Anda di halaman 1dari 5

Stem cells and the evolving notion of

cellular identity
rstb.royalsocietypublishing.org
George Q. Daley1,2
1
Stem Cell Transplantation Program, Division of Hematology/Oncology, Howard Hughes Medical Institute,
Boston Childrens Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
2
Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston,
Review MA 02115, USA

Cite this article: Daley GQ. 2015 Stem cells Stem cells are but one class of the myriad types of cells within an organism.
and the evolving notion of cellular identity. With potential to self-renew and capacity to differentiate, stem cells play
Phil. Trans. R. Soc. B 370: 20140376. essential roles at multiple stages of development. In the early embryo, plur-
http://dx.doi.org/10.1098/rstb.2014.0376 ipotent stem cells represent progenitors for all tissues while later in
development, tissue-restricted stem cells give rise to cells with highly
specialized functions. As best understood in the blood, skin and gut, stem
Accepted: 1 June 2015 cells are the seeds that sustain tissue homeostasis and regeneration, while
in other tissues like the muscle, liver, kidney and lung, various stem or pro-
One contribution of 13 to a discussion meeting genitor cells play facultative roles in tissue repair and response to injury.
Here, I will provide a brief perspective on the evolving notion of cellular
issue Cells: from Robert Hooke to cell
identity and how reprogramming and transcription factor-mediated con-
therapya 350 year journey. versions of one cell type into another have fundamentally altered our
assumptions about the stability of cell identity, with profound long-term
Subject Areas: implications for biomedical research and regenerative medicine.
cellular biology, computational biology,
developmental biology, synthetic biology
1. Background and history: from cells to stem cells
Keywords: In his groundbreaking seventeenth century treatise Micrographia: or, Some
embryonic stem cells, reprogramming, induced physiological descriptions of minute bodies made by magnifying glasses
pluripotent stem cells, cellNet (London: J. Martyn and J. Allestry, 1665) Robert Hooke described his visualiza-
tion of the tiny walled units of a thinly sliced film of cork, which he likened to a
honeycomb, and dubbed them cells. Some 350 years hence, cells are well
Author for correspondence:
known as capable of living free as whole organisms or as building blocks of
George Q. Daley living tissues within highly complex beings, and as remarkably malleable
e-mail: george.daley@childrens.harvard.edu and resilient fundamental units of life.
The twentieth century, arguably the century of genetics, began with the elu-
cidation of genes as units of heredity, mid-way through witnessed the discovery
of the double helix, and concluded with the completion of the rough draft of the
human genome sequence. Although the genome sequence provides the genetic
code, the principle research challenge of the twenty-first century is learning
how this code is read and interpreted by cellshow the information in the
single-celled human zygote is translated to produce the trillion-odd specialized
units within the human body. Developmental biology addresses many of the
questions inherent to formulating the body plan, but deciphering the expression
of the genetic code is increasingly the provenance of stem cell biology, which
seeks to understand epigenetic regulation and cell fate. Just as the gene is the
principle sub-unit of the genome, the stem cell is the chief unit of organization
for tissue formation and homeostasis.
The concept of developmental hierarchies of cells, with a stem cell at the top
of this hierarchy and multiple differentiated offspring derived from it, first
emerged in the late nineteenth century and has become an experimental main-
stay of the last 65 years, owing to the pioneering definitions of self-renewing
multipotential progenitors of all lineages of the blood by Till and McCulloch
in a series of pioneering papers [1,2]. In delineating the stem cell for the
blood-forming tissue the haematopoietic stem cell (HSC), Till and McCulloch
set the stage for understanding the developmental hierarchies of additional
tissues and organsthe skin, gut, lung, sperm and othersas well as the

& 2015 The Author(s) Published by the Royal Society. All rights reserved.
emergence of whole organisms from the pluripotent stem cell mass of the blastocyst, and hence is termed naive [6,7], 2
cells of the early embryo. To ascribe their unique proper- in contrast to the cells that more closely resemble the epiblast

rstb.royalsocietypublishing.org
ties, a stem cell must be defined clonallyat the single-cell stage of mammalian development, like human ESCs or
levelfor its ability to self-renew and differentiate, and mouse EpiSCs, which are considered primed, as if poised to
consequently stem cells represent but a tiny fraction of cells differentiate. Indeed, several groups have claimed to have
within the tissues of adult organisms. identified combinations of transgenes or purely chemical
means to convert human primed pluripotent stem cells to
their naive state. If such naive human ESCs can indeed be
introduced as an alternative to classically derived hESCs oper-
2. Developmental potency of stem cells ating since the first lines of Thomson, there may be advantages
Stem cells are defined by their range of potential fatesa con- in terms of growth rates, ease of passage from plate to plate,
cept termed developmental potency. The zygote is the most and the receptivity to efforts at genome editing. Thus, the

Phil. Trans. R. Soc. B 370: 20140376


developmentally expansive cell, yet is rarely considered a stem search for naive hESCs represents a holy grail of stem cell
cell in mammals because it cleaves into blastomeres of equal biology and promises many interesting new insights into the
developmental potency for at most three cell divisions, and metastable or heterogeneous state of pluripotent stem cells [8].
therefore manifests very limited self-renewal potential. The
zygote and early blastomeres form all the tissues of the
embryo proper as well as the supportive extraembryonic
tissuesthe fetal membranes and placentaa unique capacity
3. Multipotential stem cells
called totipotency. To date, no one has succeeded in propagating While ESCs garner considerable interest due to their multi-
the zygote or blastomeres continuously in cell culture. Embryo- faceted developmental plasticity, and may one day prove
nic stem cells (ESCs), by contrast, can be isolated from the inner invaluable as a source for autologous, personalized transplants
cell mass of blastocysts and cultured as immortal cell lines. of cells for tissue repair or regeneration, the best established
When murine ESCs are aggregated with early blastomeres or existing cell therapies typically involve another class of cells
returned to the blastocoele cavity of the blastocyst by microinjec- deemed adult or somatic stem cells. Unlike embryo-derived
tion, they chimaerize all tissues within the embryo proper stem cells, tissue-restricted somatic stem cells are limited in
but typically not the placenta, which reflects their developmen- their potency to the cell types of the tissue in which they
tal segregation from the trophectodermal lineage, which has its reside. HSCs generate blood, gut stem cells the lining of the
own master cellthe trophoblastic stem cell. ESCs are con- intestines and skin stem cells the epithelial lining of the body.
sidered pluripotential. The developmental potency of human Despite earlier claims of greater plasticity, somatic stem cells
ESCs, which for ethical reasons cannot be determined by are highly constrained and do not without considerable genetic
embryo chimaerism, is instead assessed by observing the spec- or chemical manipulation trans-differentiate into foreign cell
trum of tissue differentiation that results after subcutaneous types or tissues, and do not represent repair kits for tissues
injection of cells into immune-compromised mice. Pristine other than their own. The most widely exploited somatic stem
ESCs produce teratomas, encapsulated tumours consisting of cellHSCscan be transplanted into patients with various
disorganized masses of differentiated tissues. Because elements blood diseases, malignant and genetic, after the patients dis-
of the three primitive embryonic germ layersectoderm, meso- eased blood-forming system has been eradicated by high-dose
derm and endodermcoexist in these tumours, human ESCs chemotherapy and irradiation. But blood cells will not readily
share pluripotency with mouse ESCs. regenerate or repair other tissues like the heart, brain, liver or
When human ESCs were first described by Thomson et al. kidney after they have been injured or afflicted by disease. Con-
[3], there was an oddity that went largely unnoticed for almost sequently, multipotential stem cells represent highly valuable
a decade. The human-derived cells grew under distinct con- sources for regenerative therapies within adult tissues, but on
ditions, stimulated by fibroblast growth factor instead of account of their lineage-restricted nature, do not provide a
leukaemia inhibitory factor (LIF), which nurtured the mouse source for all potential tissue therapies in the adult. For tissues
ESCs. Human ESCs appeared not to depend on LIF signalling, that do not readily regenerate from stem cell pools in the
nor to activate the downstream pathways driven by the LIF/ adultlike the heart, kidney, pancreatic beta cells and much
gp130 receptor complex, implying a distinct physiologic or of the brainonly pluripotent stem cells directed to differentiate
developmental state [4]. Moreover, the colonies of human along specific lineages that recapitulate embryonic development
ESCs appeared flatter and were more prone to die when provide hope for future cell replacement therapies.
passaged as single cells than the mouse ESCs, which grew as
domed colonies and could be readily dissociated and passed
as single cells. Only when stem cells from the epiblast stage 4. Reverting differentiated somatic cells to
of mouse development were isolated and cultured did the dis-
tinctions become clear: human ESCs resembled more closely pluripotency
the epiblast-derived stem cells (EpiSCs) of slightly later stage During much of history since the time of Hooke, cell biologists
embryos. Hence, murine EpiSCs appear to be a closer develop- have considered development a one way thoroughfare, with
mental equivalent to human ESCs than murine ESCs [5]. the range of a cells fates becoming progressively restricted
Vigorous recent efforts to isolate human ESCs under con- as the zygote progresses from a totipotent state to the pluripo-
ditions that more closely mimic the mouse ESCs have tent state of the inner cell mass of the pre-implantation
yielded a large number of papers that report slightly different blastocyst, and ultimately devolves into the more limited
conditions and states of a putative naive human stem cell. potential of tissue-restricted cells. Ultimately, most cells in
Such a cell, like the mouse ESC, is closer in its DNA methyl- the adult organism attain a fully differentiated state, which
ation and gene expression patterns to the cells of the inner remains stable and immutable unless affected by pathologic
processes like metaplasia or frank malignancy. While today we the MyoD TF to convert fibroblasts to muscle cells [17,18], 3
comprehend that virtually all cells, save lymphocytes, retain Yamanakas TF-based approach to reverting somatic cells to

rstb.royalsocietypublishing.org
the full complement of DNA as all other cells, and become pluripotency also provoked a number of laboratories to test
specialized by virtue of expressing certain subsets of genes whether TFs might drive conversions of one somatic cell
while silencing others, early thinkers like August Weissman type directly to another, a process called transdifferentiation
speculated that development proceeded by a series of quali- or direct conversion. Among the first laboratories to demon-
tative divisions in which daughter cells differ as each strate this phenomenon was that of Doug Melton, who
inherits a different set of factors that specify function. Later succeeded in converting exocrine pancreatic cells to insulin-
Hans Spemann proved through a series of painstaking and producing beta cells by direct injection of mouse pancreas
ingenious micromanipulations of early newt blastomere with master regulators of beta-cell fate [19]. Wernigs group
nuclei that all had equal potential to generate viable organ- later converted mouse and human fibroblasts to neurons
isms, establishing the paradigm-shifting notion of nuclear using a similar strategy, only that TFs were engineered into

Phil. Trans. R. Soc. B 370: 20140376


equivalence. Spemann envisioned but never succeeded in fibroblasts in vitro [20,21]. Subsequently, a flood of papers
performing his fantastical experimenttransplanting the have reported conversions of one somatic cell type to another
nucleus of a highly differentiated cell back to the egg to test via this approach (reviewed in [22]). Indeed, these exper-
whether it would regain and manifest embryonic potential. iments have further extended the concept that a cells
Reporting success in precisely that fantastical experiment, molecular composition and thus function are malleable and
Briggs and King showed in 1952 that when nuclei were trans- can be altered for applications in research and potentially
ferred from the blastula stage cells of a frog embryo into a fresh for therapeutic purposes. But whether such cells function
egg, a third resulted in swimming tadpoles, showing that later equivalently to their native counterparts has remained a
embryonic stage cells showed nuclear equivalence. Ultimately, lingering question among stem cell biologists.
John Gurdon proved the full developmental potential of
highly differentiated intestinal cells of the frog in generating
functional organisms, thereby unequivocally establishing the 6. CellNet: a metric for discerning cell identity
principle of nuclear equivalence throughout development.
Somatic cell nuclear transfer (SCNT) ultimately succeeded in and enhancing cell engineering
proving the full developmental competency of the nuclear Numerous laboratories within the field of stem cell biology
genome of mammalian cells of the sheep [9], and now well are consumed by attempts to convert various cell types
over a dozen distinct mammalian species. Indeed, SCNT in vitro into distinct altered states: whether directing pluripo-
together with directed differentiation of murine ESCs rep- tent stem cells to differentiate along specific lineages towards
resents a classical paradigm for customized gene and cell specialized terminal cells, directing the conversion of one
therapy to treat genetic disease, as has been shown in somatic cell type to another with enforced expression of
murine models [10]. While nuclear transfer has ultimately suc- TFs, or reverting differentiated somatic cells to pluripotency
ceeded in humans [11], it has proven quite cumbersome, by expression of the Yamanaka factors. Such efforts promise
requiring harvesting of fresh oocytes from women willing to to validate cultures of human cells as improved models of
donate to research. human pathology and physiology, and raise hopes that cell-
The impractical technical and controversial aspects of based target validation, drug-screening and mechanistic
human SCNT were bound to limit its impact. Indeed, in a research will be strengthened. Of some concern however,
major paradigm-shifting demonstration that transcription fac- are the standards by which cells engineered in vitro are com-
tors (TFs) play central roles in specifying cell fates, Yamanaka pared to native tissues for both molecular identity and
and colleagues [12] demonstrated that ectopic expression of physiology. Most claims that cells engineered in a dish have
four TFs normally expressed in ESCs (Oct4, Sox2, KLF4 and achieved a given cell fate equivalent to native tissues are typi-
c-MYC) were sufficient to reset a somatic cells differentiated cally founded on analysis of a small number of diagnostic
state back to pluripotency. Within little more than a year, Yama- markers, on limited functional assays and on global assess-
nakas group as well as Thomsons and my own applied TF ments of gene expression analysed by clustering algorithms
reprogramming successfully to human cells, achieving the deri- that provide at best a qualitative suggestion of similarity
vation of induced pluripotent stem cells (iPSC; [1315]). We without an objective metric of identity.
used the reprogramming technology to assemble the first repo- Given the momentous challenge of assessing the quality
sitory of patient-derived iPSC representing a spectrum of genetic and fidelity of cells engineered in vitro, and the lack of rigor-
disease, including immune-deficiency, Downs syndrome and ous metrics for assessing cell states, my laboratory has
Huntingtons disease, among others [16]. Indeed, Yamanaka- worked closely with that of James Collins (MIT) to generate
style reprogramming has revolutionized the field of stem cell a computational platform that defines how closely an engin-
biology, rendering the notion that differentiated cellular state eered cell approaches the identity of a target cell or tissue
can be efficiently reprogrammed to pluripotency, ushering in a type, and provides a set of candidate regulators of the cells
profound set of possibilities for modelling human disease and transcriptional state that require further modulation to push
developing new drug and cell-based therapies. the engineered cells ever closer to their target. CellNet is a
publicly available network-biology platform (http://cellnet.
hms.harvard.edu/) written by Patrick Cahan with input
5. Diverting somatic lineages directly to from Hu Li and Edroaldo Lummertz de Rocha that has
been tested extensively against both published datasets [23]
alternate cell fates and used within our own laboratory to enhance the in vitro
Inspired in part by the classical experiments of Weintraub, production of several cell types [24]. At its core, CellNet is
Lassar and colleagues which exploited ectopic expression of founded on the notion that cell- and tissue-specific gene
regulatory networks (GRNs) are major molecular determi- enforced expression of Klf4/5 dampened liver marker 4
nants of cell identity that govern both the steady-state expression and fortified intestinal identity. Taken together,

rstb.royalsocietypublishing.org
transcriptional programme as well as cellular responses to these data suggested that the iHeps might in fact be poised
the environment and to various contextual perturbations as bi-potential progenitors of endoderm fates (semi-colon).
like disease and ageing. We reasoned that defining the Remarkably, these semi-colon cells could be coaxed ever
status of GRNs within cells engineered in vitro would provide closer to intestinal fates by engraftment in a murine colitis
a robust metric of cell identity as well as a means of determin- model. When the colon-engrafted cells were recovered and
ing which TFs were most dysregulated, thereby providing a analysed by CellNet, they showed a high degree of colonic epi-
plausible set of candidates to be further manipulated in thelial identity. Our laboratory is further applying CellNet to
future experiments. Using publicly available microarray optimize our efforts to convert pluripotent stem cells along
expression data and a modified version of the Context Like- various blood lineages.
lihood of Relatedness algorithm [25], Cahan produced a CellNet (v. 1.0) has notable limitations. CellNet was

Phil. Trans. R. Soc. B 370: 20140376


global GRN that defines regulatory relationships among all trained on microarray data, which are plentiful enough to pro-
annotated transcriptional regulators and target genes across vide adequate power to discover GRNs for many but by no
most cell types, and then refined these relationships into means all tissues. The current version of CellNet is powered
cell and tissue type-specific GRNs that could be used to to classify a query population of cells against a panel of
build a cell-type classifier. Other metrics included quantitat- some 20 murine or 16 human target tissues. While many
ive assessments of GRN status and a network influence medically relevant tissues currently being investigated by
score, which ranks TFs according to their effect on the GRNs. researchers are included, e.g. neurons, heart, haematopoietic
With CellNet in hand, we evaluated the outcomes of 226 stem/progenitor cells, the spectrum of human cell types is
experimentally derived cell populations drawn from 56 pub- vastly larger, especially when considering that the ideal train-
lished studies, and gleaned several overarching lessons. First, ing data would yield GRNs for every definable cell type at
virtually all reprogrammed cellsi.e. those reverted to plur- every stage of development. Furthermore, the current training
ipotency by virtue of Yamanaka-style reprogramming data derive largely from parenchymal tissues and whole
achieved near identity to the gold standard ESC, whether organs, which encompass many cell types, while most engin-
mouse or human. Second, efforts at directed differentiation eered cells are acclimated to culture in a dish. Despite these
of pluripotent stem cells (either ESCs or iPSCs) using mor- limitations, when primary cells are cultured from whole
phogens and selective culture conditions in vitro achieved organs or tissues, purified, and then subjected to analysis via
on average higher classification scores relative to their CellNet, the de-differentiation effects of cell culture and the
target tissues than experiments that endeavour to directly heterogeneity of the target tissue does not compromise the uti-
convert one differentiated somatic cell type directly into lity of CellNet as a robust classifier of a cells identity [23].
another via ectopic expression of master regulatory TFs CellNet will be augmented in future versions as transcriptional
[23]. These analyses indicate that reprogramming is a remark- profiles become available for additional target cells and
ably robust transition in cell fates, perhaps in part due to the tissues at multiple stages of development. Moreover, as more
highly selective culture conditions established for pluripotent researchers turn to RNA sequencing as a mode of transcrip-
cell types. However, when making neurons or cardiomyo- tional profiling, CellNet will need to be adapted to accept
cytes or hepatocytes from pluripotent cells, we have much these data (and such software refinement is underway). At
to learn before we can claim great success in deriving the theoretical limit, as RNA sequencing of single cells
highly specialized cell and tissue types. Even more so, our becomes more robust, diagnostic GRNs might be discoverable
attempts to convert differentiated fibroblasts directly into for every cell type at every stage of organismal development,
neurons or cardiomyocytes or hepatocytes without first enabling algorithms like CellNet to define the transcriptional
returning to the highly plastic state of pluripotency are lim- roadmap for human development. Such a platform would
ited by epigenetic modifications that stabilize the fibroblast greatly facilitate efforts at directed differentiation of cell
identity of the starting cells. Interestingly, a combination of types in vitro and accelerate the capacity for stem cells to be
transient expression of Yamanaka factors with ectopic translated for research and medical applications.
expression of lineage specifying factorsa strategy some-
times referred to as primed conversion [26], appears in
some cases highly effective at converting one somatic cell
type directly into another, and bears further exploration. 7. Prospects for the future
In an effort anchored by Samantha Morris in my labora- The definition of the cell has been refined at ever greater resol-
tory [24], we employed CellNets predictive features to refine ution since its first description by Hooke 350 years ago. In
the conversion of B cells into macrophages by ectopic recent years, the capacity to reprogram and redirect cells such
expression of CEBP/a [27,28] and fibroblasts into hepatocytes that they morph from one identity to another has highlighted
by enforced expression of Hnf4a and FoxA TFs [29,30]. In the highly plastic and malleable nature of the genome and the
converted macrophages, CellNet identified the persistence fluid state of cells. Indeed, cells have assumed a chameleon-
of B-cell-associated TFs EBF1 and Pou2af1/OBF1, and their like character. Understanding the precise molecular mechan-
knock-down enhanced macrophage functionality in vitro. In isms that define the formation, stabilization and transitions
the hepatocyte-like cells induced from fibroblasts (iHeps), Cell- among GRNs promises to inform future efforts to engineer
Net detected aberrant activation of cdx2, a hindgut-associated cells more faithfully in vitro and to diagnose and treat cellular
transcriptional regulator, as well as low-level expression of the pathology and promote repair and regeneration in vivo. More-
intestinal regulators Klf4/5. When cdx2 was knocked down, over, understanding GRNs as modules of cellular function
the putative iHeps became more robust in their liver-like prop- offers the possibility that new types of hybrid cells with highly
erties of albumin synthesis and urea metabolism, whereas engineered functions might be produced for biomedical
applications, the stuff of synthetic biology. Why be constrained biotechnology companies: True North, Solasia, Epizyme, Verastem, 5
to making a perfect hepatocyte or struggle to make a beta cell Ocata, Raze and MPM Capital. The author is an affiliate member of
the Broad Institute, and an investigator of the Howard Hughes Medical

rstb.royalsocietypublishing.org
when there might be advantages to engineering a hepatocyte
Institute and the Manton Center for Orphan Disease Research.
to perform glucose-responsive insulin secretion? It is provoca-
Funding. The author is supported by grants from the US National Insti-
tive to consider what the cell will look like in another 350 years. tutes of Health: R24-DK092760, RC4-DK090913, and UO1-HL100001
Progenitor Cell Biology Consortium; Alexs Lemonade Stand; Doris
Competing interests. The author is a member of the scientific advisory
Duke Medical Foundation and Ellison Medical Foundation.
boards and receives consulting fees or holds equity in the following

References

Phil. Trans. R. Soc. B 370: 20140376


1. McCulloch EA, Till JE. 1960 The radiation sensitivity 11. Tachibana M et al. 2013 Human embryonic stem 22. Morris SA, Daley GQ. 2013 A blueprint for
of normal mouse bone marrow cells, determined by cells derived by somatic cell nuclear transfer. Cell engineering cell fate: current technologies to
quantitative marrow transplantation into irradiated 153, 12281238. (doi:10.1016/j.cell.2013.05.006) reprogram cell identity. Cell Res. 23, 33 48.
mice. Radiat. Res. 13, 115125. (doi:10.2307/ 12. Takahashi K, Yamanaka S. 2006 Induction of (doi:10.1038/cr.2013.1)
3570877) pluripotent stem cells from mouse embryonic and 23. Cahan P, Li H, Morris SA, Lummertz da Rocha E,
2. Till JE, McCulloch EA. 1961 A direct measurement of the adult fibroblast cultures by defined factors. Cell 126, Daley GQ, Collins JJ. 2014 CellNet: network biology
radiation sensitivity of normal mouse bone marrow cells. 663 676. (doi:10.1016/j.cell.2006.07.024) applied to stem cell engineering. Cell 158,
Radiat. Res. 14, 213222. (doi:10.2307/3570892) 13. Yu J et al. 2007 Induced pluripotent stem cell lines 903915. (doi:10.1016/j.cell.2014.07.020)
3. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz derived from human somatic cells. Science 318, 24. Morris SA, Cahan P, Li H, Zhao AM, San Roman AK,
MA, Swiergiel JJ, Marshall VS, Jones JM. 1998 1917 1920. (doi:10.1126/science.1151526) Shivdasani RA, Collins JJ, Daley GQ. 2014 Dissecting
Embryonic stem cell lines derived from human 14. Park IH, Zhao R, West JA, Yabuuchi A, Huo H, Ince engineered cell types and enhancing cell fate
blastocysts. Science 282, 1145 1147. (doi:10.1126/ TA, Lerou PH, Lensch MW, Daley GQ. 2008 conversion via CellNet. Cell 158, 889 902. (doi:10.
science.282.5391.1145) Reprogramming of human somatic cells to 1016/j.cell.2014.07.021)
4. Daheron L, Opitz SL, Zaehres H, Lensch MW, pluripotency with defined factors. Nature 451, 25. Faith JJ, Hayete B, Thaden JT, Mogno I, Wierzbowski
Andrews PW, Itskovitz-Eldor J, Daley GQ. 2004 LIF/ 141 146. (doi:10.1038/nature06534) J, Cottarel G, Kasif S, Collins JJ, Gardner TS. 2007
STAT3 signaling fails to maintain self-renewal of 15. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka Large-scale mapping and validation of Escherichia
human embryonic stem cells. Stem Cells 22, T, Tomoda K, Yamanaka S. 2007 Induction of coli transcriptional regulation from a compendium
770778. (doi:10.1634/stemcells.22-5-770) pluripotent stem cells from adult human fibroblasts of expression profiles. PLoS Biol. 5, e8. (doi:10.1371/
5. De Los Angeles A, Loh YH, Tesar PJ, Daley GQ. 2012 by defined factors. Cell 131, 861872. (doi:10. journal.pbio.0050008)
Accessing naive human pluripotency. Curr. Opin. 1016/j.cell.2007.11.019) 26. Zhang Y, Li W, Laurent T, Ding S. 2012 Small
Genet. Dev. 22, 272 282. (doi:10.1016/j.gde.2012. 16. Park IH et al. 2008 Disease-specific induced molecules, big rolesthe chemical manipulation of
03.001) pluripotent stem cells. Cell 134, 877 886. (doi:10. stem cell fate and somatic cell reprogramming.
6. Kalkan T, Smith A. 2014 Mapping the route from 1016/j.cell.2008.07.041) J. Cell Sci. 125, 56095620. (doi:10.1242/jcs.
naive pluripotency to lineage specification. Phil. 17. Davis RL, Weintraub H, Lassar AB. 1987 Expression 096032)
Trans. R. Soc. B 369, 20130540. (doi:10.1098/rstb. of a single transfected cDNA converts fibroblasts to 27. Bussmann LH et al. 2009 A robust and highly
2013.0540) myoblasts. Cell 51, 987 1000. (doi:10.1016/0092- efficient immune cell reprogramming system.
7. Marks H et al. 2012 The transcriptional and 8674(87)90585-X) Cell Stem Cell 5, 554 566. (doi:10.1016/j.stem.
epigenomic foundations of ground state 18. Lassar AB, Paterson BM, Weintraub H. 1986 2009.10.004)
pluripotency. Cell 149, 590 604. (doi:10.1016/j. Transfection of a DNA locus that mediates the 28. Di Tullio A, Vu Manh TP, Schubert A, Castellano G,
cell.2012.03.026) conversion of 10T1/2 fibroblasts to myoblasts. Cell Mansson R, Graf T. 2011 CCAAT/enhancer binding
8. Cahan P, Daley GQ. 2013 Origins and implications of 47, 649656. (doi:10.1016/0092-8674(86)90507-6) protein alpha (C/EBPa)-induced transdifferentiation
pluripotent stem cell variability and heterogeneity. 19. Zhou Q, Brown J, Kanarek A, Rajagopal J, Melton of pre-B cells into macrophages involves no overt
Nat. Rev. Mol. Cell Biol. 14, 357 368. (doi:10.1038/ DA. 2008 In vivo reprogramming of adult pancreatic retrodifferentiation. Proc. Natl Acad. Sci. USA 108,
nrm3584) exocrine cells to beta-cells. Nature 455, 627 632. 17 016 17 021. (doi:10.1073/pnas.1112169108)
9. Wilmut I, Schnieke AE, McWhir J, Kind AJ, Campbell (doi:10.1038/nature07314) 29. Huang P, He Z, Ji S, Sun H, Xiang D, Liu C, Hu Y,
KH. 1997 Viable offspring derived from fetal and 20. Pang ZP et al. 2011 Induction of human neuronal Wang X, Hui L. 2011 Induction of functional
adult mammalian cells. Nature 385, 810 813. cells by defined transcription factors. Nature 476, hepatocyte-like cells from mouse fibroblasts by
(doi:10.1038/385810a0) 220 223. (doi:10.1038/nature10202) defined factors. Nature 475, 386389. (doi:10.
10. Rideout WMIII, Hochedlinger K, Kyba M, Daley GQ, 21. Vierbuchen T, Ostermeier A, Pang ZP, Kokubu Y, 1038/nature10116)
Jaenisch R. 2002 Correction of a genetic defect by Sudhof TC, Wernig M. 2010 Direct conversion of 30. Sekiya S, Suzuki A. 2011 Direct conversion of mouse
nuclear transplantation and combined cell and gene fibroblasts to functional neurons by defined factors. fibroblasts to hepatocyte-like cells by defined
therapy. Cell 109, 17 27. (doi:10.1016/S0092- Nature 463, 10351041. (doi:10.1038/ factors. Nature 475, 390 393. (doi:10.1038/
8674(02)00681-5) nature08797) nature10263)

Anda mungkin juga menyukai