expression, ED40515(+) cells were
mediated chemotaxis by ED40515(+) cells (Fig. 3 G). In con transduced with a retrovirus expressing CCR4-WT together
trast, the Gi inhibitor pertussis toxin (PTX) abrogated AKT with GFP or with a retrovirus expressing only CCR4-Q330*
activation and chemotaxis in these cells (Fig. 3, F and G). (Fig. 4 A, Exp. 1). After puromycin selection of infected cells,
Thus, CCR4-mediated chemotaxis of ATLL cells is not pri- these two cell populations were mixed in equal numbers and
marily caused by PI3K-dependent AKT signaling, but instead cultured with or without CCL22 for 12 d. The ratio of GFP/
depends on Gi, consistent with previous work (Cronshaw CCR4-Q330*expressing cells versus GFP+/CCR4-WT
et al., 2004). expressing cells was monitored every 4 d by FACS (Fig. 4 B,

JEM Vol. 211, No. 13 2501


Figure 4. Growth advantage of CCR4-Q330*
reconstituted ATLL cells. (A) Schematic of the competitive
growth assay. ED40515(+) cells depleted of endogenous
CCR4 expression by RNA interference were transduced with
the indicated CCR4-expressing vectors or mock vectors. After
puromycin selection, two transduced populations, one ex-
pressed GFP and the other did not, were mixed equally and
co-cultured for 12 d in vitro. The ratio of the two popula-
tions was determined by FACS every 4 d. (B) CCR4-Q330*
transduced cells have a competitive growth advantage.
CCL22 was added every 2 d at 50 ng/ml. The ratios of the
two populations were normalized to the value at day 0.
Growth curves represent the mean of eight replicates ob-
tained from four biologically independent experiments SEM.
*, P < 0.05; ***, P < 0.001.

Exp. 1). Without CCL22, the ratio of the two populations 1999; Vissers et al., 2001; Vulcano et al., 2001; Chong et al.,
did not change. In contrast, in the presence of CCL22, CCR4- 2004). Previous studies showed that monocyte and M2-
Q330*reconstituted cells preferentially expanded in a time- phenotype macrophages can promote ATLL cell growth through
dependent manner. To confirm this finding, we performed cellcell interaction and by paracrine mechanisms (Chen
a GFP-swapping experiment in which the GFP+ cells ex- et al., 2010; Komohara et al., 2013). Thus, the chemotaxis
pressed CCR4-Q330* and the GFP cells expressed CCR4- promoted by CCR4 mutations in ATLL might set up favor-
WT (Fig. 4 A, Exp. 2). Again, CCR4-Q330*reconstituted able interactions between ATLL cells and immune bystander
cells had a selective growth advantage in the presence of cells and/or disrupt homeostatic mechanisms that keep the
CCL22 (Fig. 4 B, Exp. 2). As a negative control, we used growth of these cells in check. Recently, WHIM syndrome-
empty vectors to create GFP+ and GFP populations and like CXCR4 somatic mutations have been detected in the
observed no change in the GFP+/GFP ratio in the presence malignant cells of patients with Waldenstrms macroglob-
or absence of CCL22 (Fig. 4, A and B, Exp. 3). ulinemia, and these mutations correlated with bone mar-
In conclusion, the present study demonstrates that muta- row involvement as well as chemotherapy drug resistance
tions in CCR4 are a frequent genetic event in ATLL and pro- (Roccaro et al., 2014; Treon et al., 2014). Given that CCR4
vides a mechanistic rationale for their selection in this cancer. mutations and WHIM syndromelike CXCR4 mutations
Mutant CCR4 isoforms enhanced migration of ATLL cells share similar biological properties in terms of chemotaxis and
toward CCL17 and CCL22 and additionally promoted PI3K/ ATK activation (Cao et al., 2014; Treon et al., 2014), it will
AKT activation in response to ligand engagement, leading us be interesting to determine whether CCR4 mutations affect
to propose two nonmutually exclusive hypotheses regarding the frequency of lymph node or skin involvement in ATLL or
the role of CCR4 mutations in ATLL pathogenesis. First, the clinical course.
enhanced migration of ATLL cells along a CCL17/CCL22 A second hypothesis raised by our findings is that CCR4
gradient might allow them to access a favorable microenvi- mutations may be selected in ATLL for their ability to
ronmental niche that could support their proliferation and/or enhance signaling downstream of this chemokine receptor,
survival. Lymph nodes and skin are plausible ATLL niches including activation of the PI3K/AKT signaling pathway.
because CCL17 and CCL22 are known to be produced by During physiological CCR4 signaling, ligand desensitization
dendritic cells and M2-phenotype macrophages in lymph nodes occurs in part because CCR4 is down-modulated from the cell
and also by Langerhans cells and venules in skin (Campbell et al., surface, a process which is disrupted by the CCR4 mutations

2502 Gain-of-function CCR4 mutations in ATLL | Nakagawa et al.


Br ief Definitive Repor t

in ATLL. Perhaps as a result, ATLL cells bearing CCR4 mu- CCL17/TARC and recombinant human CCL22/MDC were purchased from
tant isoforms displayed prolonged PI3K/AKT activation in R&D Systems. Human recombinant IL2 was obtained from Roche. Anti
human CCR4 antibodies (clone 1G1) conjugated to PE and Alexa Fluor 647
response to ligand. Given the importance of PI3K/AKT sig-
fluors were purchased from BD. The following antibodies were purchased
naling to both cellular metabolism and survival, this enhanced from Cell Signaling Technology: Akt, P-Akt (Ser473; D9E), P-Akt (S473)
PI3K/AKT response might provide a selective advantage for Alexa Fluor 647 (D9E), and Alexa Fluor 647conjugated isotype control
ATLL cells. Indeed, in our long-term competitive growth antibody (DA1E). Antirabbit IgG-HRP antibody was purchased from GE
assay, ATLL cells expressing mutant CCR4 outgrew cells Healthcare. CD8a(Lyt 2) microbeads were purchased from Miltenyi Biotec.
with WT CCR4 in the presence of CCR4 ligand. Finally, it BKM120 was purchased from Selleck Chemicals. PTX was purchased from
Sigma-Aldrich. Big Dye Terminator v1.1 was purchased from Applied Biosys-
is conceivable that both hypotheses detailed above may per-
tems. Human gamma globulin fraction II was purchased from ICN Biomedicals
tain. Specifically, the ability of CCR4 mutants to increase Inc. Paraformaldehyde was purchased from Electron Microscopy Sciences.
chemotaxis toward CCR4 ligands would expose them to higher
ligand concentrations, which might contribute to their growth RNA-seq. RNA was extracted using the AllPrep kit (QIAGEN) and
and/or survival. sequencing libraries were prepared using the TruSeq RNA sample Prep kit v2
Our findings provide a rationale to test whether inhibi- (Illumina) according to the manufacturers instructions. Paired-end 108-bp
tion of CCR4 signaling might have therapeutic potential for read sequencing was performed on a HiSeq 2000 system (Illumina). The map-
patients with ATLL. Either a CCR4 inhibitor or a PI3K ping of paired-end reads and the extraction of putative single nucleotide vari-
inhibitor might be considered (Pease and Horuk, 2014). ants (SNVs) were performed as previously described (Schmitz et al., 2012). In
brief, paired-end reads were mapped to the RNA sequences in the RefSeq
The anti-CCR4 monoclonal antibody KW-0761, which is database (NCBI build 37) using the Burrows-Wheeler Aligner (BWA) software
showing promising results in clinical trials (Yamamoto et al., with default parameters. Reads that failed to map to RefSeq were mapped
2010; Ishida et al., 2012), was designed to promote antibody- to RNA sequences in the Ensembl database. The remaining unmapped reads
dependent cellular cytotoxicity of ATLL cells. This antibody were mapped to the human genome assembly (NCBI build 37). Mutant SNV
only inhibits chemotaxis weakly (Ishida et al., 2006), and its calls were declared if more than two reads were mutated and the ratio of mu-
effect on PI3K/AKT signaling has not been evaluated. To tant reads versus total coverage was >20%. SNVs that corresponded to single
nucleotide polymorphisms in the dbSNP database (build #132), the 1000
improve the therapy of ATLL, our findings would support Genomes database (May 2011 release), and the NHLBI GO Exome Sequenc-
the development of a therapeutic anti-CCR4 antibody ing Project (ESP) database (ESP5400 December 2011 release) were excluded.
that both inhibits CCR4 signaling and mediates antibody- RNA-seq data were submitted to the NCBI Short Read Archive (SRA;
dependent cellular cytotoxicity. accession no. SRP042199).

MATERIALS AND METHODS Sanger DNA resequencing. Genomic DNA of CCR4 exon 2 region
Experimental design. All experiments presented have been repeated at was amplified by PCR using the primers CCR4-E2F, 5-CTTCCCCT
least two times, and consistent results were obtained. Data are depicted as CATTAGCTGCTTCTGGTTG-3; and CCR4-E2R, 5-CCTGAC
means SEM. Statistical comparisons were made using the Students t test. ACTGGCTCAGGAATCTCTTAC-3. The purified PCR products were
P < 0.05 was considered statistically significant. sequenced using a Big Dye Terminator v1.1 cycle sequencing kit and
analyzed on an ABI 3730 Genetic Analyzer (Applied Biosystems). The
Patient samples and cell lines. Written informed consent was obtained in following primers were used for sequencing: CCR4-E2.1F, 5-CCTTC
accordance with the Declaration of Helsinki and was approved by the Investi- CTGGCTTTCTGTTCAGCACTTG-3; and CCR4-E2.1R, 5-TGA
gational Review Board of the National Cancer Institute (NCI). Some samples TTTCCAGGGAGCTGAGAACCTTCC-3.
were obtained before cytotoxic chemotherapy, whereas others were taken after
treatment (Table S1). PBMCs were isolated from ATLL patients by Ficoll- Sanger RNA resequencing. RNA was DNase-digested and reverse tran-
Hypaque. In cases where the ATLL cells were <70% of the mononuclear cells, scribed using the Omniscript RT kit (QIAGEN). The CCR4 coding region
the ATLL cells were purified by an initial negative selection magnetic column was amplified by PCR using primers CCR4-E2F and CCR4-E2R. The
method (Miltenyi Biotec) to enrich for CD4+ cells followed by a positive purified PCR products were cloned using the TOPO XL PCR Cloning kit
selection on a CD25 column. The resultant population consisted of 7095% (Invitrogen). Sequencing was performed as described in the Sanger DNA
CD4+CD25+ ATLL cells. The ATLL cell lines were provided by the follow- resequencing section.
ing researchers: M. Maeda (Kyoto University, Kyoto, Japan; ED40515(+),
ED40515(), ED41214(+), ED41214C(), ATL43T(+), ATL43Tb(), CCR4 mutation search of public databases. cBioPortal and COSMIC
ATL55T(+), and ATL42T(+)),Y.Yamada (Nagasaki University, Nagasaki, Japan; were used.
ST1, KOB, KK1, and LMY1), T. Hata (Nagasaki University; ST1), T. Naoe
(Nagoya University, Nagoya, Japan; ATN1), and N. Arima (Kagoshima Uni- Retroviral vectors and retroviral transduction. CCR4-WT and CCR4-
versity, Kagoshima, Japan; Su9T01). 32D (a variant of the mouse 32D my- Q330* cDNA was PCR amplified from ED40515(+) cDNA using the prim-
eloid cell line engineered to express the human IL-2 receptor subunit) was ers HindIII-CCR4-S, 5-GGAAGCTTTTGAAAGGCACCGGGTC-3;
also used. IL2-dependent cell lines (ED40515(+), ED41214(+), ATL43T(+), and CCR4-TAG-BamHI-AS, 5-CGGGATCCCTACAGAGCATCATG-
ATL55(+), ATL42T(+), KOB, KK, LMY1, and 32D) were cultured with GAGAT-3. The amplified fragments were cloned into HindIIBamHI sites
RPMI/10% FCS/penicillin-streptomycin with 100 IU/ml human recom- of doxycycline-inducible pRetroCMV/TO/puro and pRetroCMV/TO/PG
binant IL2. Other lines were cultured with RPMI/10% FCS/penicillin- vectors. MSCV-CCR4-WT-ires-huKO or MSCV-CCR4-Q330*-ires-GFP
streptomycin. ED40515(+) and KOB were engineered to express ecotropic was made by inserting HindIII (blunted)XhoI fragments from pRetroCMV/
retroviral receptors and TET repressor for transduction of retroviral shRNA TO/CCR4-WT-puro or pRetroCMV/TO/CCR4-Q330*-puro vectors
vectors and expression vectors as previously described (Schmitz et al., 2012). into EcoRI(blunted)XhoI sites of MSCV-ires-huKO or MSCV-ires-GFP
(provided by S. Tsuzuki, Aichi Cancer Center Research Institute, Nagoya,
Reagents and antibodies. Doxycycline, puromycin, ethidium bromide, and Japan). shRNA targeting the 3 UTR of CCR4 was cloned into doxycycline-
Lipofectamine 2000 were purchased from Invitrogen. Recombinant human inducible pRSMX-puro vector with these two annealed oligos (shRNA target

JEM Vol. 211, No. 13 2503


sequences are underlined): shCCR4-4A11_F, 5-GATCCCGAATGAAGTT- in 53 cases of ATLL samples. Online supplemental material is available at
GTAGGTAATTTCAAGAGAATTACCTACAACTTCATTCTTTTT-3; http://www.jem.org/cgi/content/full/jem.20140987/DC1.
and shCCR4-4A11_R, 5-AGCTAAAAAGAATGAAGTTGTAGGTA
ATTCTCTTGAAATTACCTACAACTTCATTCGG-3. pBMN-shCCR4- We thank the following researchers for providing ATLL cell lines: Michiyuki Maeda,
ires-Lyt2 was made by insertion of XbaIBsmI fragment from pRSMX- Yasuaki Yamada, Tomoko Hata, Tomoki Naoe, and Naomichi Arima. We also thank
shCCR4-puro vector into XbaIBsmI sites of pBMN-ires-Lyt2 (provided by Richard N. Bamford, Liyanage P. Perera, Ryan Young, Art Shaffer, and Michele
G. Nolan, Stanford University, Stanford, CA). Retroviruses for ED40515(+) Ceribelli for helpful discussions.
and KOB were produced by cotransfection of 293T cells with helper plasmids This research was supported by the Intramural Research Program of the
expressing gag-pol genes and an ecotropic pseudotyping env gene and a retro- National Institutes of Health, National Cancer Institute, Center for Cancer Research.
Roland Schmitz was supported by the Dr. Mildred Scheel Stiftung fr
viral vector using Lipofectamine 2000 as described previously (Schmitz et al.,
Krebsforschung (Deutsche Krebshilfe).
2012). For 32D, retroviruses were produced by transfection of PlatE with ret-
The authors declare no competing financial interests.
roviral vectors. Target cells were infected with the retroviruses with 8 g/ml
polybrene with spin infection at 1,290 g for 1.5 h. pRetroCMV/TO-puro,
Submitted: 23 May 2014
pRetroCMV/TO-PG, and pRSMX-infected cells was purified by using Accepted: 13 November 2014
2 g/ml puromycin. pBMN-ires-Lyt2infected cells were purified by MACS
mouse CD8a (Lyt2) microbeads (Miltenyi Biotech). Doxycycline was used at
40 ng/ml for induction of shRNA and cDNA. REFERENCES
Arisawa, K., M. Soda, S. Endo, K. Kurokawa, S. Katamine, I. Shimokawa, T.
Chemotaxis assay. HTS Transwell 96-well permeable supports with an Koba, T. Takahashi, H. Saito, H. Doi, and S. Shirahama. 2000.
8.0-m pore polyester membrane (Corning) were used for chemotaxis assays Evaluation of adult T-cell leukemia/lymphoma incidence and its
of ED40515(+) and KOB. 235 l of culture medium containing the indi- impact on non-Hodgkin lymphoma incidence in southwestern Japan.
cated dose of chemokine was added to the lower chamber, followed by the Int. J. Cancer. 85:319324. http://dx.doi.org/10.1002/(SICI)1097-
addition of the Transwell inserts and the addition of cells in growth medium 0215(20000201)85:3<319::AID-IJC4>3.0.CO;2-B
(75 l) at a concentration of 5 105 cells/ml into the upper chamber. ChemoTx Balabanian, K., B. Lagane, J.L. Pablos, L. Laurent, T. Planchenault,
(5 M pore; Neuro Probe) was used for chemotaxis assays of 32D. 300 l O. Verola, C. Lebbe, D. Kerob, A. Dupuy, O. Hermine, et al. 2005.
of chemokine-containing medium was added to the lower chamber, and WHIM syndromes with different genetic anomalies are accounted for
55 l of cells in growth media (2 106 cells/ml) were added to the upper by impaired CXCR4 desensitization to CXCL12. Blood. 105:2449
chamber. After incubation for 2 h at 37C in 5% CO2, the upper chamber 2457. http://dx.doi.org/10.1182/blood-2004-06-2289
was removed and 50 l of 1 PBS with a 1:100 dilution of SPHERO parti- Campbell, J.J., G. Haraldsen, J. Pan, J. Rottman, S. Qin, P. Ponath, D.P. Andrew,
R. Warnke, N. Ruffing, N. Kassam, et al. 1999. The chemokine receptor
cles (SpheroTECH) and 1:2,500 dilution of 10 mg/ml ethidium bromide
CCR4 in vascular recognition by cutaneous but not intestinal memory
was added into each lower chamber. The numbers of cells and SPHERO
T cells. Nature. 400:776780. http://dx.doi.org/10.1038/23495
particles in lower chamber were quantified with a FACSCalibur (BD), and
Campo, E., S.H. Swerdlow, N.L. Harris, S. Pileri, H. Stein, and E.S. Jaffe.
the numbers of cells that had migrated in each condition were normalized 2011. The 2008 WHO classification of lymphoid neoplasms and be-
based on the SPHERO particle count. yond: evolving concepts and practical applications. Blood. 117:5019
5032. http://dx.doi.org/10.1182/blood-2011-01-293050
Flow cytometry for CCR4 surface staining. Cells were washed with
Cao, Y., Z.R. Hunter, X. Liu, L. Xu, G. Yang, J. Chen, C.J. Patterson, N.
PBS containing 200 g/ml of human gamma globulin (Human Gamma
Tsakmaklis, S. Kanan, S. Rodig, et al. 2014. The WHIM-like CXCR4S338X
Globulin Fraction II; ICN Biomedicals Inc.) twice and incubated for 30 min
somatic mutation activates AKT and ERK, and promotes resistance
on ice with antihuman CCR4-PE or CCR4Alexa Fluor 647 (1G1; BD) to ibrutinib and other agents used in the treatment of Waldenstroms
or control isotype IgG. After washing with FACS buffer (PBS/1% FCS) Macroglobulinemia. Leukemia. In press. http://dx.doi.org/10.1038/
twice, the cells were analyzed on a FACSCalibur (BD). For the CCR4 in- leu.2014.187
ternalization assay, cells were exposed to CCL22 for varying lengths of time Chen, J., M. Petrus, B.R. Bryant, V.P. Nguyen, C.K. Goldman, R. Bamford,
and were then treated for 1 min at 37C to an acid buffer (pH 3.0, consisting J.C. Morris, J.E. Janik, and T.A. Waldmann. 2010. Autocrine/paracrine
of 50 mM glycine and 100 mM NaCl) to remove cell-bound CCL22. The cytokine stimulation of leukemic cell proliferation in smoldering and
cells were washed twice and then stained with antihuman CCR4Alexa chronic adult T-cell leukemia. Blood. 116:59485956. http://dx.doi
Fluor 647 on ice for 30 min, fixed with 2% paraformaldehyde (Electron .org/10.1182/blood-2010-04-277418
Microscopy Sciences) on ice for 10 min, and analyzed on a FACSCalibur. Chong, B.F., J.E. Murphy, T.S. Kupper, and R.C. Fuhlbrigge. 2004. E-selectin,
thymus- and activation-regulated chemokine/CCL17, and intercellular
Immunoblot analysis. Cells were washed and resuspended in 2 SDS adhesion molecule-1 are constitutively coexpressed in dermal microves-
sample buffer at 106 cells/100 l and boiled for 5 min. Samples were sepa- sels: a foundation for a cutaneous immunosurveillance system. J. Immunol.
rated on Novex 412% Tris-Glycine gel (Invitrogen) and transferred to a 172:15751581. http://dx.doi.org/10.4049/jimmunol.172.3.1575
PVDF membrane (Immobilon-P; EMD Millipore). Proteins were detected Cronshaw, D.G., C. Owen, Z. Brown, and S.G. Ward. 2004. Activation of
with the following primary antibodies: p-AKT(S473) (D9E) and AKT (5G3; phosphoinositide 3-kinases by the CCR4 ligand macrophage-derived
Cell Signaling Technology). An antirabbit IgG-HRP antibody (GE Health- chemokine is a dispensable signal for T lymphocyte chemotaxis. J. Immunol.
care) was used as the secondary antibody. 172:77617770. http://dx.doi.org/10.4049/jimmunol.172.12.7761
DAmbrosio, D., C. Albanesi, R. Lang, G. Girolomoni, F. Sinigaglia, and C.
Laudanna. 2002. Quantitative differences in chemokine receptor en-
AKT phospho-flow analysis. After incubation with 50 ng/ml CCL22 for
gagement generate diversity in integrin-dependent lymphocyte adhe-
the indicated time, cells were fixed in 2% paraformaldehyde for 10 min at sion. J. Immunol. 169:23032312. http://dx.doi.org/10.4049/jimmunol
room temperature and then permeabilized with cold methanol at 20C .169.5.2303
overnight. Cells were washed twice in FACS buffer and stained with p-Akt Diaz, G.A. 2005. CXCR4 mutations in WHIM syndrome: a misguided
(S473)Alexa Fluor 647 (D9E) or isotype control (DA1E; Cell Signaling immune system? Immunol. Rev. 203:235243. http://dx.doi.org/10.1111/
Technology) for 20 min at room temperature. Cells were washed and resus- j.0105-2896.2005.00226.x
pended in FACS buffer for analysis on a FACSCalibur. Elliott, N.E., S.M. Cleveland, V. Grann, J. Janik, T.A. Waldmann, and U.P. Dav.
2011. FERM domain mutations induce gain of function in JAK3 in
Online supplemental material. Table S1, included as a separate Excel adult T-cell leukemia/lymphoma. Blood. 118:39113921. http://dx.doi
file, shows Sanger sequencing analysis of exon 2 of CCR4 (NM_005508.4) .org/10.1182/blood-2010-12-319467

2504 Gain-of-function CCR4 mutations in ATLL | Nakagawa et al.


Br ief Definitive Repor t

Imai, T., M. Baba, M. Nishimura, M. Kakizaki, S. Takagi, and O.Yoshie. 1997. Pease, J.E., and R. Horuk. 2014. Recent progress in the development of
The T cell-directed CC chemokine TARC is a highly specific biological antagonists to the chemokine receptors CCR3 and CCR4. Expert
ligand for CC chemokine receptor 4. J. Biol. Chem. 272:1503615042. Opin Drug Discov. 9:467483. http://dx.doi.org/10.1517/17460441
http://dx.doi.org/10.1074/jbc.272.23.15036 .2014.897324
Imai, T., D. Chantry, C.J. Raport, C.L. Wood, M. Nishimura, R. Godiska, Roccaro, A.M., A. Sacco, C. Jimenez, P. Maiso, M. Moschetta, Y. Mishima,
O. Yoshie, and P.W. Gray. 1998. Macrophage-derived chemokine is a Y. Aljawai, I. Sahin, M. Kuhne, P. Cardarelli, et al. 2014. C1013G/
functional ligand for the CC chemokine receptor 4. J. Biol. Chem. CXCR4 acts as a driver mutation of tumor progression and modulator
273:17641768. http://dx.doi.org/10.1074/jbc.273.3.1764 of drug resistance in lymphoplasmacytic lymphoma. Blood. 123:4120
Iqbal, J., D.D. Weisenburger, T.C. Greiner, J.M. Vose, T. McKeithan, 4131. http://dx.doi.org/10.1182/blood-2014-03-564583
C. Kucuk, H. Geng, K. Deffenbacher, L. Smith, K. Dybkaer, et al. Schmitz, R., R.M. Young, M. Ceribelli, S. Jhavar, W. Xiao, M. Zhang, G.
International Peripheral T-Cell Lymphoma Project. 2010. Molecular Wright, A.L. Shaffer, D.J. Hodson, E. Buras, et al. 2012. Burkitt lym-
signatures to improve diagnosis in peripheral T-cell lymphoma and prog- phoma pathogenesis and therapeutic targets from structural and func-
nostication in angioimmunoblastic T-cell lymphoma. Blood. 115:1026 tional genomics. Nature. 490:116120. http://dx.doi.org/10.1038/
1036. http://dx.doi.org/10.1182/blood-2009-06-227579 nature11378
Ishida, T., A. Utsunomiya, S. Iida, H. Inagaki, Y. Takatsuka, S. Kusumoto, Treon, S.P., Y. Cao, L. Xu, G. Yang, X. Liu, and Z.R. Hunter. 2014. Somatic
G. Takeuchi, S. Shimizu, M. Ito, H. Komatsu, et al. 2003. Clinical sig- mutations in MYD88 and CXCR4 are determinants of clinical presen-
nificance of CCR4 expression in adult T-cell leukemia/lymphoma: its tation and overall survival in Waldenstrom macroglobulinemia. Blood.
close association with skin involvement and unfavorable outcome. Clin. 123:27912796. http://dx.doi.org/10.1182/blood-2014-01-550905
Cancer Res. 9:36253634.
Vissers, J.L., F.C. Hartgers, E. Lindhout, M.B. Teunissen, C.G. Figdor,
Ishida, T., T. Ishii, A. Inagaki, H.Yano, H. Komatsu, S. Iida, H. Inagaki, and R.
and G.J. Adema. 2001. Quantitative analysis of chemokine expres-
Ueda. 2006. Specific recruitment of CC chemokine receptor 4-positive
sion by dendritic cell subsets in vitro and in vivo. J. Leukoc. Biol. 69:
regulatory T cells in Hodgkin lymphoma fosters immune privilege.
785793.
Cancer Res. 66:57165722. http://dx.doi.org/10.1158/0008-5472.
Vulcano, M., C. Albanesi, A. Stoppacciaro, R. Bagnati, G. DAmico, S. Struyf,
CAN-06-0261
P.Transidico, R. Bonecchi, A. Del Prete, P. Allavena, et al. 2001. Dendritic
Ishida, T., T. Joh, N. Uike, K. Yamamoto, A. Utsunomiya, S. Yoshida,
Y. Saburi, T. Miyamoto, S. Takemoto, H. Suzushima, et al. 2012. cells as a major source of macrophage-derived chemokine/CCL22 in vitro
Defucosylated anti-CCR4 monoclonal antibody (KW-0761) for relapsed and in vivo. Eur. J. Immunol. 31:812822. http://dx.doi.org/10.1002/1521-
adult T-cell leukemia-lymphoma: a multicenter phase II study. J. Clin. 4141(200103)31:3<812::AID-IMMU812>3.0.CO;2-L
Oncol. 30:837842. http://dx.doi.org/10.1200/JCO.2011.37.3472 Yamagishi, M., and T. Watanabe. 2012. Molecular hallmarks of adult
Kawai, T., U. Choi, N.L. Whiting-Theobald, G.F. Linton, S. Brenner, J.M. T cell leukemia. Front Microbiol. 3:334. http://dx.doi.org/10.3389/fmicb
Sechler, P.M. Murphy, and H.L. Malech. 2005. Enhanced function .2012.00334
with decreased internalization of carboxy-terminus truncated CXCR4 Yamamoto, K., A. Utsunomiya, K. Tobinai, K. Tsukasaki, N. Uike, K. Uozumi,
responsible for WHIM syndrome. Exp. Hematol. 33:460468. http:// K.Yamaguchi,Y.Yamada, S. Hanada, K. Tamura, et al. 2010. Phase I study
dx.doi.org/10.1016/j.exphem.2005.01.001 of KW-0761, a defucosylated humanized anti-CCR4 antibody, in re-
Komohara, Y., D. Niino, Y. Saito, K. Ohnishi, H. Horlad, K. Ohshima, lapsed patients with adult T-cell leukemia-lymphoma and peripheral
and M. Takeya. 2013. Clinical significance of CD163+ tumor-associated T-cell lymphoma. J. Clin. Oncol. 28:15911598. http://dx.doi.org/
macrophages in patients with adult T-cell leukemia/lymphoma. Cancer 10.1200/JCO.2009.25.3575
Sci. 104:945951. http://dx.doi.org/10.1111/cas.12167 Yoshie, O. 2005. Expression of CCR4 in adult T-cell leukemia. Leuk. Lymphoma.
Luttrell, L.M., and R.J. Lefkowitz. 2002. The role of beta-arrestins in the 46:185190. http://dx.doi.org/10.1080/10428190400007607
termination and transduction of G-protein-coupled receptor signals. Yoshie, O., R. Fujisawa, T. Nakayama, H. Harasawa, H. Tago, D. Izawa, K.
J. Cell Sci. 115:455465. Hieshima,Y. Tatsumi, K. Matsushima, H. Hasegawa, et al. 2002. Frequent
Matsuoka, M., and K.T. Jeang. 2007. Human T-cell leukaemia virus type 1 expression of CCR4 in adult T-cell leukemia and human T-cell leukemia
(HTLV-1) infectivity and cellular transformation. Nat. Rev. Cancer. virus type 1-transformed T cells. Blood. 99:15051511. http://dx.doi.org/
7:270280. http://dx.doi.org/10.1038/nrc2111 10.1182/blood.V99.5.1505

JEM Vol. 211, No. 13 2505


Brief Definitive Report

Enhancement of an anti-tumor immune


response by transient blockade
of central T cell tolerance
Imran S. Khan,1 Maria L. Mouchess,1 Meng-Lei Zhu,3,4 Bridget Conley,3,4
Kayla J. Fasano,1 Yafei Hou,2 Lawrence Fong,2 Maureen A. Su,3,4
and Mark S. Anderson1
1DiabetesCenter and 2Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco,
San Francisco, CA 94143
3Department of Pediatrics and 4Department of Microbiology/Immunology, School of Medicine, and Lineberger Comprehensive

Cancer Center, University of North Carolina, Chapel Hill, NC 27599

Thymic central tolerance is a critical process that prevents autoimmunity but also presents
a challenge to the generation of anti-tumor immune responses. Medullary thymic epithelial
cells (mTECs) eliminate self-reactive T cells by displaying a diverse repertoire of tissue-
specific antigens (TSAs) that are also shared by tumors. Therefore, while protecting against
autoimmunity, mTECs simultaneously limit the generation of tumor-specific effector T cells
by expressing tumor self-antigens. This ectopic expression of TSAs largely depends on
autoimmune regulator (Aire), which is expressed in mature mTECs. Thus, therapies to
deplete Aire-expressing mTECs represent an attractive strategy to increase the pool of
tumor-specific effector T cells. Recent work has implicated the TNF family members RANK
and RANK-Ligand (RANKL) in the development of Aire-expressing mTECs. We show that in
vivo RANKL blockade selectively and transiently depletes Aire and TSA expression in the
thymus to create a window of defective negative selection. Furthermore, we demonstrate
that RANKL blockade can rescue melanoma-specific T cells from thymic deletion and that
persistence of these tumor-specific effector T cells promoted increased host survival in
response to tumor challenge. These results indicate that modulating central tolerance
through RANKL can alter thymic output and potentially provide therapeutic benefit by
enhancing anti-tumor immunity.

CORRESPONDENCE Medullary thymic epithelial cells (mTECs) con- autoimmune syndrome in patients or mice with
Mark S. Anderson: tribute to self-tolerance through the ectopic ex- defective AIRE expression (Consortium, 1997;
manderson@diabetes.ucsf.edu
pression of tissue-specific antigens (TSAs) in the Nagamine et al., 1997; Anderson et al., 2002).
Abbreviations used: Aire, thymus (Derbinski et al., 2001; Anderson et al., Although central tolerance provides pro-
autoimmune regulator; cTEC, 2002; Metzger and Anderson, 2011). This TSA tection against autoimmunity, this process also
cortical thymic epithelial cell; expression in mTECs is largely dependent on represents a challenge for anti-tumor immunity
IRBP, interphotoreceptor
retinoid-binding protein; mTEC, autoimmune regulator (Aire), which is expressed (Kyewski and Klein, 2006; Malchow et al., 2013).
medullary thymic epithelial cell; in mature mTECs (Gbler et al., 2007; Gray et al., Because many of the TSAs expressed in the thy-
OPG, Osteoprotegerin; TSA, 2007; Metzger and Anderson, 2011). Through mus are also expressed in tumors, high-affinity
tissue-specific antigen.
the recognition of TSAs, developing autoreactive effector T cells capable of recognizing tumor
T cells are either negatively selected from the pool self-antigens may normally be deleted in the thy-
of developing thymocytes or recruited into the mus (Bos et al., 2005; Cloosen et al., 2007; Trger
regulatory T (T reg) cell lineage (Liston et al., 2003; et al., 2012; Zhu et al., 2013). Transiently sup-
Anderson et al., 2005; DeVoss et al., 2006; Shum pressing central tolerance by depleting mTECs
et al., 2009; Taniguchi et al., 2012; Malchow et al., or modulating Aire expression may provide a
2013). The overall importance of this process is
2014 Khan et al. This article is distributed under the terms of an Attribution
underscored by the development of a multi-organ NoncommercialShare AlikeNo Mirror Sites license for the first six months
after the publication date (see http://www.rupress.org/terms). After six months
it is available under a Creative Commons License (AttributionNoncommercial
I.S. Khan and M.L. Mouchess contributed equally to Share Alike 3.0 Unported license, as described at http://creativecommons.org/
this paper. licenses/by-nc-sa/3.0/).

The Rockefeller University Press $30.00


J. Exp. Med. 2014 Vol. 211 No. 5 761-768 761
www.jem.org/cgi/doi/10.1084/jem.20131889
Figure 1. Selective depletion of mTECs with anti-
RANKL blockade. (A) Wild-type mice were treated for 2 wk
with either isotype (gray) or anti-RANKL (RANKL, blue)
antibody, and absolute numbers of mTECs and cTECs were
enumerated by flow cytometry. Bar graphs of total cell num-
bers are depicted by mean SEM. (B) Representative flow
cytometry plots of mTECs in A showing relative composition
of mTEC subsets. Values represent mean SEM. Bar graphs
(right) of total cell numbers of each mTEC subset depicted by
mean SEM. (C) Top panels show immunostaining for keratin-8
(green) and keratin-5 (red) on thymic sections, and bottom
panels show immunostaining for keratin-5 (red) and
Aire (green). Bars: (top) 500 m; (bottom) 50 m. (D) Gene
expression analysis on mTECs sorted from wild-type mice
treated with either isotype (gray) or anti-RANKL (blue) anti-
body. Results standardized to Cyclophilin A and normalized
to isotype-treated mice with bars depicting mean SD.
(E) Representative flow cytometry plots of thymocytes from
antibody-treated wild-type mice. Values depict mean SD.
Total thymocyte numbers (right) are indicated with each
circle depicting an individual animal and bars showing the
mean. (F) Flow cytometric analysis of Foxp3 staining in CD4
SP thymocytes from E. Plots (right) show percentage and
number of thymic CD4+ Foxp3+ cells, with bars showing the
mean. Data shown in Fig. 1 is representative of two to four
independent experiments containing three or more indi-
vidual mice within each group. *, P 0.05; ***, P 0.001,
Students t test.

therapeutic window for the generation of T cells capable of has also demonstrated that mTECs in particular have a rela-
recognizing tumor self-antigens. Many current cancer immune tively fast turnover in adult mice with an estimated half-life of
therapies rely on activating relatively weak tumor-specific T cell 2 wk (Gbler et al., 2007; Gray et al., 2007). Given these find-
responses through modulating peripheral tolerance (Swann and ings, we speculated that in vivo blockade of RANKL in adult
Smyth, 2007; Chen and Mellman, 2013). In contrast, manipu hosts could both selectively and transiently inhibit the develop-
lation of central tolerance has the potential to increase the ment and turnover of mTECs with potential to alter central
pool and affinity of effector T cells that can recognize and con- T cell tolerance. To this end, we performed in vivo RANKL
tribute to effective anti-tumor responses. Furthermore, such blockade in adult mice and investigated its effects on both TECs
high-affinity, self-reactive T cells may be more resistant to pe- and developing thymocytes.We show that anti-RANKL treat-
ripheral tolerance mechanisms that typically restrain an anti- ment not only depleted mTECs but could also be used thera-
tumor response (Swann and Smyth, 2007).Thus, the development peutically to break central tolerance and, as a result, increase
of methods that selectively and transiently deplete Aire-expressing the generation of tumor-specific T cells.
mTECs may be an attractive method to enhance tumor-specific
immune responses.
Previous work has identified agents that can inhibit the RESULTS AND DISCUSSION
growth and development of TECs such as corticosteroids, cy- Depletion of mTECs with RANKL blockade
closporine, and some inflammatory cytokines (Anz et al., 2009; The RANKRANKL signaling pathway is important for
Fletcher et al., 2009). Despite their clear inhibitory effects on mTEC development, but it remains unclear what impact per-
TECs, however, these agents do not appear to have selectivity turbation of this pathway might have on the adult thymus.
for blocking mTEC development. Interestingly, recent studies Previous work has linked the RANKRANKL pathway to
have demonstrated a role for TNF family member pairs the development of Aire-expressing mTECs (Rossi et al.,
RANKRANKL and CD40-CD40L in the embryological 2007; Akiyama et al., 2008; Hikosaka et al., 2008; Roberts
development of Aire+ mTECs (Rossi et al., 2007; Akiyama et al., et al., 2012), and we hypothesized that treating mice with
2008; Hikosaka et al., 2008; Roberts et al., 2012). Recent work blocking anti-RANKL antibody would decrease Aire+ mTECs.

762 RANKL blockade enhances anti-tumor immunity | Khan et al.


Br ief Definitive Repor t

We treated wild-type mice with either anti-RANKL or isotype


control antibody for 2 wk and harvested their thymi for analy
sis. Interestingly, although mice treated with anti-RANKL
showed only a modest loss of cortical thymic epithelial cell
(cTEC) cellularity, they exhibited a severe depletion of >80%
of mTECs (Fig. 1 A). Using MHC II and Aire as markers of
mTEC maturation (Gbler et al., 2007; Gray et al., 2007), we
further analyzed the immature mTEClo (MHC IIlo Aire),
intermediate mTEChi (MHC IIhi Aire), and mature Aire+
mTEC subsets (MHC IIhi Aire+).The relative mTEC compo-
sition in anti-RANKLtreated mice revealed a substantial loss
of Aire+ and mTEChi cells along with enrichment of the re-
maining mTEClo cells (Fig. 1 B). Although absolute numbers
of mTECs were decreased across all mTEC subsets, mTEC
depletion was mostly due to the loss of >90% of all Aire+ and
mTEChi cells (Fig. 1 B). Immunostaining revealed a slight de-
crease in the density of keratin-5+ (K5) cells in the medulla of
anti-RANKLtreated mice, whereas Aire+ cells were nearly
undetectable (Fig. 1 C). Importantly, staining for keratin-8
(K8) and K5 showed that the overall corticomedullary thymic
architecture was preserved despite the loss of mature mTECs
Figure 2. Increased Aire+ mTECs in OPG/ mice. (A) Quantitative
(Fig. 1 C). Furthermore, consistent with the loss of the Aire+ PCR analysis of Aire and OPG gene expression in MHC IIlo and MHC IIhi
and mTEChi subsets, Aire-dependent TSA gene expression in mTECs sorted from wild-type B6 mice. Results standardized to Cyclophilin
sorted mTECs was decreased in anti-RANKLtreated mice A and normalized to MHC IIlo, with error bars depicting mean SD.
(Fig. 1 D). (B) Absolute numbers of mTECs and cTECs in OPG+/+ (black) and OPG/
Next, we characterized the impact of anti-RANKLmedi- (red) mice were enumerated by flow cytometry. Bar graphs of total cell
ated mTEC depletion on thymocyte selection. In the poly- numbers are depicted by mean SEM. (C) Representative flow cytometry
clonal T cell repertoire of wild-type mice treated with anti- plots of mTECs in B showing relative composition of indicated mTEC sub-
RANKL, we observed a modest increase in frequencies of sets. Values depict mean SEM. Bar graphs (right) depict total cell num-
bers of each mTEC subset and indicate mean SEM. (D) Immunostaining
both CD4 single-positive (SP) and CD8 SP thymocytes, con-
for keratin-8 (green) and keratin-5 (red) on frozen thymic sections from
sistent with a lack of negative selection (Fig. 1 E). Importantly, OPG+/+ and OPG/ mice. Data are representative of at least three inde-
anti-RANKLtreated mice showed only a slight reduction in pendent experiments with at least three mice per group. *, P 0.05;
the frequency of double-positive thymocytes while absolute **, P 0.01; ***, P 0.001, Students t test. Bars, 500 m.
numbers were maintained, confirming normal positive selec-
tion. In addition, total thymocyte numbers were also maintained
in mice treated with anti-RANKL (Fig. 1 E). Furthermore, corticomedullary thymic architecture despite these changes
anti-RANKLtreated mice showed a 50% reduction of Foxp3+ in TEC cellularity and mTEC composition (Fig. 2 D).
T reg cells within the CD4 SP subset (Fig. 1 F). Collectively, these data suggest that RANKRANKL
Given the dramatic impact of RANKL blockade on OPG interactions regulate both TEC cellularity and mTEC
mTECs, we sought to determine whether this effect could be maturation, and are particularly important for the induction of
reversed in the context of increased RANK signaling. Osteo- Aire+ mTECs. Furthermore, anti-RANKL treatment selec-
protegerin (OPG; Tnfrsf11b) is a soluble decoy receptor for tively targeted mature mTECs and altered negative selection
RANKL and its role as a negative regulator of RANK signal- without affecting the development of new thymocytes.
ing has been well described in bone physiology (Kearns et al.,
2008). Interestingly, in sorted wild-type mTECs, OPG ex- Regeneration of mTECs after withdrawal of anti-RANKL
pression was up-regulated in MHC IIhi mTECs when com- We next examined the kinetics of mTEC recovery after with-
pared with MHC IIlo mTECs (Fig. 2 A). To test whether drawal from anti-RANKL blockade. After a 2-wk treatment
OPG negatively regulates RANKRANKL signaling in window, we harvested mice at 2-wk intervals and observed
mTECs, we analyzed thymi from OPG/ mice.While cTEC gradual recovery of the mTEChi and Aire+ subsets (Fig. 3,
cellularity was increased threefold in OPG/ mice, mTEC A and B). By 10 wk, we observed the complete recovery of
cellularity was increased by nearly 10-fold when compared all mTEC subsets and a normal level of Aire expression after
with OPG+/+ mice (Fig. 2 B). Although absolute numbers of anti-RANKL treatment (Fig. 3 B). Interestingly, the pattern of
all mTEC subsets were increased in OPG/ mice, we ob- mTEC recovery appeared consistent with published reports
served an enrichment of Aire+ mTECs and a proportional of mTEClo cells giving rise to mTEChi cells before the induction
loss of the mTEClo subset (Fig. 2 C). Immunostaining of of Aire+ mTECs (Fig. 3 B; Gbler et al., 2007; Gray et al., 2007;
K8 and K5 showed that OPG/ thymi maintained their Rossi et al., 2007). Notably, thymi from anti-RANKL and

JEM Vol. 211, No. 5 763


Figure 3. Regeneration of mTECs after anti-RANKL with-
drawal. (A) Experimental layout for analysis of mTEC recovery
after anti-RANKL withdrawal. Wild-type mice were treated for
1 wk with isotype or anti-RANKL (black arrows) and harvested at
indicated time points (red arrows). Graph (bottom) depicts rela-
tive composition of mTEClo mTEChi and Aire+ mTECs at each time
point in anti-RANKLtreated mice. Values represent mean
SEM. (B) Representative flow cytometry plot of mTECs from
each of the indicated time points from A. Values represent the
frequency of Aire+ mTECs. (C) Immunostaining for keratin-8
(green) and keratin-5 (red) on thymic sections from isotype or
anti-RANKLtreated mice harvested at 10 wk. Bars, 500 m.
Data shown is representative of at least two independent experi-
ments with three to five mice per group.

isotype-treated mice at 10 wk were indistinguishable by im- transgenic mice express a membrane form of OVA under the
munostaining (Fig. 3 C). Thus, anti-RANKLmediated mTEC control of the rat insulin promoter which results in its expres-
depletion is a transient phenomenon with return of normal sion in both the pancreatic islets and in mTECs (Anderson et al.,
thymic composition after withdrawal of antibody treatment. 2005). When the OT-II CD4+ TCR transgenic line is crossed
to the RIP-mOVA transgenic line, OVA-specific OT-II T cells
Manipulation of thymic negative selection are deleted in the thymus (Anderson et al., 2005). We treated
with RANKL blockade both OT-II and OT-II x RIP-mOVA mice with either anti-
To further characterize the impact of anti-RANKL treatment RANKL or isotype control antibody and performed thymocyte
on negative selection, we first used the OT-II CD4+ TCR analysis. Consistent with previous reports, OT-II x RIP-mOVA
x RIP-mOVA double transgenic mouse model. RIP-mOVA mice treated with control antibody showed a significant

Figure 4. Anti-RANKLmediated mTEC


ablation leads to altered negative selection.
(A) OT-II and OT-II x RIP-mOVA mice were har-
vested after 2 wk of indicated antibody treat-
ment. Percentages of CD4 SP (top) and OT-II
clonotype-specific (bottom) thymocytes are
shown. Values represent mean SD. (B) Quanti-
fication of OT-II clonotype-specific CD4 SP cells
from A. Graph shows mean SEM. **, P 0.01,
Students t test. (C) Representative histograms
for Foxp3 staining in CD4 SP events from A.
(D) Wild-type mice were treated with isotype or
anti-RANKL antibody for 3 wk, immunized with
P2 peptide, and then harvested 10 d later for
tetramer analysis. Representative flow cytom-
etry plots of CD4+ T cells are shown for each
condition. Values indicate absolute numbers of
CD44+ P2-I-Abspecific T cells. IRBP/ mice
were included as positive controls. Each dot on
the graph represents an individual mouse, bars
show mean, and the dotted line represents the
limit of detection. Data shown is representative
of two to three independent experiments.
*, P 0.05, Mann-Whitney test.

764 RANKL blockade enhances anti-tumor immunity | Khan et al.


Br ief Definitive Repor t

Figure 5. RANKL blockade increases


anti-melanoma immune response.
(A and B) RAG1/ x TRP-1 TCR Tg mice were
treated with isotype or anti-RANKL antibody
for 2 wk and thymus and spleen were har-
vested for analysis. Flow cytometry plots in A
show percentage of CD4+ (top) and CD4-gated
V14+ (bottom) T cells. Bar graphs in B show
quantification of data in A. Values represent
mean SEM. *, P 0.05; **, P 0.01;
***, P 0.001, Students t test. Data shown is
representative of at least three independent
experiments with n = 47 mice per group.
(C) Ear skin from mice treated with isotype (gray)
or anti-RANKL (blue) antibody was analyzed
by qPCR for Tyrosinase and Tyrp1 expression.
Results standardized to 2m and normalized
to untreated B6 wild-type mice (black) with
bars depicting mean SD. (D) Survival curves
of RAG1/ x TRP-1 TCR Tg mice inoculated
with B16 melanoma after treatment with iso-
type (gray) or anti-RANKL (blue) antibody,
n = 78 mice for each group. P 0.05, Log-rank
test. Data shown is representative of four inde-
pendent experiments. (E) Tumor sections from
isotype or anti-RANKLtreated mice in D were
immunostained for CD3 (pink) with DAPI coun-
terstain (purple). Bars, 50 m. Mean densities
of CD3+ cells per tumor area are quantified on
the right. Four sections were evaluated for
each treatment group and 6 imaging fields
were randomly scored from each section.
Graphs depict mean SEM. *, P 0.05, Stu-
dents t test. (F) RAG1/ x TRP-1 TCR Tg mice
were inoculated with B16 melanoma after
treatment with isotype (gray) or anti-RANKL
(blue) antibody. Mice were harvested at 21 d
after tumor inoculation and tumor-infiltrating
lymphocytes (TILs) were analyzed by flow cy-
tometry. Graph depicts percentage of Foxp3+
CD25+ T reg cells among CD4+ T cells. Values
represent mean SEM. ***, P 0.001, Students t test. Data shown is n = 79 mice per group pooled from two independent experiments. (G) Survival curves
of RAG1/ recipients inoculated with B16 melanoma after adoptive transfer of splenocytes from either anti-RANKL (blue) or isotype (gray)treated
RAG1/ x TRP-1 TCR Tg mice, n = 10 mice for each group. P 0.001, Log-rank test. Data shown is representative of two independent experiments.

reduction in the proportions of CD4 SP thymocytes and also thymus of Aire+/+ mice, whereas these cells escape deletion
had decreased frequencies and numbers of OT-II+ T cells (Fig. 4, in Aire/ mice and cause autoimmune uveitis (DeVoss et al.,
A and B; Anderson et al., 2005). In contrast, thymocyte pro- 2006; Taniguchi et al., 2012). Through the use of an IRBP pep-
files of anti-RANKLtreated OT-II x RIP-mOVA mice were tide class II tetramer, P2-I-Ab, such autoreactive CD4+ T cells
indistinguishable from that of OT-II mice, demonstrating that can be detected in Aire/ mice. Given both the severe deple-
RANKL blockade prevented thymic deletion of OT-II T cells tion of Aire+ mTECs and the loss of thymic IRBP expression
yet allowed their positive selection (Fig. 4, A and B). We also in anti-RANKLtreated mice, we hypothesized that P2-I-Ab
observed a loss of thymic T reg cell development in these specific T cells could be detected in treated mice. After treating
mice, which suggested a loss of cognate antigen (OVA) ex- wild-type mice with anti-RANKL antibody, we immunized
pression from mTECs within the thymus (Fig. 4 C). mice with a MHC IIbinding IRBP peptide epitope (P2) to
To expand these observations to the polyclonal T cell rep- expand T cells for detection. 10 d after immunization, lymph
ertoire, we analyzed the development of Aire-dependent auto- nodes and spleen were pooled for the enumeration of CD4+
reactive T cells using a tetramer enrichment protocol. Previously, P2-I-Abspecific T cells by flow cytometry. Consistent with the
we had shown that T cells specific for the self-antigen inter- loss of Aire+ mTECs, tetramer analysis of anti-RANKLtreated
photoreceptor retinoid-binding protein (IRBP) are deleted in mice revealed an expansion of CD4+ P2-I-Abspecific T cells

JEM Vol. 211, No. 5 765


(Fig. 4 D). Overall, these data show a clear defect in negative challenged with B16 melanoma, recipients of splenocytes from
selection associated with the loss of Aire+ mTECs in anti- anti-RANKLtreated donors again showed a statistically sig-
RANKLtreated mice. nificant increase in survival (Fig. 5 G). Collectively, these data
demonstrate that short-term, reversible RANKL blockade of
Treatment with anti-RANKL enhances anti-tumor immunity mTEC development can be used to create a therapeutic win-
Given its ability to selectively block mTECs and central toler- dow that allows tumor self-antigenspecific T cells to escape
ance, we next sought to determine whether anti-RANKL thymic deletion. Furthermore, the generation of these high-
treatment could be used to therapeutically enhance anti-tumor affinity tumor-specific T cells confers a survival benefit in a
immunity. Previous work has shown that many tumor self- melanoma tumor model.
antigens are expressed by mTECs as TSAs, and that high-affinity In conclusion, our findings provide strong evidence that
T cells capable of recognizing these tumor self-antigens are ef- mTECs can be selectively and therapeutically targeted by
ficiently deleted in the thymus (Bos et al., 2005; Trger et al., RANKL blockade in adult mice and that anti-RANKL can
2012; Zhu et al., 2013). To test whether anti-RANKL treat- be used as an approach to enhance anti-tumor responses. To
ment could rescue tumor self-antigenspecific T cells from date, previous efforts on the therapeutic manipulation of
thymic deletion, we used the TRP-1 CD4+ TCR transgenic TECs have shown global effects that involve both cTECs and
mouse model which generates T cells specific for the mela- mTECs and disrupt thymocyte development (Fletcher et al.,
noma antigen TRP-1 (Tyrp1; Muranski et al., 2008). TRP-1 is 2009). The selectivity for mTECs with anti-RANKL thus
expressed in B16 melanoma cells, and TRP-1 T cells are effica- provides evidence that central tolerance can be transiently sup-
cious against established B16 melanoma tumors (Muranski et al., pressed in adult hosts while maintaining T cell generation. Al-
2008). Importantly, mTECs endogenously express TRP-1 in an though autoimmunity is a dangerous potential consequence
Aire-dependent manner, such that TRP-1specific T cells are of this approach, the treatment may also be an attractive new
deleted in the thymi of Aire-sufficient, TRP-1sufficient hosts method to help break tolerance for cancer immunotherapy.
(Muranski et al., 2008; Zhu et al., 2013). Given the depletion of Interestingly, although we could detect an expansion of
Aire+ mTECs and loss of thymic TRP-1 expression in anti- retina-specific T cells in anti-RANKLtreated mice, we did
RANKLtreated mice (Fig. 1 D), we hypothesized that anti- not observe development of spontaneous uveitis (unpublished
RANKL treatment could rescue TRP-1 T cells from thymic data), suggesting that this brief window of central tolerance
deletion. We treated RAG1/ x TRP-1 TCR transgenic mice suppression was countered by peripheral tolerance mecha-
with either anti-RANKL or control antibody and harvested nisms such as T reg cells which existed before treatment. Im-
their thymi and spleens for analysis. Consistent with previous portantly, we find that the medullary epithelial compartment
reports, isotype-treated mice showed efficient deletion of CD4 of the thymus recovers after withdrawal of anti-RANKL an-
SP T cells in the thymus (Fig. 5 A; Muranski et al., 2008; Zhu tibody and, thus, only a transient window of central tolerance
et al., 2013). In contrast, anti-RANKL treatment prevented suppression occurs. This window may be an attractive feature
deletion of CD4 SP cells and also resulted in a much higher of this approach, especially if coupled with methods that pref-
percentage of V14+ T cells (Fig. 5, A and B). Furthermore, erentially expand tumor-specific T cells over pathogenic auto-
TRP-1 T cells were detectable in the spleens of anti-RANKL reactive T cells. Currently, there has been intense interest and
treated mice and also expressed higher levels of the V14 TCR progress in manipulating peripheral tolerance for immuno
(Fig. 5, A and B). In addition, qPCR analysis revealed a loss of therapy (Chen and Mellman, 2013). Given our results, it will
Tyrosinase and Tyrp1 expression in the ear skin of anti-RANKL also be of interest to determine if a combination of methods
treated mice which appeared consistent with the destruction of that target both central and peripheral tolerance could fur-
melanocytes by TRP-1 T cells (Fig. 5 C). ther enhance anti-tumor immune responses. Finally, it is impor-
Next, we challenged anti-RANKLtreated RAG1/ x tant to note that our results also have implications for patients
TRP-1 mice with B16 melanoma to determine whether a lim- in the clinical setting receiving denosumab, a humanized mono-
ited break in central tolerance could improve overall survival. clonal antibody that blocks RANKL which is widely used in
We observed a statistically significant increase in the survival of the treatment of osteoporosis in adults (Cummings et al., 2009).
anti-RANKLtreated mice compared with the isotype-treated Further study will be needed in such patients regarding their
cohort (Fig. 5 D).We also found evidence of an enhanced T cell susceptibility to autoimmunity and for other potential defects
response in anti-RANKLtreated mice by measuring CD3+ in central tolerance.
T cell infiltrates within the tumors of these mice (Fig. 5 E). Of
note, given the overall increase in T reg cells in the spleen and MATERIALS AND METHODS
tumors of anti-RANKLtreated mice, it appears unlikely that Mice. C57BL/6, B6.RAG1/ Tyrp1B-w TRP-1 TCR transgenic, and B6.
the survival benefit observed in TRP-1 mice is due to differ- OPG/ mice were purchased from The Jackson Laboratory. Mice were
ences in T reg cell numbers (Fig. 5, B and F). Furthermore, to treated intraperitoneally with 100 g anti-RANKL (IK22/5) or isotype con-
trol (2A3; Bio X Cell) antibody three times per week as stated in the text.
exclude potential effects of anti-RANKL antibody on the
B6.OT-II, B6.RIP-mOVA, and B6.IRBP/ mice were described previously
tumor microenvironment, we performed adoptive transfer (Anderson et al., 2005; DeVoss et al., 2006). Mice were treated at 35 wk of
studies in which splenocytes from antibody-treated RAG1/ age and harvested at time points indicated in the text. All mice were housed
x TRP-1 mice were transferred into RAG1/ mice. When and bred in specific pathogen-free conditions in animal facilities at UCSF or

766 RANKL blockade enhances anti-tumor immunity | Khan et al.


Br ief Definitive Repor t

UNC, Chapel Hill. Animal experiments were approved by the Institutional Submitted: 6 September 2013
Animal Care and Use Committee (IACUC) at UCSF or UNC, Chapel Hill. Accepted: 3 April 2014

Quantitative PCR. RNA was extracted from sorted mTECs using the REFERENCES
RNeasy Micro Plus kit (QIAGEN) and cDNA was synthesized with the In- Akiyama, T.,Y. Shimo, H.Yanai, J. Qin, D. Ohshima,Y. Maruyama, Y. Asaumi,
vitrogen Superscript III kit. TaqMan gene expression assays (Applied Biosys- J. Kitazawa, H. Takayanagi, J.M. Penninger, et al. 2008. The tumor ne-
tems) were used for all targets. crosis factor family receptors RANK and CD40 cooperatively establish
the thymic medullary microenvironment and self-tolerance. Immunity.
Histology and immunofluorescence. Thymi were harvested and embed- 29:423437. http://dx.doi.org/10.1016/j.immuni.2008.06.015
Anderson, M.S., E.S.Venanzi, L. Klein, Z. Chen, S.P. Berzins, S.J. Turley, H. von
ded in O.C.T. media (Tissue-Tek). 8-m frozen thymic sections were fixed in
Boehmer, R. Bronson, A. Dierich, C. Benoist, and D. Mathis. 2002. Projec
100% acetone, blocked, and stained for keratin-5, keratin-8 (Abcam), or Aire
tion of an immunological self shadow within the thymus by the aire pro-
(eBioscience). Secondary antibodies were purchased from Invitrogen. Thymic
tein. Science. 298:13951401. http://dx.doi.org/10.1126/science.1075958
sections were visualized using a widefield microscope (Apotome; Carl Zeiss). Anderson, M.S., E.S.Venanzi, Z. Chen, S.P. Berzins, C. Benoist, and D. Mathis.
For tumor immunostaining, tumors were harvested and fixed in 10% formalin 2005.The cellular mechanism of Aire control of T cell tolerance. Immunity.
as previously described (Zhu et al., 2013). Fixed tumors were embedded in 23:227239. http://dx.doi.org/10.1016/j.immuni.2005.07.005
paraffin and sectioned for staining with anti-CD3 antibody and counterstained Anz, D., R.Thaler, N. Stephan, Z.Waibler, M.J.Trauscheid, C. Scholz, U. Kalinke,
with DAPI. Tumor sections were visualized using a fluorescent microscope W. Barchet, S. Endres, and C. Bourquin. 2009. Activation of melanoma
(BX60; Olympus) and analyzed using ImageJ software (National Institutes differentiation-associated gene 5 causes rapid involution of the thymus. J.
of Health). Immunol. 182:60446050. http://dx.doi.org/10.4049/jimmunol.0803809
Bos, R., S. van Duikeren,T. van Hall, P. Kaaijk, R.Taubert, B. Kyewski, L. Klein,
Flow cytometry. TECs were isolated as previously described (Gardner C.J. Melief, and R. Offringa. 2005. Expression of a natural tumor anti-
et al., 2008). In brief, thymi were minced and digested with DNase I and gen by thymic epithelial cells impairs the tumor-protective CD4+ T-cell
Liberase TM (Roche) before gradient centrifugation with Percoll PLUS (GE repertoire. Cancer Res. 65:64436449. http://dx.doi.org/10.1158/0008-
Healthcare). Enriched stromal cells were stained with the indicated surface 5472.CAN-05-0666
marker antibodies (BioLegend). mTECs were defined as CD45, EpCAM+, Chen, D.S., and I. Mellman. 2013. Oncology meets immunology: the cancer-
MHC II+, Ly51 events and cTECs were defined as CD45, EpCAM+, immunity cycle. Immunity. 39:110. http://dx.doi.org/10.1016/j.immuni
MHC II+, Ly51+ events. For lymphocyte staining, all surface marker anti- .2013.07.012
Cloosen, S., J. Arnold, M. Thio, G.M. Bos, B. Kyewski, and W.T. Germeraad.
bodies were obtained from BioLegend. For intracellular staining, cells were
2007. Expression of tumor-associated differentiation antigens, MUC1
stained using the Foxp3 Staining Buffer Set and stained with anti-Foxp3
glycoforms and CEA, in human thymic epithelial cells: implications for
or anti-Aire (eBioscience). All data were collected using a flow cytometer
self-tolerance and tumor therapy. Cancer Res. 67:39193926. http://
(LSR II; BD) and analyzed with either FlowJo software (Tree Star) or FACS dx.doi.org/10.1158/0008-5472.CAN-06-2112
Diva (BD). Cell sorting was performed using a FACS Aria III cell sorter (BD). Consortium, F.G.A.; Finnish-German APECED Consortium. 1997. An auto
immune disease, APECED, caused by mutations in a novel gene featuring
IRBP P2 peptide immunization and tetramer analysis. Mice were two PHD-type zinc-finger domains. Nat. Genet. 17:399403. http://
immunized with 100 g P2 peptide (IRBP; aa 271290) emulsified in com- dx.doi.org/10.1038/ng1297-399
plete Freunds adjuvant as described previously (Taniguchi et al., 2012). Cummings, S.R., J. San Martin, M.R. McClung, E.S. Siris, R. Eastell, I.R.
Tetramer analysis was performed on pooled lymph nodes and spleen from Reid, P. Delmas, H.B. Zoog, M. Austin, A. Wang, et al. FREEDOM
treated mice 10 d after immunization. P2-I-Ab tetramer was generated by the Trial. 2009. Denosumab for prevention of fractures in postmenopausal
National Institutes of Health Tetramer Core Facility, and tetramer staining women with osteoporosis. N. Engl. J. Med. 361:756765. http://dx.doi
was performed as described previously (Taniguchi et al., 2012). After tetramer .org/10.1056/NEJMoa0809493
enrichment, cells were stained with antibodies for flow cytometry, and count- Derbinski, J., A. Schulte, B. Kyewski, and L. Klein. 2001. Promiscuous gene
ing beads (Invitrogen) were used to enumerate tetramer+ cells. expression in medullary thymic epithelial cells mirrors the peripheral
self. Nat. Immunol. 2:10321039. http://dx.doi.org/10.1038/ni723
B16 melanoma tumor challenge. B6.RAG1/TRP-1 TCR transgenic DeVoss, J., Y. Hou, K. Johannes, W. Lu, G.I. Liou, J. Rinn, H. Chang,
mice were injected subcutaneously in the left flank with 1.0 105 B16 mela- R.R. Caspi, L. Fong, and M.S. Anderson. 2006. Spontaneous autoim-
noma cells. For adoptive transfer studies, spleens from donor mice were munity prevented by thymic expression of a single self-antigen. J. Exp.
pooled and CD25-depleted before transfer into B6.RAG1/ hosts. Recipi- Med. 203:27272735. http://dx.doi.org/10.1084/jem.20061864
ent mice were inoculated with 7.5 104 B16 melanoma cells 7 d after trans- Fletcher, A.L., T.E. Lowen, S. Sakkal, J.J. Reiseger, M.V. Hammett, N.
fer. Tumor growth was monitored by taking measurements of length (L) and Seach, H.S. Scott, R.L. Boyd, and A.P. Chidgey. 2009. Ablation and
width (W), and tumor volume was calculated as LW2/2. For generation of regeneration of tolerance-inducing medullary thymic epithelial cells
after cyclosporine, cyclophosphamide, and dexamethasone treatment.
survival curves, death was defined as tumor size >1,000 mm3.
J. Immunol. 183:823831. http://dx.doi.org/10.4049/jimmunol.0900225
Gbler, J., J. Arnold, and B. Kyewski. 2007. Promiscuous gene expression and
Statistical analysis. Statistical analysis was performed using Prism 6.0 the developmental dynamics of medullary thymic epithelial cells. Eur.
(GraphPad Software). Mann-Whitney Rank sum testing was performed on J. Immunol. 37:33633372. http://dx.doi.org/10.1002/eji.200737131
tetramer analysis. Students t test was performed for TEC and lymphocyte Gardner, J.M., J.J. Devoss, R.S. Friedman, D.J. Wong,Y.X. Tan, X. Zhou, K.P.
analyses. Log-rank test was performed for Kaplan-Meier survival curves. Johannes, M.A. Su, H.Y. Chang, M.F. Krummel, and M.S. Anderson.
2008. Deletional tolerance mediated by extrathymic Aire-expressing
We thank T. Metzger, T. LaFlam, M. Cheng, and W. Purtha for critical reading of the cells. Science. 321:843847. http://dx.doi.org/10.1126/science.1159407
manuscript. We thank the National Institutes of Health Tetramer Core Facility for Gray, D., J. Abramson, C. Benoist, and D. Mathis. 2007. Proliferative arrest
providing tetramer reagent. and rapid turnover of thymic epithelial cells expressing Aire. J. Exp. Med.
This work was supported by the US National Institutes of Health Grants 204:25212528. http://dx.doi.org/10.1084/jem.20070795
AI097457 (M.S. Anderson) and K12-GM081266 (M.L. Mouchess), and the UCSF Hikosaka, Y., T. Nitta, I. Ohigashi, K. Yano, N. Ishimaru, Y. Hayashi, M.
Medical Scientist Training Program (I.S. Khan). Flow cytometry data were generated Matsumoto, K. Matsuo, J.M. Penninger, H.Takayanagi, et al. 2008.The
in the UCSF Parnassus Flow Cytometry Core, which is supported by the Diabetes cytokine RANKL produced by positively selected thymocytes fosters
and Endocrinology Research Center (DERC) grant NIH P30 DK063720. medullary thymic epithelial cells that express autoimmune regulator.
The authors declare no competing financial interests. Immunity. 29:438450. http://dx.doi.org/10.1016/j.immuni.2008.06.018

JEM Vol. 211, No. 5 767


Kearns, A.E., S. Khosla, and P.J. Kostenuik. 2008. Receptor activator of nuclear et al. 2012. Rank signaling links the development of invariant T cell
factor kappaB ligand and osteoprotegerin regulation of bone remodeling progenitors and Aire(+) medullary epithelium. Immunity. 36:427437.
in health and disease. Endocr. Rev. 29:155192. http://dx.doi.org/10 http://dx.doi.org/10.1016/j.immuni.2012.01.016
.1210/er.2007-0014 Rossi, S.W., M.Y. Kim, A. Leibbrandt, S.M. Parnell, W.E. Jenkinson, S.H.
Kyewski, B., and L. Klein. 2006. A central role for central tolerance. Annu. Glanville, F.M. McConnell, H.S. Scott, J.M. Penninger, E.J. Jenkinson,
Rev. Immunol. 24:571606. http://dx.doi.org/10.1146/annurev.immunol et al. 2007. RANK signals from CD4+3 inducer cells regulate develop-
.23.021704.115601 ment of Aire-expressing epithelial cells in the thymic medulla. J. Exp.
Liston, A., S. Lesage, J. Wilson, L. Peltonen, and C.C. Goodnow. 2003. Med. 204:12671272. http://dx.doi.org/10.1084/jem.20062497
Aire regulates negative selection of organ-specific T cells. Nat. Immunol. Shum, A.K., J. DeVoss, C.L. Tan, Y. Hou, K. Johannes, C.S. OGorman,
4:350354. http://dx.doi.org/10.1038/ni906 K.D. Jones, E.B. Sochett, L. Fong, and M.S. Anderson. 2009.
Malchow, S., D.S. Leventhal, S. Nishi, B.I. Fischer, L. Shen, G.P. Paner, A.S. Identification of an autoantigen demonstrates a link between interstitial
Amit, C. Kang, J.E. Geddes, J.P. Allison, et al. 2013. Aire-dependent thy- lung disease and a defect in central tolerance. Sci. Transl. Med. 1:9ra20.
mic development of tumor-associated regulatory T cells. Science. 339:1219 http://dx.doi.org/10.1126/scitranslmed.3000284
1224. http://dx.doi.org/10.1126/science.1233913 Swann, J.B., and M.J. Smyth. 2007. Immune surveillance of tumors. J. Clin.
Metzger, T.C., and M.S. Anderson. 2011. Control of central and periph- Invest. 117:11371146. http://dx.doi.org/10.1172/JCI31405
eral tolerance by Aire. Immunol. Rev. 241:89103. http://dx.doi.org/10 Taniguchi, R.T., J.J. DeVoss, J.J. Moon, J. Sidney, A. Sette, M.K. Jenkins, and
.1111/j.1600-065X.2011.01008.x M.S. Anderson. 2012. Detection of an autoreactive T-cell population
Muranski, P., A. Boni, P.A. Antony, L. Cassard, K.R. Irvine, A. Kaiser, C.M. within the polyclonal repertoire that undergoes distinct autoimmune
Paulos, D.C. Palmer, C.E. Touloukian, K. Ptak, et al. 2008. Tumor- regulator (Aire)-mediated selection. Proc. Natl. Acad. Sci. USA. 109:7847
specific Th17-polarized cells eradicate large established melanoma. Blood. 7852. http://dx.doi.org/10.1073/pnas.1120607109
112:362373. http://dx.doi.org/10.1182/blood-2007-11-120998 Trger, U., S. Sierro, G. Djordjevic, B. Bouzo, S. Khandwala, A. Meloni, M.
Nagamine, K., P. Peterson, H.S. Scott, J. Kudoh, S. Minoshima, M. Heino, K.J. Mortensen, and A.K. Simon. 2012. The immune response to melanoma
Krohn, M.D. Lalioti, P.E. Mullis, S.E. Antonarakis, et al. 1997. Positional is limited by thymic selection of self-antigens. PLoS ONE. 7:e35005.
cloning of the APECED gene. Nat. Genet. 17:393398. http://dx.doi http://dx.doi.org/10.1371/journal.pone.0035005
.org/10.1038/ng1297-393 Zhu, M.L., A. Nagavalli, and M.A. Su. 2013. Aire deficiency promotes
Roberts, N.A., A.J. White, W.E. Jenkinson, G. Turchinovich, K. Nakamura, TRP-1-specific immune rejection of melanoma. Cancer Res. 73:2104
D.R. Withers, F.M. McConnell, G.E. Desanti, C. Benezech, S.M. Parnell, 2116. http://dx.doi.org/10.1158/0008-5472.CAN-12-3781

768 RANKL blockade enhances anti-tumor immunity | Khan et al.


Brief Definitive Report

T cellderived interleukin (IL)-21 promotes


brain injury following stroke in mice
Benjamin D.S. Clarkson,1,3 Changying Ling,1 Yejie Shi,2 Melissa G. Harris,1,4
Aditya Rayasam,4 Dandan Sun,2,5 M. Shahriar Salamat,1 Vijay Kuchroo,6
John D. Lambris,7 Matyas Sandor,1 and Zsuzsanna Fabry1
1Department of Pathology and Laboratory Medicine, 2Department of Neurological Surgery, 3Department of Cellular
and Molecular Pathology, 4Neuroscience Training Program, School of Medicine and Public Health, University
of Wisconsin-Madison, Madison, WI 53792
5Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Educational and Clinical Center, Pittsburgh, PA 15213
6Center for Neurological Diseases, Brigham and Womens Hospital Harvard Medical School, Boston, MA 02115
7Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104

T lymphocytes are key contributors to the acute phase of cerebral ischemia reperfusion
injury, but the relevant T cellderived mediators of tissue injury remain unknown. Using a
mouse model of transient focal brain ischemia, we report that IL-21 is highly up-regulated
in the injured mouse brain after cerebral ischemia. IL-21deficient mice have smaller
infarcts, improved neurological function, and reduced lymphocyte accumulation in the
brain within 24 h of reperfusion. Intracellular cytokine staining and adoptive transfer
experiments revealed that brain-infiltrating CD4+ T cells are the predominant IL-21 source.
Mice treated with decoy IL-21 receptor Fc fusion protein are protected from reperfusion
injury. In postmortem human brain tissue, IL-21 localized to perivascular CD4+ T cells in the
area surrounding acute stroke lesions, suggesting that IL-21mediated brain injury may be
relevant to human stroke.

CORRESPONDENCE Stroke is one of the leading causes of death and Conventionally, the protective role of T cells has
Zsuzsanna Fabry: disability worldwide. Clinical and preclinical ex- been attributed to the accumulation of regula-
zfabry@facstaff.wisc.edu
perimental studies highlight the importance of tory T cells within the CNS in later stages of re-
Abbreviations used: BA, basilar inflammation in both acute and delayed neuro- perfusion injury. These T cells produce a variety
artery; ECA, external carotid nal tissue damage after ischemic stroke; however, of cytokines including TGF and IL-10, which
artery; ICA, internal carotid the mechanisms and cells involved in this neuro- are both antiinflammatory and neuroprotective.
artery; I/R, ischemia/reperfu-
sion; MCA, middle cerebral inflammation are not fully understood. There is (Liesz et al., 2009; Stubbe et al., 2013). In addi-
artery; PCA, posterior cerebral currently no available treatment targeting the tion to having an established role in delayed
artery; PComA, posterior com- acute immune response that develops in the neuroprotection, Kleinschnitz et al. (2013) have
municating artery; RAG,
recombination activating
brain after transient focal ischemia. Therefore, recently shown that CD4+ CD25+ regulatory
gene; rCBF, regional cerebral we sought to identify novel T cellderived cyto- T cells also promote acute ischemic injury through
blood flow; tMCAO, transient kines that contribute to acute cerebral reperfu- interaction with the cerebral vasculature. The
MCA occlusion; TTC, 2,3,5-
triphenyltetrazolium chloride;
sion using the mouse model of transient middle acute detrimental effects can be further divided
VA, vertebral artery. cerebral artery occlusion (tMCAO). into early (24 h) and late (72 h) phases, with
During the reperfusion of infarcted brain tis- IL-17 production by nonconventional T cells
sue, leukocytes accumulate in the injured brain (less common T cell subset associated with mu-
where, in addition to clearing cell debris, they cosal tissues) possibly accounting for the latter by
promote secondary tissue injury (Yilmaz and promoting neutrophil accumulation (Gelderblom
Granger, 2010). Within the acute phase of isch- et al., 2012).
emic reperfusion (I/R) injury there are multiple The mechanisms of the early detrimental ef-
waves of cell infiltration of macrophages, neutro- fects of T cells after cerebral ischemia are least
phils, and lymphocytes (Gelderblom et al., 2009).
Brain-infiltrating T cells have also been widely 2014 Clarkson et al. This article is distributed under the terms of an Attribution
NoncommercialShare AlikeNo Mirror Sites license for the first six months
reported in stroke and animal models of stroke after the publication date (see http://www.rupress.org/terms). After six months
and are thought to have acute detrimental and it is available under a Creative Commons License (AttributionNoncommercial
Share Alike 3.0 Unported license, as described at http://creativecommons.org/
delayed protective effects (Magnus et al., 2012). licenses/by-nc-sa/3.0/).

The Rockefeller University Press $30.00


J. Exp. Med. 2014 Vol. 211 No. 4 595-604 595
www.jem.org/cgi/doi/10.1084/jem.20131377
understood. Several laboratories have reported reduced neuro- of several previously reported inflammatory genes, we found
logical deficit and infarct volumes at 2448 h reperfusion in that IL-21 was one of the most highly expressed inflammatory
T celldeficient mice after tMCAO (Yilmaz and Granger, 2010). genes among those measured (Fig. 1 a). Gene expression levels
After tMCAO, recombination activating gene 1deficient from arrays were normalized to interquartile spot intensity.
(RAG1 KO) mice, which lack T and B lymphocytes, have sig- Arrays did not differ systemically in gene expression levels be-
nificantly smaller brain injury compared with controls; whereas, fore or after normalization (not depicted).This increase in IL-21
adoptive transfer of WT CD4+ helper T cells or CD8+ cyto gene expression was confirmed by real-time (RT) PCR analy-
toxic T cells increases stroke infarct volumes within 24 h after sis, which detected a >24-fold relative increase in IL-21 gene
ischemia in these mice (Kleinschnitz et al., 2010). Additionally, expression in the ipsilateral ischemic brain tissues compared
TCR-transgenic mice and mice lacking co-stimulatory TCR with the contralateral hemisphere at 24 h reperfusion (Fig. 1 b).
signaling molecules were fully susceptible to acute I/R injury, IL-21 was not detectable by this method in healthy brain tis-
indicating that T cell involvement at early time points is anti- sue (Fig. 1 b).
gen-independent (Kleinschnitz et al., 2010). These data dem-
onstrate that conventional CD4+ or CD8+ T cells exacerbate
acute injury after cerebral ischemia independently of TCR li- IL-21deficient mice are protected from acute
gation, and this effect seems to be concomitant with an early neuronal injury after cerebral I/R injury
increase in T cell infiltration into the postischemic brain, which Whether IL-21 contributes to ischemic tissue injury had not
many have reported to be between 3 and 48 h (Yilmaz et al., been directly studied. However, in the last few years it has be-
2006; Gelderblom et al., 2009). come evident that IL-21 expression is associated with acute
Recent findings suggest that, in the postischemic brain, rejection in mice after kidney, heart, or liver allograft (Baan et al.,
within hours of reperfusion T cells accumulate in postcapillary 2007; Hecker et al., 2009; Xie et al., 2010). Because these
segments of periinfarct inflamed cerebral microvasculature models also involve reperfusion of ischemic tissues, these find-
characterized by high endothelial expression of chemokines ings support the potential role of IL-21 in I/R injury.
and adhesion molecules. These postcapillary venules have been We evaluated the levels of cerebral I/R injury in IL-21
postulated to allow accumulating immune cells to activate each deficient (IL-21 KO) mice. Infarct volumes in IL-21 KO mice
other and promote platelet adhesion in a process termed were reduced to 35% of the infarct volumes observed in con-
thrombo-inflammation (Nieswandt et al., 2011). Much research genic C57BL6/J WT mice as early as 24 h after tMCAO, as
has been devoted to identifying T cell factors that promote measured by triphenyltetrazolium chloride (TTC; Fig. 1 c).
thrombo-inflammation (Barone et al., 1997; Hedtjrn et al., Similar effects were also seen at 4 d (not depicted) and 7 d after
2002; Yilmaz et al., 2006; Shichita et al., 2009; Gelderblom et al., tMCAO (Fig. 1 d), indicating that IL-21 contributes to both
2012); however, to our knowledge no study has yet identified immediate and delayed brain injury and suggesting that in the
the T cellderived factors responsible for the early increase in absence of IL-21 tissue repair can occur. Analysis of intracra-
infarct volumes at 24 h reperfusion. Here, we present data that nial vascular anatomy revealed no differences between WT and
identify IL-21 as a key CD4+ T cellderived inflammatory fac- IL-21 KO mice in the patency of the posterior communicating
tor that contributes to increased early ischemic tissue injury. artery that would account for the observed differences in tissue
injury (Fig. 1 g). Nor did we observe differences between WT
and IL-21 KO mice in heart rate, or blood pressure before or
RESULTS AND DISCUSSION after tMCAO (Fig. 1 h). WT and IL-21 KO CD4+ T cells,
Robust up-regulation of IL-21 during cerebral I/R injury CD8+ T cells, and CD11b+ myeloid cells showed no difference
IL-21 is closely related to IL-2 and IL-15 and signals through in IL-2, IL-17A, IFN-, or TNF production when stimulated
the IL-21 receptor, which is comprised of an IL-21specific in vitro (Fig. 2, gl). The reduction in infarct volumes corre-
subunit and a common subunit shared with IL-2, IL-7, IL-9, sponded with less weight loss (unpublished data), less spleen
and IL-15. IL-21 is known to regulate immune responses by atrophy (Fig. 2 c), and improved neurological functioning in
promoting antibody production, T cellmediated immunity, IL-21 KO mice compared with WT mice as assessed by both
and NK cell and CD8+ T cell cytotoxicity. Recently, others grip strength (Fig. 1 e) and Bederson (Fig. 1 f) scoring 1, 4, and
have shown that stress signals from necrotic tissue can induce 7 d after tMCAO.
rapid IL-21 production from naive T cells (Holm et al., 2009), We measured accumulation of monocytes and lympho-
and co-stimulation with TLR3 ligands during polyclonal T cell cytes in the brain after tMCAO in IL-21 KO and WT animals
activation significantly increases IL-21 secretion that contributes (gating strategy in Fig. 2 a).We did not observe significant dif-
to small intestine localized pediatric celiac disease (van Leeuwen ferences in the rate of accumulation of monocytes (CD45high
et al., 2013).To test whether IL-21 is up-regulated in brain after CD11b+Ly6chigh), microglia (CD45intCD11b+), B cells (B220+),
ischemic necrosis induced by MCAO and to better understand T cells (TCR+), NK, or NKT cells (NK1.1+) in the
the cytokines involved in T cellmediated cerebral I/R injury, ischemic brain of WT and IL-21 KO mice after 1, 4, and 7 d
we measured changes in inflammatory gene expression in the of reperfusion (unpublished data). In contrast, as early as 1 d
brain within 24 h after tMCAO in mice using PCR-based after tMCAO, IL-21 KO mice showed significantly dimin-
gene array analysis. In addition to verifying the up-regulation ished cerebral accumulation of CD4+ T cells and CD8+ T cells

596 IL-21 promotes brain injury after stroke | Clarkson et al.


Br ief Definitive Repor t

Figure 1. IL-21 is up-regulated early in


mouse brain, and IL-21deficient mice are
protected after tMCAO. GeArray S Series
Mouse Autoimmune and Inflammatory Re-
sponse gene array of transcripts expressed in
pooled brain tissues 24 h after tMCAO or
sham procedure (n = 36 mice per group).
(a) Bar graphs show PCR array spot intensity
of genes with a greater than sixfold difference
in gene expression in tMCAO compared with
sham, normalized to the interquartile mean
spot intensity. (b) IL-21 mRNA expression
level in ipsilateral hemisphere relative to con-
tralateral hemisphere 24 h after tMCAO (n = 3
per group). Mann-Whitney rank sum test
*, P < 0.05. Plots show median, lower, and upper
quartile (box) and range (error bars). Infarct
volumes of WT and IL-21 KO mice 24 h after
tMCAO (c) and 7 d after tMCAO (d). Represen-
tative images of TTC-stained 2-mm brain
slices shown below (n = 57 mice per group).
WT and IL-21 KO mice grip strength (e) and
Bederson score (f) at 1, 4, and 7 d after 1 h
tMCAO (n = 78 for each group). (g) Brain
vasculature of C57BL/6 and IL-21KO mice
perfused transcardially with carbon lampblack
(C198-500; Thermo Fisher Scientific) in 20%
gelatin ddH2O. Arrows indicate anterior cere-
bral (ACA), MCA, posterior cerebral (PCA),
basilar (BA), and vertebral arteries (VA), show-
ing point of occlusion (white circle). High
magnification images demonstrate no differ-
ence in patency of the posterior communicat-
ing artery (PComA, arrows). (h) Heart rate and
blood pressure of WT and IL-21 KO mice be-
fore and after tMCAO (n = 34 mice per
group). Data are representative of 23 inde-
pendent experiments. **, P < 0.01; ***, P <
0.001; ****, P < 0.0001 by Students t test
(single comparison) or one-way ANOVA (mul-
tiple comparisons). Error bars indicate SD.

after tMCAO compared with WT mice (Fig. 2 e) and these in either tissue compared with WT experimental animals. Nor
differences persisted at day 7 (Fig. 2 f ). These differences were did we observe a difference between WT and IL-21 KO mice
not reflected in the spleen before or after tMCAO (Fig. 2 b). in the frequency of lymphocytes producing the antiinflam-
IL-21 has also been shown to be produced by and modulate matory cytokine IL-10 among B cells (B220+), CD8+ T cells,
the function of regulatory T cells (Peluso et al., 2007; Battaglia or CD4+ T cells (unpublished data). These data demonstrate
et al., 2013), which begin to accumulate in the brain after that IL-21 deficiency is protective at acute time points after
tMCAO. Thus, we compared the frequency of regulatory tMCAO and IL-21 levels in the CNS correlate with early in-
CD4 T cells expressing the marker Foxp3 in the brain and filtration of T cells without affecting regulatory T or B cell
spleen 24 h after tMCAO in WT and IL-21 KO mice. IL-21 accumulation or IL-10 cytokine production during the acute
KO mice exhibited no difference in regulatory T cell abundance period (day 14).

JEM Vol. 211, No. 4 597


Figure 2. Lymphocyte recruitment to brain is diminished in IL-21 deficient mice. (a) Gating strategy for leukocytes isolated from brain after
MCAO. (b) WT and IL-21 KO spleen cells 24 h after tMCAO or sham procedure (n = 3 mice per group). (c) Relative change in spleen weight of WT and IL-21
KO mice after tMCAO (n = 37 mice per group). (d) Percentage of blood and spleen CD4+ T cells expressing IL-21 after 5-h ex vivo stimulation with PMA
(10 ng/ml) and Ionomycin (1 g/ml) 4 d after MCAO or control treatment. (e and f) Leukocyte accumulation in the brain of WT mice compared with IL-21
KO mice 1, 4, and 7 d after tMCAO (n = 36 mice per group). (gk) In vitro cytokine expression by WT and IL-21 KO CD4+ and CD8+ T cells after 5-h
stimulation under indicated conditions with or without recombinant mouse IL-21 (100 ng/ml). (l) TNF production by CD11b+ myeloid cells stimulated with
LPS (500 ng/ml) for 5 h with or without recombinant mouse IL-21 (100 ng/ml). Cells isolated from n = 3 mice per group. Data are representative of two to
four independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001 by Students t test (single comparison) or one-way ANOVA (multiple
comparisons). Error bars indicate SD (bd and gl) and SEM (e, f).

598 IL-21 promotes brain injury after stroke | Clarkson et al.


Br ief Definitive Repor t

IL-21 is primarily produced IL-21 blockade is protective in tMCAO


by brain-infiltrating CD4+ T cells We treated WT mice with IL-21 receptor Fc protein (IL-21R.
We measured intracellular IL-21 production by various cell Fc) using a previously described protocol (Jang et al., 2009;
populations. IL-21producing cells were not detected by flow McGuire et al., 2011; Spolski et al., 2012). We administered
cytometry among cells isolated from healthy brain, but could 500 g of IL-21R.Fc i.p. 1 h before tMCAO. As measured by
be detected among mononuclear cells isolated from isch- TTC staining, treated mice showed significantly reduced in-
emic brain 24 h after tMCAO. Gating on IL-21+ cells revealed farct volumes compared with control-treated mice 24 h after
that the majority of these cells were CD4+ T cells (Fig. 3 a). tMCAO (Fig. 4 a). We found a similarly protective effect in
IL-21producing CD4+ T cells were also detected at low lev- mice treated with IL-21R.Fc protein (500 g i.p.) 2 h after ini-
els among cells isolated from blood, but not spleen, of healthy tiation of reperfusion (Fig. 4 a). These differences were associ-
WT mice, and these levels were unaffected after transient ce- ated with decreased locomotor function (decreased resistance
rebral ischemia (Fig. 2 d), indicating that the increase in IL-21 to lateral push and increased circling behavior) in control-
production was limited to CD4+ T cells recovered from the treated mice compared with those treated with IL-21R.Fc
postischemic brain. Next, we adoptively transferred WT or (Fig. 4 b and Video 1). Although we cannot exclude the pos-
IL-21 KO CD4+ T cells into lymphocyte-deficient RAG2 sibility that IL-21R.Fc exhibits its blocking effect in periph-
KO mice. Purity of transferred CD4+ T cells was confirmed eral immune compartments, using ELISA for human IgG4 we
by flow cytometry to be 95% (Fig. 3 b). As shown previ- were able to observe thatupon i.p. injectionsoluble IL-21R.
ously (Kleinschnitz et al., 2010), we observed markedly re- Fc accumulates in the CNS of mice after tMCAO (Fig. 4 c).
duced infarcts in RAG KO mice compared with WT mice,
and infarct volumes could be restored to WT levels in RAG2 IL-21 presence localizes with CD4 staining
KO by adoptively transferring WT CD4+ T cells. Most im- in human stroke tissue
portantly, RAG2 KO mice that received WT CD4+ T cells IL-21+ cells were recently detected in human brain tissue
had significantly larger infarcts than those receiving IL-21 during different neuroinflammatory conditions (Tzartos et al.,
KO CD4+ T cells (Fig. 3 c). 2011).Thus, we stained groups of postmortem brain tissue from

Figure 3. IL-21 is primarily produced by brain-infiltrating CD4+ T cells. (a) Intracellular cytokine staining of lymphocytes isolated from n = 5
pooled healthy WT, ischemic IL-21/, or ischemic WT mouse brains 24 h after tMCAO or sham procedure showing IL-21 versus CD8 expression. Histo-
grams show CD4, NK1.1, and TCR expression on IL-21+ cells from ischemic WT brain. (b) CD45, CD4, and LFA-1 expression by negative fractions purified
from WT and IL-21/ lymph node cells by CD4+ negative selection using magnetic cell separation before transfer into RAG2/ recipients. (c) Infarct
volume in WT mice (n = 4), RAG2/ mice (n = 4), RAG2/ mice + WT CD4 T cells (n = 10), and RAG2/ mice + IL-21/ CD4 T cells (n = 10) 24 h
after tMCAO. Representative TTC-stained 2-mm mouse brain slices shown on top. Data are representative of two independent experiments. **, P < 0.01;
***, P < 0.001; ****, P < 0.0001 one-way ANOVA. Error bars indicate SD.

JEM Vol. 211, No. 4 599


Figure 4. Blockade of IL-21 signaling before or after tMCAO reduces infarct size in WT mice. (a) Infarct volumes 24 h after tMCAO in WT mice
treated with 500 g recombinant mIL-21R.Fc or PBS 1 h before (pretreatment) or 2 h after (posttreatment) surgery. Representative TTC-stained brain
slices shown on left (n = 34 mice per group). (b) Still image from Video 1 depicting behavioral differences between WT mice posttreated with IL-21R.Fc
or PBS. (c) IL-21R.Fc protein levels in the indicated organs 2024 h after tMCAO in WT mice injected with 500 g IL-21R.Fc 2 h after start of reperfusion
(n = 24 mice per group). N.D., not detected. Data are representative of two independent experiments. **, P < 0.01; ***, P < 0.001, by Students t test. Error
bars indicate SEM. Representative images of postmortem paraffin-embedded human acute stroke lesions stained with control sera (d), or primary anti-
bodies against CD4 (e and g [ii-iii]), IL-21 (f and g [iii]), or eosin (g [i]) visualized with Fast Red (d, e, and g) and/or DAB (d, f, and g [iii]) and counterstained
with hematoxylin. High magnification images are shown on right. Arrows indicate positive staining. Bars, 50 m.

600 IL-21 promotes brain injury after stroke | Clarkson et al.


Br ief Definitive Repor t

patients with acute and chronic stroke lesions. In acute infarcts, T cells that can secrete IL-21 were detected within the
rare CD4+ T cells were found in the necrotic brain parenchyma CSF-filled subarachnoid and perivascular spaces during cere-
(Fig. 4 g, ii, arrows), which was predominantly infiltrated by bral infarction in humans.
foamy macrophages (Fig. 4 g, i). In contrast, CD4+ T cells were
consistently found within the Virchow Robins space of vessels Neuronal cells express IL-21 receptor and up-regulate
bordering acute infarcts (not depicted) and in the subarachnoid autophagy genes in response to IL-21
space adjacent to meningeal vessels (arrows, Fig. 4 e, i). IL-21 RT-PCR analysis of primary mouse neurons and murine neu-
staining was limited almost exclusively to these perivascular ronal cell lines (Neuro2A) indicated that IL-21R expression
spaces. Compared with control stained tissue (Fig. 4 d, i), anti was higher on neuronal cells than on other brain cells, includ-
IL-21 staining labeled cells extensively in the subarachnoid ing astrocytes and endothelial cells (Fig. 5, a and c). This is con-
space of deep sulci penetrating the infarcted tissueshowing sistent with another report where in situ hybridization detected
a similar distribution to CD4+ cells in serial sections (arrows, neuron-restricted IL-21R expression in inflamed human brain
Fig. 4 f ). Additionally, double staining with antibodies for tissue (Tzartos et al., 2011). Moreover, treating Neuro2a cells
CD4 and IL-21 revealed the presence of CD4+ IL-21+ cells with IL-21 after in vitro oxygen glucose deprivation (OGD)
in this perivascular niche (arrow, Fig. 4 g, iii). In summary, CD4+ significantly increased cell death as measured by XTT cell

Figure 5. IL-21 promotes autophagy expres-


sion in neuronal cells after hypoxia/ischemia.
(a) Il21r mRNA expression relative to GAPDH
expression levels is shown in normoxic and hy-
poxic primary mouse neurons after OGD or con-
trol treatment. (b) Viability of Neuro2A cells
treated with the indicated doses of IL-21 after
OGD. (c) Il21r mRNA expression relative to
GAPDH in neuronal (Neura2A), astrocytic, and
endothelial cell lines (MB114) expressed relative
to BMDC expression. (d) ATG6 expression in pri-
mary neurons treated with PBS, etoposide, or
32256 ng/ml rIL-21 for 4 h after 12 h oxygen
glucose deprivation as measured by RT-PCR. Cells
treated in triplicate. (e) Number of ATG6+ cells per
field in the same regions of WT and IL-21 KO
mouse brains after tMCAO as assessed by im-
mune staining (n = 3 mice per group). Arrows
indicate ATG-6+ cells in periinfarcted brain tissue
of WT and IL-21 KO mice. Bars, 100 m. Data
are representative of two independent experi-
ments. *, P < 0.05; **, P < 0.01; ***, P < 0.001;
****, P < 0.0001 by Students t test (single com-
parison) or one-way ANOVA (multiple compari-
sons). Error bars indicate SEM.

JEM Vol. 211, No. 4 601


viability assay (Fig. 5 b). In subsequent studies we found that of the superior thyroid artery, the ECA was dissected distally and coagulated
treatment of primary neurons with IL-21 up-regulated mRNA along with the terminal lingual and maxillary artery branches.The internal ca-
rotid artery (ICA) was isolated, and the extracranial branch of the ICA was then
levels of the autophagy associated gene ATG6 (Fig. 5 d). These
dissected and ligated. A standardized polyamide resin glue-coated 6.0 nylon
data suggest that IL-21 could directly affect neuronal autoph- monofilament (3021910; Doccol Corp) was introduced into the ECA lumen,
agy during ischemic injury, which has been implicated in neu- and then advanced 99.5 mm in the ICA lumen to block MCA blood flow.
ronal death in infarcted and periinfarcted brain tissue.Thus, we During the entire procedure, mouse body temperature was kept between 37
stained WT and IL-21 KO postischemic brain tissues for ATG6. and 38C with a heating pad. The suture was withdrawn 60 min after occlu-
We observed significantly fewer ATG6+ cells in infarcted brain sion. The incision was closed, and the mice underwent recovery.
tissue of IL-21 KO mice compared with WT, suggesting that
Infarction size measurement. After 24 h reperfusion, mice were sacrificed
IL-21 may contribute to increased cerebral autophagy after
and brains were removed and frozen at 80C for 5 min. 2-mm coronal
stroke (Fig. 5 e). slices were made with a rodent brain matrix (Ted Pella, Inc.). The sections
In conclusion, we implicate IL-21 as a lymphocyte-derived were stained for 20 min at 37C with 2% TTC (Sigma-Aldrich). Infarction
factor with a pronounced effect on brain injury after focal isch- volume was calculated with the method reported by Swanson et al. (1990) to
emia in mice. We also present data demonstrating that IL-21 compensate for brain swelling in the ischemic hemisphere. In brief, the sec-
producing CD4+ T cells are present in the brain of patients tions were scanned, and the infarction area in each section was calculated by
subtracting the noninfarct area of the ipsilateral side from the area of the
with acute stroke.These data warrant investigation of the thera-
contralateral side with National Institutes of Health image analysis software,
peutic potential of IL-21modifying treatments in isolation ImageJ. Infarction areas on each section were summed and multiplied by sec-
and combination with current anti-thrombotic treatments for tion thickness to give the total infarction volume.
ischemic stroke.
Gene array and RT-PCR. Ipsilateral brain hemispheres were dissected and
stored in RNAlater (QIAGEN) at 4C until further use. Total RNA was ex-
MATERIALS AND METHODS tracted and purified with RNeasy Protect Mini kit (QIAGEN) according to
Ethics statement. C57BL/6 WT mice were obtained from The Jackson the manufacturers instructions. Purified RNA samples were analyzed by
Laboratory. IL-21deficient mice (IL-21tm1Lex) were purchased from the Mu- GeArray S Serious Mouse Autoimmune and Inflammatory Gene array
tant Mouse Regional Resources Center. All mice underwent 1 h tMCAO (SuperArray; Bioscience Corporation). Results from GeArray were filtered
and 24 h reperfusion. All animal procedures used in this study were con- for genes with spot intensities higher than the mean local background of the
ducted in strict compliance with the National Institutes of Health Guide for bottom 75% of nonbleeding spots. For RT-PCR, 1 g total RNA from each
the Care and Use of Laboratory Animals and approved by the University of sample was reverse transcribed using SuperScript II first strand cDNA syn-
Wisconsin Center for Health Sciences Research Animal Care Committee. thesis kit (Invitrogen). RT-PCR was performed on a Smart Cycler (Model SC
All mice (25 g) were anesthetized with 5% halothane for induction and 1001; Cepheid) using IL-21 TaqMan gene expression assay (Mm00517640_m1;
1.0% halothane for maintenance vaporized in N2O and O2 (3:2), and all ef- Applied Biosystems), RT2 qPCR Primer assay for mouse Becn1
forts were made to minimize suffering. (PPM32434A; SABiosciences), or RT2 qPCR Primer Assay for Mouse IL21r
(PPM03762A; SABiosciences). The data were normalized to an internal ref-
Regional cerebral blood flow (rCBF) measurement. Changes in rCBF erence gene, GAPDH.
at the surface of the left cortex were recorded using a blood perfusion monitor
(Laserflo BPM2;Vasamedics) with a fiber optic probe (0.7 mm diam). The tip
Mononuclear cell isolation and flow cytometry. Brains were removed
of the probe was fixed with glue on the skull over the core area supplied by the
from perfused animals, weighed, minced, transferred to Medicon inserts, and
MCA (2 mm posterior and 6 mm lateral from bregma). Changes in rCBF after
ground in a MediMachine (BD) for 2030 s.The cell suspension was washed
MCAO were recorded as a percentage of the baseline value. Mice included in
with HBSS, and cells were resuspended in 70% Percoll (Pharmacia) and over-
these investigations had >80% relative decrease in rCBF during MCAO.
laid with 30% Percoll. The gradient was centrifuged at 2,250 g for 30 min at
4C without brake. The interface was removed and washed once for further
Investigation of intracranial vasculature. WT and IL-21 KO mice were
analysis. CD11b-positive and -negative fractions were isolated using Imag
anesthetized with ketamine (100 mg/kg, i.p.) and xylazine (10 mg/kg, i.p.).
anti-CD11b magnetic particles (BD), following the manufacturers protocol.
After thoracotomy was performed, a cannula was introduced into the ascend-
A total of 106 cells were incubated for 30 min on ice with saturating concen-
ing aorta through the left ventricle. Transcardial perfusion fixation was per-
trations of labeled antibodies with 40 g/ml unlabeled 2.4G2 mAb to block
formed with 2 ml saline and 2 ml of 3.7% formaldehyde. Carbon lampblack
binding to Fc receptors, and then washed 3 times with 1% BSA in PBS.
(C198-500; Thermo Fisher Scientific) in an equal volume of 20% gelatin in
Single-cell suspensions from various tissues were cultured at 37C in
ddH2O (1 ml) was injected through the cannula. The brains were removed
10% FBS in RPMI 1640 media supplemented with GolgiStop (BD) in the pres-
and fixed in 4% PFA overnight at 4C. Posterior communicating arteries
ence of either phorbol myristate acetate (50 ng/ml) and ionomycin (1 g/ml)
(PComA) connect vertebrobasilar arterial system to the Circle of Willis and
for 5 h. After surface staining with antibodies against CD4, NK1.1, and
internal carotid arteries, and its development affects brain sensitivity to isch-
TCR, cell suspensions were fixed and permeabilized by Cytofix/Cy-
emia among different mouse strains (Barone et al., 1993). Development of
toperm solution (BD), followed by staining with antiIL-21 antibodies.
PComA in both hemispheres was examined and graded on a scale of 03, as
Fluorochrome-labeled antibodies against CD45, CD11b, Ly6c, B220, CD4,
reported previously (Majid et al., 2000). 0, no connection between anterior
CD8a, NK1.1, IFN-, and appropriate isotype controls were purchased from
and posterior circulation; 1, anastomosis in capillary phase (present but poorly
BD. Fluorochrome-labeled antibody against IL-21 and TCR was pur-
developed); 2, small truncal PComA; 3, truncal PComA.
chased from eBioscience. Cell staining was acquired on a FACSCalibur or
LSRII (BD) and analyzed with FlowJo (Tree Star) software version 5.4.5.
Focal ischemia model. Focal cerebral ischemia in mice was induced by oc-
clusion of the left MCA, as described previously (Longa et al., 1989). Operators Neurofunctional assessment. Neuromuscular coordination was assessed by
performing surgeries were masked to experimental groups. In brief, the left grip strength test, as previously described (Kleinschnitz et al., 2010). For this
common carotid artery was exposed, and the occipital artery branches of the test, mice were placed on a horizontal string midway between two supports.
external carotid artery (ECA) were isolated and coagulated. After coagulation Mice were scored from 0 to 5 as follows: 0, falls off within 2 s; 1, hangs on with

602 IL-21 promotes brain injury after stroke | Clarkson et al.


Br ief Definitive Repor t

forepaw(s); 2, hangs on with forepaws and moves laterally on string; 3, hangs Statistical analyses and quality standards. All surgeries were performed
onto string with forepaws and hindpaw(s); 4, hangs onto string with forepaws, in a blinded manner by a third party and measurements masked where possi-
hindpaw(s) and tail; 5, escape to supports. Mice were allowed to rest between ble. Infarct volume measurements from TTC stained sections were averaged
trials. Scores for each mouse were determined by averaging 510 trials (each from two to three independent blinded observers. Based on power calcula-
lasting 15 s). Global neurological deficit was determined by a modified Bed- tions, n = 310 sex- and age-matched mice were used for each experiment
erson scoring system: 0, no deficit; 1, forelimb flexion; 2, unidirectional circling and group assignment was randomized. Among animals receiving MCAO
after being lifted by tail; 3, spontaneous unidirectional circling; 4, longitudinal procedure, 86.5% of WT mice, 93.5% of IL-21KO mice, and 100% of
rolling upon being lifted by tail; 5, spontaneous longitudinal rolling. RAG2KO mice were included in analysis. Mice were excluded due to prema-
ture death (13.5% of WT mice, 3.2% of IL-21 KO mice) or vessel variation
Generation of IL-21 receptor Fc fusion protein. Chinese hamster ovary (3.2% of IL-21KO mice). Results are given as means 1 SD. Multiple com-
cell line (Korn et al., 2007) expressing the extracellular domain (aa 20236) parisons were made using one-way ANOVA. Where appropriate, two-tailed
of mouse IL-21R fused to the fragment crystallizable (Fc) portion of human Students t test analysis was used for comparing measures made between two
IgG4 (IL-21R.Fc) were maintained in UltraCHO (BioWhittaker). IL-21R. groups. For comparison of RT-PCR data, nonparametric Mann-Whitney
Ig was purified from the culture supernatant by passage through a protein rank sum analysis was used. P-values <0.05 were considered significant.
GSepharose column and concentrated by ultrafiltration. Concentration was
determined spectrophotometrically. Purity and molecular weight were con- Online supplemental material. Video 1 shows groups of WT C57BL6
firmed by sodium dodecyl-sulfate PAGE and human-IgG4 ELISA (eBiosci- mice treated with 500 g IL-21R.Fc or PBS control via i.p. injection. On-
ence) following the manufacturers instructions. The IL-21R.Fc reagent was line supplemental material is available at http://www.jem.org/cgi/content/
tested in vitro for its ability to suppress IL-21induced T cell proliferation. full/jem.20131377/DC1.

Immunohistochemistry. Paraffin-embedded postmortem brain tissue sec- We thank Satoshi Kinoshita for expert histopathology services, Guoqing Song for
tions from individuals with acute and chronic stroke lesions were obtained assisting in the surgical procedures, Dr. Wenda Gao for providing reagents and
from the Neuropathology Laboratory of the University of Wisconsin De- protocols for the purification of IL-21R.Fc protein, and members of our laboratory
partment of Pathology. After rehydration and deparaffinization, sections un- for helpful discussions and constructive criticisms of this work. We also thank Khen
derwent heat-induced antigen retrieval in 10 mM sodium citrate, pH 6.0, for Macvilay and Sinarack Macvilay for their expertise provided for cytofluorimetry and
surface antigens or Tris-EDTA (10 mM/1 mM) with 0.05% Tween-20 for immunohistochemistry studies and Samuel (Joe) Ollar for assisting in the OGD procedure.
intracellular antigens. Sections were blocked for 30 min with secondary This work was supported by awards from the American Heart Association (pre-
serum (10% in Tris-buffered saline) and then stained with primary antibodies, doctoral fellowship #12PRE12060020 to B.D.S. Clarkson) and the National Institutes
0.5% chicken antiIL-21 (Lifespan Biosciences) or prediluted mouse anti- of Health (NS037570 and NS076946 to ZF, AI048087 to M.S. Salamat, and
CD4 ([1F6]; ab17131; Abcam) for 12 h at 37C or overnight at 4C. Nor- AI068730 to J.D. Lambris).
mal primary sera (510%) were used for negative control. After several washes, The authors have no competing financial interests.
secondary antibodies (biotin-labeled goat antichicken or biotin-labeled
goat antimouse;Vector Laboratories) were applied to sections and incubated Submitted: 1 July 2013
for 2 h at room temperature. Staining was developed using the VECTA- Accepted: 24 February 2014
STAIN ABC-HRP kit (Vector Laboratories) with diaminobenzidine sub-
strate (BD) or streptavidin-alkaline phosphatase with Fast Red substrate REFERENCES
(Laboratory Vision), following the manufacturers instructions. Slides were Baan, C.C., A.H. Balk, I.E. Dijke, S.S. Korevaar, A.M. Peeters, R.P. de Kuiper, M.
lightly counterstained with hematoxylin, rinsed with running tap water, and Klepper, P.E. Zondervan, L.A. Maat, and W. Weimar. 2007. Interleukin-21:
mounted. For frozen mouse sections WT and IL-21 KO mice underwent 1-h an interleukin-2 dependent player in rejection processes. Transplantation.
tMCAO and 17-d reperfusion. Brains were perfused with PBS and 3% for- 83:14851492. http://dx.doi.org/10.1097/01.tp.0000264998.23349.54
malin, embedded in OCT, and cut into 8-m frozen sections for immuno Barone, F.C., D.J. Knudsen, A.H. Nelson, G.Z. Feuerstein, and R.N. Willette.
histochemistry. Frozen sections were thawed for 10 min at room temperature 1993. Mouse strain differences in susceptibility to cerebral ischemia are
and blocked with 5% goat serum solution in PBS for 15 min. Sections were related to cerebral vascular anatomy. J. Cereb. Blood Flow Metab. 13:683
stained with rabbit polyclonal antibodies for ATG6 for 1 h at room tempera- 692. http://dx.doi.org/10.1038/jcbfm.1993.87
ture, followed by phycoerythrin-labeled goat antirabbit IgG (Santa Cruz Barone, F.C., B. Arvin, R.F. White, A. Miller, C.L. Webb, R.N. Willette, P.G.
Biotechnology, Inc.). Images were acquired on a BX40 microscope equipped Lysko, and G.Z. Feuerstein. 1997.Tumor necrosis factor-alpha. A mediator
with a Q-Color 3 camera using Q-Capture software (Olympus). Digital of focal ischemic brain injury. Stroke. 28:12331244. http://dx.doi.org/
images were processed and analyzed using Photoshop CS4 software (Adobe). 10.1161/01.STR.28.6.1233
Color balance, brightness, and contrast settings were manipulated to generate Battaglia, A., A. Buzzonetti, C. Baranello, M. Fanelli, M. Fossati, V. Catzola,
final images. All changes were applied equally to entire image. G. Scambia, and A. Fattorossi. 2013. Interleukin-21 (IL-21) synergizes
with IL-2 to enhance T-cell receptor-induced human T-cell proliferation
Neuronal cell oxygen glucose deprivation. Primary neuronal cultures and counteracts IL-2/transforming growth factor--induced regulatory
derived from embryonic day 1418 mouse cortices were grown to 80% con- T-cell development.Immunology.139:109120.http://dx.doi.org/10.1111/
fluency in neural basal media supplemented with B27 (2%) and penicillin/ imm.12061
streptomycin (1%), as previously described (Kintner et al., 2010). Astrocytic Gelderblom, M., F. Leypoldt, K. Steinbach, D. Behrens, C.U. Choe, D.A. Siler,
T.V. Arumugam, E. Orthey, C. Gerloff, E. Tolosa, and T. Magnus. 2009.
and microglial contamination was excluded based on the absence of GFAP+
Temporal and spatial dynamics of cerebral immune cell accumulation in
and CD11b+ cells when stained by immunocytochemistry. For OGD, media
stroke. Stroke. 40:18491857. http://dx.doi.org/10.1161/STROKEAHA
was replaced with neural basal media with or without glucose and placed in .108.534503
a hypoxic chamber or under normoxic conditions for 2 h at 37C. Afterward, Gelderblom, M., A. Weymar, C. Bernreuther, J. Velden, P. Arunachalam, K.
cells were lysed and mRNA isolated using RNeasy mini kit (QIAGEN). For Steinbach, E. Orthey, T.V. Arumugam, F. Leypoldt, O. Simova, et al.
XTT viability assay, Neuro2A underwent OGD and were treated with dose 2012. Neutralization of the IL-17 axis diminishes neutrophil invasion
curve of IL-21 immediately after return to normoxic media and incubated and protects from ischemic stroke. Blood. 120:37933802. http://dx.doi
for 4 h at 37C. XTT labeling mixture (50 l per well; Roche) was added ac- .org/10.1182/blood-2012-02-412726
cording to manufacturers instructions, and at18 h fluorescence was read on Hecker,A.,A. Kaufmann, M. Hecker,W. Padberg, andV. Grau. 2009. Expression
a GENious Microplate reader (Tecan). Cell number was calculated using a of interleukin-21, interleukin-21 receptor alpha and related type I
standard curve of known untreated cells kept under normoxic conditions. cytokines by intravascular graft leukocytes during acute renal allograft

JEM Vol. 211, No. 4 603


rejection. Immunobiology. 214:4149. http://dx.doi.org/10.1016/j.imbio McGuire, H.M., S. Walters, A.Vogelzang, C.M. Lee, K.E. Webster, J. Sprent, D.
.2008.04.004 Christ, S. Grey, and C. King. 2011. Interleukin-21 is critically required in
Hedtjrn, M., A.L. Leverin, K. Eriksson, K. Blomgren, C. Mallard, and H. autoimmune and allogeneic responses to islet tissue in murine models.
Hagberg. 2002. Interleukin-18 involvement in hypoxic-ischemic brain Diabetes. 60:867875. http://dx.doi.org/10.2337/db10-1157
injury. J. Neurosci. 22:59105919. Nieswandt, B., C. Kleinschnitz, and G. Stoll. 2011. Ischaemic stroke: a
Holm, C.K., C.C. Petersen, M. Hvid, L. Petersen, S.R. Paludan, B. Deleuran, thrombo-inflammatory disease? J. Physiol. 589:41154123.
and M. Hokland. 2009. TLR3 ligand polyinosinic:polycytidylic acid Peluso, I., M.C. Fantini, D. Fina, R. Caruso, M. Boirivant, T.T. MacDonald,
induces IL-17A and IL-21 synthesis in human Th cells. J. Immunol. F. Pallone, and G. Monteleone. 2007. IL-21 counteracts the regulatory
183:44224431. http://dx.doi.org/10.4049/jimmunol.0804318 T cell-mediated suppression of human CD4+ T lymphocytes. J. Immunol.
Jang, E., S.H. Cho, H. Park, D.J. Paik, J.M. Kim, and J.Youn. 2009. A positive 178:732739.
feedback loop of IL-21 signaling provoked by homeostatic CD4+CD25- Shichita, T., Y. Sugiyama, H. Ooboshi, H. Sugimori, R. Nakagawa, I. Takada,
T cell expansion is essential for the development of arthritis in auto T. Iwaki,Y. Okada, M. Iida, D.J. Cua, et al. 2009. Pivotal role of cerebral in-
immune K/BxN mice. J. Immunol. 182:46494656. http://dx.doi.org/10 terleukin-17-producing gammadeltaT cells in the delayed phase of isch-
.4049/jimmunol.0804350 emic brain injury. Nat. Med. 15:946950. http://dx.doi.org/10.1038/
Kintner, D.B., X. Chen, J. Currie, V. Chanana, P. Ferrazzano, A. Baba, T. nm.1999
Matsuda, M. Cohen, J. Orlowski, S.Y. Chiu, et al. 2010. Excessive Na+/ Spolski, R., L. Wang, C.K. Wan, C.A. Bonville, J.B. Domachowske, H.P. Kim,
H+ exchange in disruption of dendritic Na+ and Ca2+ homeostasis Z. Yu, and W.J. Leonard. 2012. IL-21 promotes the pathologic immune
and mitochondrial dysfunction following in vitro ischemia. J. Biol. Chem. response to pneumovirus infection. J. Immunol. 188:19241932. http://
285:3515535168. http://dx.doi.org/10.1074/jbc.M110.101212 dx.doi.org/10.4049/jimmunol.1100767
Kleinschnitz, C., N. Schwab, P. Kraft, I. Hagedorn, A. Dreykluft, T. Schwarz, Stubbe, T., F. Ebner, D. Richter, O. Engel, J. Klehmet, G. Royl, A. Meisel,
M. Austinat, B. Nieswandt, H. Wiendl, and G. Stoll. 2010. Early detri- R. Nitsch, C. Meisel, and C. Brandt. 2013. Regulatory T cells accu-
mental T-cell effects in experimental cerebral ischemia are neither related mulate and proliferate in the ischemic hemisphere for up to 30 days
to adaptive immunity nor thrombus formation. Blood. 115:38353842. after MCAO. J. Cereb. Blood Flow Metab. 33:3747. http://dx.doi.org/10
http://dx.doi.org/10.1182/blood-2009-10-249078 .1038/jcbfm.2012.128
Kleinschnitz, C., P. Kraft, A. Dreykluft, I. Hagedorn, K. Gbel, M.K. Swanson, R.A., M.T. Morton, G. Tsao-Wu, R.A. Savalos, C. Davidson, and
Schuhmann, F. Langhauser, X. Helluy, T. Schwarz, S. Bittner, et al. 2013. F.R. Sharp. 1990. A semiautomated method for measuring brain infarct
Regulatory T cells are strong promoters of acute ischemic stroke in mice volume. J. Cereb. Blood Flow Metab. 10:290293. http://dx.doi.org/10
by inducing dysfunction of the cerebral microvasculature. Blood. 121:679 .1038/jcbfm.1990.47
691. http://dx.doi.org/10.1182/blood-2012-04-426734 Tzartos, J.S., M.J. Craner, M.A. Friese, K.B. Jakobsen, J. Newcombe, M.M.
Korn,T., E. Bettelli,W. Gao,A.Awasthi,A. Jger,T.B. Strom, M. Oukka, andV.K. Esiri, and L. Fugger. 2011. IL-21 and IL-21 receptor expression in lym-
Kuchroo. 2007. IL-21 initiates an alternative pathway to induce proinflam- phocytes and neurons in multiple sclerosis brain. Am. J. Pathol. 178:794
matory T(H)17 cells. Nature. 448:484487. http://dx.doi.org/10.1038/ 802. http://dx.doi.org/10.1016/j.ajpath.2010.10.043
nature05970 van Leeuwen, M.A., D.J. Lindenbergh-Kortleve, H.C. Raatgeep, L.F. de
Liesz, A., E. Suri-Payer, C.Veltkamp, H. Doerr, C. Sommer, S. Rivest, T. Giese, Ruiter, R.R. de Krijger, M. Groeneweg, J.C. Escher, and J.N. Samsom.
and R.Veltkamp. 2009. Regulatory T cells are key cerebroprotective im- 2013. Increased production of interleukin-21, but not interleukin-17A,
munomodulators in acute experimental stroke. Nat. Med. 15:192199. in the small intestine characterizes pediatric celiac disease. Mucosal
http://dx.doi.org/10.1038/nm.1927 Immunol. 6:12021213. http://dx.doi.org/10.1038/mi.2013.19
Longa, E.Z., P.R. Weinstein, S. Carlson, and R. Cummins. 1989. Reversible Xie, X.J., Y.F. Ye, L. Zhou, H.Y. Xie, G.P. Jiang, X.W. Feng, Y. He, Q.F. Xie,
middle cerebral artery occlusion without craniectomy in rats. Stroke. and S.S. Zheng. 2010. Th17 promotes acute rejection following liver
20:8491. http://dx.doi.org/10.1161/01.STR.20.1.84 transplantation in rats. J. Zhejiang Univ. Sci. B. 11:819827. http://dx.doi
Magnus, T., H. Wiendl, and C. Kleinschnitz. 2012. Immune mechanisms of .org/10.1631/jzus.B1000030
stroke. Curr. Opin. Neurol. 25:334340. http://dx.doi.org/10.1097/WCO Yilmaz, G., and D.N. Granger. 2010. Leukocyte recruitment and ischemic
.0b013e328352ede6 brain injury. Neuromolecular Med. 12:193204. http://dx.doi.org/10
Majid, A., Y.Y. He, J.M. Gidday, S.S. Kaplan, E.R. Gonzales, T.S. Park, J.D. .1007/s12017-009-8074-1
Fenstermacher, L.Wei, D.W. Choi, and C.Y. Hsu. 2000. Differences in vulner- Yilmaz, G.,T.V.Arumugam, K.Y. Stokes, and D.N. Granger. 2006. Role of T lym-
ability to permanent focal cerebral ischemia among 3 common mouse strains. phocytes and interferon-gamma in ischemic stroke. Circulation. 113:2105
Stroke. 31:27072714. http://dx.doi.org/10.1161/01.STR.31.11.2707 2112. http://dx.doi.org/10.1161/CIRCULATIONAHA.105.593046

604 IL-21 promotes brain injury after stroke | Clarkson et al.


The Pinnacle of Discovery
Recombinant Proteins for Bioassay

BioLegend has a large variety of recombinant cytokines, chemokines, growth factors, and proteinases (e.g., MMPs, Granzymes A
and B). The high quality of our recombinant proteins anchor your research, and the exceptional pricing allows you to focus on
reaching those lofty research goals.

BioLegend Recombinant Proteins are:


Diverse, with over 400 Exceptionally priced, to Functionally tested and Available in carrier-free
human, mouse, and rat meet even the tightest validated for bioassay. and animal-free formats
proteins. budget. to suit your assay needs.

Recombinant Mouse FLT3L Recombinant Human MMP-9


Mouse FLT3L induces The activity of
BioLegend IL-6 production in monomeric hMMP-9
Competitor murine myeloid was measured with
OD (450 - 570 nm)

leukemia cell line M1. 10 M of fluorogenic


MMP substrate, Mca-
RFU/sec

PLGL-Dpa-AR-NH2 , in
the presence of 2.5,
5.0, 10, 20, 40, and
80 ng of activated
hMMP-9. The activity
of activated hMMP-9 is
>800 pmole/min/g.
ng/mL hMMP-9 (ng)

To learn more, visit: biolegend.com/recombinant_proteins


BioLegend is ISO 9001:2008 and ISO 13485:2003 Certified
Toll-Free Tel: (US & Canada): 1.877.BIOLEGEND (246.5343)
Tel: 858.768.5800
biolegend.com
08-0050-01

World-Class Quality | Superior Customer Support | Outstanding Value

Anda mungkin juga menyukai