Anda di halaman 1dari 5

Neuroscience Research 126 (2018) 39–43

Contents lists available at ScienceDirect

Neuroscience Research
journal homepage: www.elsevier.com/locate/neures

Review article

Astrocyte reactivity and astrogliosis after spinal cord injury


Seiji Okada a,b,∗ , Masamitsu Hara a,b , Kazu Kobayakawa b , Yoshihiro Matsumoto b ,
Yasuharu Nakashima b
a
Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
b
Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan

a r t i c l e i n f o a b s t r a c t

Article history: After traumatic injuries of the central nervous system (CNS), including spinal cord injury (SCI), astrocytes
Received 25 August 2017 surrounding the lesion become reactive and typically undergo hypertrophy and process extension. These
Received in revised form 9 September 2017 reactive astrocytes migrate centripetally to the lesion epicenter and aid in the tissue repair process,
Accepted 9 September 2017
however, they eventually become scar-forming astrocytes and form a glial scar which produces axonal
Available online 17 October 2017
growth inhibitors and prevents axonal regeneration. This sequential phenotypic change has long been
considered to be unidirectional and irreversible; thus glial scarring is one of the main causes of the limited
Keywords:
regenerative capability of the CNS. We recently demonstrated that the process of glial scar formation is
Spinal cord injury
Astrocytes
regulated by environmental cues, such as fibrotic extracellular matrix material. In this review, we discuss
Glial scar the role and mechanism underlying glial scar formation after SCI as well as plasticity of astrogliosis, which
Axonal regeneration helps to foster axonal regeneration and functional recovery after CNS injury.
Laser microdissection © 2017 Elsevier Ireland Ltd and Japan Neuroscience Society. All rights reserved.

Contents

1. Spinal cord injury and glial scar formation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39


2. The beneficial role of reactive astrocytes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
3. The plasticity of astrocytic changes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
4. Preventing glial scar formation through environmental regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41
5. Cell transplantation therapy and glial scar for SCI . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42
6. Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43
Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43

in the USA; approximately 100,000 people in Japan and 1.2 million


1. Spinal cord injury and glial scar formation people in the USA are paralyzed due to SCI, mainly because of motor
vehicle, sports, and work accidents (Lee-Liu et al., 2013; Ide-Okochi
There are approximately 5000 new cases of traumatic spinal et al., 2013). The incidence of fall-related injuries in the elderly pop-
cord injury (SCI) recorded each year in Japan, and 17,000 new cases ulation has significantly increased with the aging of the population
(Okada et al., 2009). Traumatic SCI often causes irreversible motor
and sensory dysfunction as well as the loss of bladder, bowel and
sexual function, resulting in a significant reduction in the quality
Abbreviations: CNS, central nervous system; SCI, spinal cord injury; USA, United
States of America; GFAP, glial fibrillary acidic protein; STAT, signal transducer
of life (Adams and Hicks, 2005). Since there are no approved ther-
and activator of transcription; SOCS, Suppressor of cytokine signaling; LMD, laser apies to restore the spinal cord function, a better understanding of
microdissection; GFP, green fluorescent protein; FACS, fluorescence activated cell the mechanism for compromising functional recovery after SCI is
sorting; Col I, the type 1 collagen; NAs, naïve astrocytes; RAs, reactive astrocytes; significantly important.
SAs, scar-forming astrocytes.
∗ Corresponding author at: Department of Orthopaedic Surgery, Graduate School
Glial scarring, which impedes axonal regeneration and func-
of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-
tional recovery has been considered to be the main cause of the
8582, Japan. limited regenerative capability in the mammalian CNS (Silver and
E-mail address: seokada@ortho.med.kyushu-u.ac.jp (S. Okada). Miller, 2004). The impeded axonal regeneration and functional

https://doi.org/10.1016/j.neures.2017.10.004
0168-0102/© 2017 Elsevier Ireland Ltd and Japan Neuroscience Society. All rights reserved.
40 S. Okada et al. / Neuroscience Research 126 (2018) 39–43

after injury. During this functional recovery phase, we found that


reactive astrocytes emerged and eventually migrated centripetally
to the area of the lesion and compacted the infiltrating inflamma-
tory cells, resulting in the compaction of the area of the lesion.
After the migration of reactive astrocytes and the completion of
glial scar formation, the functional improvement also reached a
plateau. Although reactive astrocytes were considered to be harm-
Fig. 1. The changes of astrocytes after CNS injury. Naïve astrocytes are activated by ful for the pathophysiology of SCI due to their contribution to glial
several types of injury (i.e., inflammation, infection, ischemia, and traumatic injury) scar formation, this phenomenon indicates that the emergence
and they exhibit sequential phenotypic changes, including cellular hypertrophy and and migration of reactive astrocytes had a prominent role in the
process extension; these are referred to as reactive astrocytes. Around the area of
repair of injured tissue and the restoration of the motor function
the lesion, reactive astrocytes express cell adhesion molecules, such as N-cadherin,
and transform into scar-forming astrocytes. Scar-forming astrocytes adhere to each
before the completion of glial scar formation (Okada et al., 2006). In
other and form the glial scar. fact, selective ablation of dividing astrocytes using ganciclovir after
SCI in the GFAP-TK transgenic mice resulted in severe leukocyte
infiltration, tissue disruption, demyelination and neuronal death
recovery have been shown to be irreversible and to permanently (Faulkner et al., 2004), which also suggested the beneficial role
inhibit axonal regrowth in both rodents and humans with SCI (Lee- of reactive astrocytes after SCI. Since we found that phosphory-
Liu et al., 2013). In fact, in some non-mammalian vertebrate animals lation and nuclear translocation of signal transducer and activator
(i.e., urodele amphibians and lampreys) in which little glial scar of transcription-3 (STAT3) were mainly observed in reactive astro-
formation is detected, the spinal cord has been shown to regener- cytes, we examined the role of STAT3 in reactive astrocytes in the
ate, substantially restoring the motor function (Lee-Liu et al., 2013; mouse model of SCI (Okada et al., 2006). In the genetically modified
Diaz Quiroz and Echeverri, 2013). Chondroitin and keratin sulphate mice in which STAT3 was selectively depleted in reactive astro-
proteoglycans are among the main inhibitory extracellular matrix cytes under the control of Nestin gene promoter/enhancer, reactive
molecules to be produced by reactive astrocytes in glial scars, and astrocytes showed limited migration to the lesion epicenter, which
they play a crucial part in regeneration failure (Silver, 2016). Due resulted in a wide area of injury, impaired contraction of infiltrating
to the presence of these inhibitory molecules, severed axons can- inflammatory cells, and limited functional recovery (Fig. 2). Sim-
not regenerate past the lesion. In fact, after SCI, treatment with ilar results were observed after SCI in the STAT3-depleted mice
chondroitinase results in the degradation of chondroitin sulphate in GFAP-expressing astrocytes, indicating that STAT3 is a critical
proteoglycans at the lesion site and allows axonal regeneration and regulator of astrogliosis (Herrmann et al., 2008). In contrast, in
functional recovery (Bradbury et al., 2002). the genetically modified mice in which the protein suppressor of
Glial scar formation after CNS insult is regulated by complex and cytokine signaling-3 (SOCS3, negative feedback molecule of STAT3)
combinatorial inter- and intracellular signal mechanisms. Under was selectively depleted in reactive astrocytes, the rapid migration
normal conditions, astrocytes are the predominant subtype of glial of reactive astrocytes to secluded inflammatory cells, enhanced
cells in the CNS; they maintain neurons as well as the blood-brain- contraction of the area of the lesion, and a dramatic improve-
barrier. They are activated in response to various types of injury, ment in functional recovery were observed after SCI. Similarly, in
including inflammation, infection, ischemia, and traumatic injury, transgenic mice that showed the expression of astrocyte-targeted
and play a crucial role in the pathophysiology of each through interleukin-6 (IL-6, one of the upstream effectors of STAT3 path-
a phenotypic change known as reactive astrogliosis. After SCI, way) the prompt migration of reactive astrocytes and compaction
naïve astrocytes sequentially exhibit contrasting phenotypes, first of infiltrating inflammatory cells were observed after CNS injury
as reactive astrocytes and then as scar-forming astrocytes (Fig. 1). (Penkowa et al., 2003). These result suggest that STAT3 signaling,
Although the mechanism underlying reactive response of naïve which is associated with the migration of reactive astrocytes, is a
astrocytes has not been fully elucidated, Shinozaki et al., recently key regulator in the healing process after SCI. In contrast, in the
have demonstrated that microglia transformed astrocytes pheno- chronic phase of injury, reactive astrocytes form glial scars, which
type via downregulation of the P2Y1 purinergic receptor (Shinozaki function as a physical and chemical barrier, impairing axonal regen-
et al., 2017). Reactive astrocytes are necessary in the acute wound eration.
healing and tissue remodeling processes; however, they eventu-
ally become scar-forming astrocytes and form a dense glial scar.
Although these different astrocytic functions are multifaceted and 3. The plasticity of astrocytic changes
phase-dependent, the term of astrogliosis has only been roughly
defined, which makes the role of astrocytes unclear in the patho- Although astrocytic scars have been studied for more than half
physiology of SCI. a century, the cellular and molecular mechanisms of this pro-
cess remain unclear. One factor that limits basic research is the
2. The beneficial role of reactive astrocytes absence of specific markers of differentiation and reactivity. For
example, the characterization of astrocytes is usually performed
Although limited functional recovery occurs during the chronic using intermediate filament GFAP; however, all phenotypes of
phase of SCI, temporary functional recovery is often observed soon astrocytes including reactive astrocytes and scar-forming astro-
after incomplete SCI (Kobayakawa et al., 2014). The mechanism cytes are strongly expressed with GFAP, even though these cell
underlying this phenomenon has been unclear. We previously types show distinct morphological characteristics. These cells are
demonstrated that the astrocytic responses are crucial to tissue conventionally distinguished from each other based on a histo-
repair and functional recovery after SCI (Okada et al., 2006). We logical analysis. Reactive astrocytes are characterized by cellular
first created a contusive SCI (70 Kdyn) on the mouse 9th tho- hypertrophy, process extension, and the increased expression of
racic spinal cord using a commercially available SCI device (Infinite intermediate filaments such as GFAP and Nestin (Frisén et al.,
Horizons Impactor, Precision Systems Instrumentation, Lexington, 1995). However, this immunohistological discrimination is neither
KY), performed an immunohistological analysis, and evaluated the objective nor quantitative. To investigate the regulatory mecha-
functional recovery. In this model of incomplete SCI, there was a nism underlying astrogliosis, clear definitions for naïve, reactive,
substantial recovery in lower extremity movement until 2–3 weeks and scar-forming astrocytes are necessary; we therefore developed
S. Okada et al. / Neuroscience Research 126 (2018) 39–43 41

Fig. 2. The beneficial role of reactive astrocytes in SCI. The migration of reactive astrocytes is important for tissue repair and functional recovery. The impaired migration of
reactive astrocytes results in a wide lesion and poor functional recovery.

a method for distinguishing these cells based on the expression promotor using FACS from 7 days after spinal cord injury, and
of marker genes by selectively isolating these cells with laser confirmed that the reactive astrocyte marker genes were upreg-
microdissection (LMD) (Hara et al., 2017). Naïve astrocytes were ulated in these isolated cells. We then grafted these GFP-positive
isolated from the intact spinal cord, while reactive astrocytes from reactive astrocytes into a new cohort of mice in which the spinal
the spinal cord of B6 mice in the subacute phase of injury (7 days cord was either intact or injured. Notably, the engrafted reac-
after a contusive injury [70 Kdyn]), and scar-forming astrocytes tive astrocytes formed astrocytic scars when transplanted into the
were isolated from the chronic phase of injury (14 days after the injured spinal cord, whereas they reverted (morphologically) back
same SCI). This method revealed that in reactive astrocytes, Nes, to naïve astrocytes when transplanted into the naïve spinal cord
Ctnnb1, Axin2, Plaur, Mmp2, Mmp13 were selectively upregulated (Fig. 3). Consistent with the morphological conversion, a selective
in comparison to naïve and scar-forming astrocytes. These genes gene expression analysis of the engrafted cells using LMD demon-
were therefore defined as the marker genes of reactive astrocytes. strated that scar-forming astrocyte marker genes were significantly
Similarly, Cdh2, Sox9, Xylt1, Chst11, Csgalnact1, Acan, Pcan, Slit2 were upregulated at 7 days after transplantation when the cells were
defined as the marker genes of scar-forming astrocytes. The com- transplanted into the injured spinal cord. In contrast, the original
bination of the expression profile of these marker genes and the expression patterns of reactive astrocyte marker genes were sig-
morphological definitions is useful for identifying the phenotype nificantly suppressed when they were transplanted into the naïve
of astrocytes. Using these markers, we examined whether glial scar spinal cord. In addition, a whole-transcriptome analysis of isolated
formation represented a cell-autonomous phenotypic change or a reactive astrocytes, reactive astrocytes transplanted into the naïve
non-cell-autonomous change (Hara et al., 2017). spinal cord (at 7 days after transplantation), and host astrocytes
First, we harvested the green fluorescent protein (GFP)-labeled in the naïve spinal cord, revealed that the global gene expression
naïve astrocytes from primary astrocyte cultures of the green profiles of transplanted and host astrocytes were comparable; how-
mice where GFP is ubiquitously expressed and then transplanted ever, they were markedly different from the global gene expression
them into the spinal cords of either naïve mice or mice with profiles of isolated reactive astrocytes. These results clearly indi-
SCI, immediately after spinal contusion. At 7 and 14 days after cated that the process of reactive astrogliosis is reversible under
transplantation, we examined the morphological and immuno- certain conditions (Hara et al., 2017).
histochemical characteristics of grafted cells as well as the gene
expression by cell-selective isolation with LMD. The morphological
and gene expression characteristics of the grafted cells were both 4. Preventing glial scar formation through environmental
unchanged when they were grafted into the naïve spinal cord. In regulation
contrast, the grafted cells transformed along with host astrocytes
during various processes, such as process extension and cellular As described above, reactive astrocytes have beneficial func-
hypertrophy, and the reactive astrocyte marker genes were sig- tions, while scar-forming astrocytes are harmful to the repair
nificantly upregulated in the injured spinal cord at 7 days after process after SCI (Okada et al., 2006). Thus, preventing the transfor-
transplantation; they gradually overlapped and were integrated mation of reactive astrocytes into scar-forming astrocytes through
into astrocytic scars and scar-forming astrocyte marker genes were environmental modification may be a new therapeutic strategy for
significantly upregulated at 14 days after transplantation. These SCI. To detect the environmental cues associated with the trans-
findings suggest that astrocytes change their phenotype in an formation of reactive astrocytes into scar-forming astrocytes, we
environment-dependent manner. Next, we examined the plastic- performed time-course genome-wide expression analyses in naïve
ity of the sequential astrocytic changes and whether the reactive and injured spinal cord models. These analyses revealed that in
astrocytes could revert back to naïve astrocytes after transplan- comparison to the naïve spinal cord, the type 1 collagen (Col I)
tation into the naive spinal cord. We selectively isolated reactive genes were the more highly expressed in the injured spinal cord
astrocytes that were genetically-labeled with GFP under the Nes during the scar forming phase (Hara et al., 2017). We performed an
immunohistochemical analysis to investigate the influence of Col
42 S. Okada et al. / Neuroscience Research 126 (2018) 39–43

Fig. 3. The plasticity of astrocytic change. The engrafted reactive astrocytes formed astrocytic scars when transplanted into the injured spinal cord, whereas they reverted
to naïve astrocytes when transplanted into the naïve spinal cord, indicating that the process of reactive astrogliosis occurs in an environment-dependent manner and has
plasticity.

Fig. 4. Preventing glial scar formation through environmental regulation. The administration of anti-␤1 Ab during the scarring phase inhibited the interaction between Col
I and RAs as well as the transformation of RAs to SAs, which resulted in loosened scar formation. Axonal regeneration at the glial scar was impeded in the control group
whereas axonal regeneration into and beyond the epicenter of the lesion was observed in the treatment group.

I on the fate of reactive astrocytes in vitro; the analysis revealed and functional recovery after SCI, providing a potential therapeutic
that astrocytic scars were formed by scar-forming astrocytes colo- target for CNS injury (Hara et al., 2017).
calised with the expression of Col I, while the reactive astrocytes
retained their phenotypes in the Col I-absent area. Reactive astro- 5. Cell transplantation therapy and glial scar for SCI
cytes showed process retraction and the decreased expression of
GFAP when cultured in control dishes. In contrast, they adhered In recent years, cell transplantation has emerged as a potential
closely to adjacent cells and showed the increased expression of therapeutic strategy for SCI and several early-phase clinical trials
GFAP and N-cadherin (characteristic of scar-forming astrocytes), have been completed. Their results showed that cell transplan-
when cultured on Col I-coated dishes. Interfering in the interaction tation is generally feasible; however, the efficacy and long-term
between reactive astrocytes and Col I using antibodies to collagen- safety remain unproven (Assinck et al., 2017). Notably, most
binding integrin (anti-␤1 Ab) as well as N-cadherin-neutralizing preclinical and experimental transplantation studies have been
antibodies, inhibited the transformation of reactive astrocytes into performed in the acute or subacute phase of SCI; few studies
scar-forming astrocytes, even when they were cultured on Col I- have investigated the initiation of treatment in the chronic phase.
coated dishes. This suggests that Col I induces the transformation In addition, transplantation was reported to be effective in the
of reactive astrocytes into scar-forming astrocytes via the integrin- acute or subacute phase of SCI in experimental models. Moreover,
N-cadherin pathway. Similarly, the administration of anti-␤1 Ab few studies have shown its efficacy in chronic SCI—even in ani-
during the scarring phase after SCI loosened the scar formation and mal experimental models. In fact, Fehlings’ group reported that
allowed for significant axonal regeneration into and beyond the the transplantation of neural precursor cells improved the func-
epicenter of the lesion, which resulted in better functional recovery tional recovery in the subacute phase of SCI, but not in the chronic
in comparison to the control SCI mice (Fig. 4). These results indicate phase (Karimi-Abdolrezaee et al., 2006). We also have reported that
that environmental modification in the injured spinal cord pre- chronic transplantation of neural precursor cells failed to improve
vents astrocytic scar formation and promotes axonal regeneration functional recovery although their engraftment, neuronal differen-
S. Okada et al. / Neuroscience Research 126 (2018) 39–43 43

tiation potential, trophic factor secretion were comparable to those Bradbury, E.J., Moon, L.D., Popat, R.J., King, V.R., Bennett, G.S., Patel, P.N., Fawcett,
of the acute transplantation (Kumamaru et al., 2013). Considering J.W., McMahon, S.B., 2002. Chondroitinase ABC promotes functional recovery
after spinal cord injury. Nature 416, 636–640.
that the engrafted neural precursor cells promote functional recov- Diaz Quiroz, J.F., Echeverri, K., 2013. Spinal cord regeneration: where fish, frogs and
ery through synapse reorganization with spared host neurons after salamanders lead the way, can we follow? Biochem. J. 451, 353–364.
SCI (Yokota et al., 2015), the failure of the chronic transplantation Faulkner, J.R., Herrmann, J.E., Woo, M.J., Tansey, K.E., Doan, N.B., Sofroniew, M.V.,
2004. Reactive astrocytes protect tissue and preserve function after spinal cord
can likely be ascribed to the glial scar, which strictly segregates the injury. J. Neurosci. 24, 2143–2155.
lesion and the spared area after SCI. Since the demand for therapies Frisén, J., Johansson, C.B., Török, C., Risling, M., Lendahl, U., 1995. Rapid, widespread,
that are effective for chronic SCI is much greater than that for acute and longlasting induction of nestin contributes to the generation of glial scar
tissue after CNS injury. J. Cell Biol. 131, 453–464.
SCI, a combined approach of cell transplantation therapy with the
Hara, M., Kobayakawa, K., Ohkawa, Y., Kumamaru, H., Yokota, K., Saito, T., Kijima, K.,
attenuation of glial scarring is needed. Yoshizaki, S., Harimaya, K., Nakashima, Y., Okada, S., 2017. Interaction of reactive
astrocytes with type I collagen induces astrocytic scar formation through the
integrin-N-cadherin pathway after spinal cord injury. Nat. Med. 23, 818–828.
6. Concluding remarks
Herrmann, J.E., Imura, T., Song, B., Qi, J., Ao, Y., Nguyen, T.K., Korsak, R.A., Takeda, K.,
Akira, S., Sofroniew, M.V., 2008. STAT3 is a critical regulator of astrogliosis and
In this review, we concisely summarized the pathophysiology as scar formation after spinal cord injury. J. Neurosci. 28, 7231–7243.
Ide-Okochi, A., Yamazaki, Y., Tadaka, E., Fujimura, K., Kusunaga, T., 2013. Illness
well as the reversibility of astrogliosis after SCI with an emphasis on
experience of adults with cervical spinal cord injury in Japan: a qualitative
the selective analysis of each astrocyte phenotype. The regulation of investigation. BMC Public Health 13, 69.
astrogliosis and the transition of naïve, reactive, and scar-forming Karimi-Abdolrezaee, S., Eftekharpour, E., Wang, J., Morshead, C.M., Fehlings, M.G.,
astrocytes could be a feasible and novel therapeutic strategy for SCI. 2006. Delayed transplantation of adult neural precursor cells promotes remyeli-
nation and functional neurological recovery after spinal cord injury. J. Neurosci.
However, a therapeutic approach for chronic SCI, that either atten- 26, 3377–3389.
uates or eliminates a glial scar after it has already formed is most Kobayakawa, K., Kumamaru, H., Saiwai, H., Kubota, K., Ohkawa, Y., Kishimoto, J.,
needed. In addition, the precise mechanism of functional recovery Yokota, K., Ideta, R., Shiba, K., Tozaki-Saitoh, H., Inoue, K., Iwamoto, Y., Okada, S.,
2014. Acute hyperglycemia impairs functional improvement after spinal cord
by promoting the migration of reactive astrocytes and attenuat- injury in mice and humans. Sci. Transl. Med. 6, 256ra137.
ing glial scar formation remains elusive, since the reorganization Kumamaru, H., Saiwai, H., Kubota, K., Kobayakawa, K., Yokota, K., Ohkawa, Y.,
of interactions between descending inputs from the brain to the Shiba, K., Iwamoto, Y., Okada, S., 2013. Therapeutic activities of engrafted neu-
ral stem/precursor cells are not dormant in the chronically injured spinal cord.
spinal cord neurons controlling paroxysmal muscles is required. Stem Cells 31, 1535–1547.
Further elucidation of the astrocytic response during neuronal cir- Lee-Liu, D., Edwards-Faret, G., Tapia, V.S., Larraín, J., 2013. Spinal cord regeneration:
cuit restoration will provide a better understanding of the glial lessons for mammals from non-mammalian vertebrates. Genesis 51, 529–544.
Okada, S., Nakamura, M., Katoh, H., Miyao, T., Shimazaki, T., Ishii, K., Yamane, J.,
biology and a novel therapeutic strategy for CNS injury.
Yoshimura, A., Iwamoto, Y., Toyama, Y., Okano, H., 2006. Conditional ablation
of Stat3 or Socs3 discloses a dual role for reactive astrocytes after spinal cord
Conflict of interest injury. Nat. Med. 12, 829–834.
Okada, S., Maeda, T., Ohkawa, Y., Harimaya, K., Saiwai, H., Kumamaru, H., Matsumoto,
Y., Doi, T., Ueta, T., Shiba, K., Iwamoto, Y., 2009. Does ossification of the posterior
The authors declare no conflicts of interest. longitudinal ligament affect the neurological outcome after traumatic cervical
cord injury? Spine 34, 1148–1152.
Penkowa, M., Giralt, M., Lago, N., Camats, J., Carrasco, J., Hernández, J., Molinero, A.,
Acknowledgments
Campbell, I.L., Hidalgo, J., 2003. Astrocyte-targeted expression of IL-6 protects
the CNS against a focal brain injury. Exp. Neurol. 181, 130–148.
This study was supported in part by a Grants-in-Aid for Scien- Shinozaki, Y., Shibata, K., Yoshida, K., Shigetomi, E., Gachet, C., Ikenaka, K., Tanaka,
K.F., Koizumi, S., 2017. Transformation of astrocytes to a neuroprotective
tific Research (16H05450) and Challenging Exploratory Research
phenotype by microglia via P2Y1 receptor downregulation. Cell Reports 19,
(16K15668) from the Ministry of Education, Science, Sports and 1151–1164.
Culture of Japan. Silver, J., Miller, J.H., 2004. Regeneration beyond the glial scar. Nat. Rev. Neurosci. 5,
146–156.
Silver, J., 2016. The glial scar is more than just astrocytes. Exp. Neurol. 286, 147–149.
References Yokota, K., Kobayakawa, K., Kubota, K., Miyawaki, A., Okano, H., Ohkawa, Y., Iwamoto,
Y., Okada, S., 2015. Engrafted neural Stem/progenitor cells promote functional
Adams, M.M., Hicks, A.L., 2005. Spasticity after spinal cord injury. Spinal Cord 43, recovery through synapse reorganization with spared host neurons after spinal
577–586. cord injury. Stem Cell Rep. 5, 264-277.3.
Assinck, P., Duncan, G.J., Hilton, B.J., Plemel, J.R., Tetzlaff, W., 2017. Cell transplanta-
tion therapy for spinal cord injury. Nat. Neurosci. 20, 637–647.

Anda mungkin juga menyukai