Você está na página 1de 10

Available online at www.sciencedirect.

com

Experimental Neurology 209 (2008) 368 – 377


www.elsevier.com/locate/yexnr

Review
Stem cells for the treatment of spinal cord injury
Margaret Coutts, Hans S. Keirstead ⁎
Reeve-Irvine Research Center, Stem Cell Research Center, Department of Anatomy and Neurobiology, 2111 Gillespie Neuroscience Research Facility,
College of Medicine, University of California Irvine, Irvine, CA, USA

Received 21 August 2007; revised 29 August 2007; accepted 1 September 2007


Available online 12 September 2007

Abstract

This article reviews stem cell-based strategies for spinal cord injury repair, and practical issues concerning their translation to the clinic. Recent
progress in the stem cell field includes clinically compliant culture conditions and directed differentiation of both embryonic stem cells and
somatic stem cells. We provide a brief overview of the types of stem cells under evaluation, comparing their advantages and disadvantages for use
in human clinical trials. We review the practical considerations and risks that must be addressed before human treatments can begin. With a
growing understanding of these practical issues, stem cell biology, and spinal cord injury pathophysiology, stem cell-based therapies are moving
closer to clinical application.
© 2007 Elsevier Inc. All rights reserved.

Keywords: Human embryonic stem cell; Neural stem cell; Bone marrow stem cell; Differentiation; Clinical trial; Regulatory

Introduction clinical improvements are modest and many patients still face
significant neurologic dysfunction and disability.
This review summarizes stem cell-based therapeutic strate- Stem cells offer several approaches for SCI repair. Stem cell-
gies to treat spinal cord injury (SCI). Here we present the based therapies could, 1) replace damaged or diseased cells, 2)
emerging perspective that stem cell administration is a viable provide a cell-based electrical ‘relay’ between neurons above and
therapeutic strategy and discuss the challenges to its develop- below the injury, 3) ameliorate clinical deterioration and/or
ment. Pre-clinical research has demonstrated the effectiveness of facilitate regeneration by providing neuroprotective or growth
some stem cell-based treatments; however, several significant factors, or 4) play other indirect roles such as promoting neovas-
obstacles must be overcome before this research translates to the cularization or providing a permissive substrate for regeneration
clinic. These hurdles include: identifying the best source of stem of endogenous cells.
cells, optimizing their characteristics prior to transplant, re-
ducing the risks of stem cell therapy, developing large-scale Pathophysiology of SCI
manufacturing technologies, and fulfilling regulatory considera-
tions for government approval. Transplanting stem cell derivates will not be sufficient to
Current treatments for SCI (Baptiste and Fehlings, 2007) achieve complete functional restoration following SCI. The
include surgery to stabilize the injury site, high doses of corti- complex, reactive, and oftentimes multifocal nature of SCI
costeroids to help limit secondary injury processes and reha- presents several clinical challenges that must be overcome before
bilitative care. Preclinical studies with neuroprotective agents stem cell-based therapies can become clinical realities.
(such as minocycline and the Rho antagonist, Cethrinr) are SCI results in inflammation, progressive hemorrhagic ne-
encouraging, and clinical trials with these agents will soon crosis, edema, demyelination and cellular destruction. In the
begin. While these treatment options may provide benefits, earlier stages of SCI, there is vascular destruction, a loss of
neurons within the grey matter, and a loss of myelinating
⁎ Corresponding author. Fax: +1 949 824 5352. oligodendrocytes in the white matter. Axonopathy leads to a loss
E-mail address: hansk@uci.edu (H.S. Keirstead). of functional connections (denervation) and retraction of the
0014-4886/$ - see front matter © 2007 Elsevier Inc. All rights reserved.
doi:10.1016/j.expneurol.2007.09.002
M. Coutts, H.S. Keirstead / Experimental Neurology 209 (2008) 368–377 369

proximal axon. Cell death as a result of traumatic insult leads to reviewed in this section, including a discussion of their po-
further cell death via excitotoxicity. Excitotoxicity occurs as a tential for the treatment of SCI. Considerations include their
result of the accumulation of excitatory molecules such as potential for self-renewal, multipotency for providing a range
glutamate in the extracellular fluid, leading to overactivation of of differentiated cell types, and the potential to meet clinical
neurotransmitter receptors. High levels of calcium enter the cell needs.
and activate enzymes (phospholipases, proteases, etc.) that go on
to damage cell structures. This cell damage and other changes Human embryonic stem cells
often lead to induced cell death (apoptosis) and free radical-
mediated lipid oxidation (Liu et al., 1997). Thus, during the first Embryonic stem cells (ESCs) owe their discovery and
few days after injury there are many features detrimental to the potential to the early work of Kleinsmith and Pierce (1964).
survival and integration of transplanted cells (Hausmann, 2003). They demonstrated that a single embryonal carcinoma cell had
Inflammation plays a role in both the early and chronic stages the capacity to self-renew, and to generate multiple mature cell
of spinal cord injury (Hausmann, 2003; Fleming et al., 2006; types. Later work with cell lines derived from the inner cell mass
Donnelly and Popovich, in press). Macrophages, neutrophils of cultured blastocyst stage embryos underscored the plasticity
and T cells migrate in from the peripheral circulation and become and potential of ESCs (Evans and Kaufman, 1981; Martin,
activated. Microglia (normally resident in the spinal cord) also 1981). A landmark experiment used ESCs to generate an entire
become activated. Both activated microglia and macrophages mouse (Nagy et al., 1993). This astonishing plasticity was of
remove dead cells and debris via phagocytosis. The cytokines interest not only to the scientific community, but also to medical
and chemokines that these cells produce propagate the in- community interested in clinical approaches to repair and
flammatory processes, as does complement activation (Ander- replace damaged tissues. Successful isolation of human ESCs
son et al., 2004). Thus, inflammation in both the early and (hESCs) (Thomson et al., 1998) further heightened the interest in
chronic stages of spinal cord injury could also damage trans- the field. ESCs can be prepared from pre-implantation or
planted cells. blastocyst-stage embryos (from unused embryos created during
After the initial injury, the damage site expands from the in vitro fertilization procedures), by somatic cell nuclear transfer,
injury epicenter (many centimeters in a human). Analysis of or by parthenogenetic activation of eggs (Cibelli et al., 2002;
chronic SCI shows that, typically, portions of the more external Vrana et al., 2003). Unlike normal somatic cells, ESCs can be
white matter are spared while there is extensive damage of the grown in virtually unlimited quantities because they do not
more internally located grey matter. Within white matter, there is undergo senescence, retain high telomerase activity and normal
degeneration of both ascending and descending axons, and cell cycle signaling. With proper culture techniques, they do not
demyelination due to loss of oligodendrocytes. Chronic, prog- undergo the genomic, mitochondrial and epigenetic changes that
ressive demyelination is a persistent feature of SCI (Totoiu and lead to transformation (Zeng and Rao, 2007).
Keirstead, 2005). The astrocyte response begins immediately Of all stem cell types, ESCs currently show the greatest
after injury (proliferation, hypertrophy, etc.) and evolves over potential for the widest range of cell replacement therapies.
time. Reactive astrocytes produce extracellular matrix compo- They can be propagated in vitro almost indefinitely, can be
nents such as chondroitin and keratan sulfate proteoglycans. stably banked, and maintain a normal karyotype and
Ultimately, a scar-encapsulated cavity many times the size of the differentiation potential even after years of culture. These
initial injury forms. The glial scar presents both a physical barrier attributes make hESCs a commercially viable possibility for
and an inhibitory environment for axonal regeneration and production-scale processes. Originally, hESCs where cultivat-
remyelination (Fitch and Silver, 2008; Silver and Miller, 2004). ed on mouse fibroblast feeder layers with medium containing
Work from our laboratory strongly suggests that the gliotic bovine serum. Great progress has been made in cultivating
environment of scarred, chronic lesions contributes to the failure these cells, as current approaches use defined media with few
of remyelination by prohibiting transplant-derived oligoden- or no reagents derived from animal sources (Richards et al.,
drocytes from remyelinating (Keirstead et al., 2005). Therefore, 2002; Lee et al., 2005). This is important to preclude the
effective cell therapies for SCI likely necessitate transplantation possibility of cellular incorporation of zoonotic pathogens
during a brief therapeutic window: following acute inflamma- from the media. Other types of stem cells are not as thoroughly
tion, and prior to glial scar formation (Keirstead et al., 2005; characterized, and the methods for directing hESC differen-
Okano et al., 2003). tiation are most advanced. Several studies have indicated that
rodent ESCs can be directed in their differentiation to neuronal
Types of stem cells (Finley et al., 1996; Lang et al., 2004) or glial fates (Liu et al.,
2000; Billon et al., 2006). ESC-derived neurons can survive,
Stem cells can be divided into two broad categories, em- integrate and help restore function following transplantation
bryonic stem cells and somatic stem cells. Somatic stem cells into spinal cord injured rats (Deshpande et al., 2006). Human
include endogenous progenitor cells that repair and replace ESCs have been directed to differentiate into multipotent neural
tissues in our bodies. The category of somatic stem cells also precursors (Carpenter et al., 2001; Reubinoff et al., 2001), into
includes cells and cell lines derived from fetal tissues, neonatal low-purity motor neurons (Li et al., 2005) and recently into high-
tissues and adult tissues (e.g. neural stem cells, mesenchymal purity oligodendrocyte progenitors (Keirstead et al., 2005; Nistor
stem cells). These different types of stem cells will be briefly et al., 2005). Cumulatively, these studies demonstrate that the
370 M. Coutts, H.S. Keirstead / Experimental Neurology 209 (2008) 368–377

directed differentiation of hESCs into high-purity neural popula- reasons, it is not as conducive for neuronal differentiation. Some
tions is possible. studies show that more mature populations of neural precursors
will generate neurons, either because these cells do not respond
Neural stem cells to inhibitory signals, or alternatively, no longer need positive
environmental influences to attain a neuronal phenotype (Han
Endogenous neural stem cells (NSCs) exist within the central et al., 2002; Roy et al., 2004; Yan et al., 2007).
nervous system (CNS) of higher mammals, and recently, several Human neural tissue is generally obtained from cadavers,
groups have successfully isolated and expanded human NSCs which restricts supply. Furthermore, human neural tissue has a
from specific regions of the developing and adult brain (Snyder limited expansion potential in vitro. These factors limit the
et al., 1992; Lois and Alvarez-Buylla, 1993; Uchida et al., 2000) usefulness of NSCs in the clinical setting.
(Reynolds and Weiss, 1992), spinal cord (Mayer-Proschel et al.,
1997) and optic nerve (Shi et al., 1998). NSCs are of particular Olfactory ensheathing cells
interest for SCI repair. It is believed that they are already com-
mitted to a neural fate and hence will be easier to differentiate Olfactory ensheathing cells (OECs) are support cells that
into mature neural phenotypes, and less likely than ESCs to wrap olfactory axons and facilitate their regeneration throughout
become neoplastic. the life of mammalian species. They are reported to have
NSCs are neurogenic and gliogenic within the CNS exceptional plasticity, and importantly, allow neurons to cross a
throughout both development and adulthood. They can be glial scar as well as the PNS–CNS boundary (reviewed in
expanded in vitro by exposure to different growth factors, (Richter and Roskams, 2008) and (Raisman and Li, 2007)).
maintain some capacity for self-renewal even after several These cells are relatively easy to obtain from nasal biopsies and
freeze–thaw cycles, and are capable of generating differenti- could provide a source of autologous cells for transplant. Over
ated progeny that can functionally integrate (Caldwell et al., the last decade, a number of labs have used OECs in several
2001) and repair the damaged CNS (Nunes et al., 2003; different acute and chronic models of rodent SCI. In some cases,
Cummings et al., 2006; Ogawa et al., 2002; Iwanami et al., remyelination of axons and regeneration of damaged axons was
2005). NSCs can also protect against excitotoxicity and secrete reported along with a surprising degree of functional recovery.
neurotrophic growth factors (Llado et al., 2004; Lu et al., Other groups have not been able to reproduce these results, due
2003). Multipotent NSCs have recently been isolated from perhaps in part to differences in biological properties of primary
adult human subcortical white matter, and can be maintained in OECs with increasing age and/or passage number (Pastrana
vitro before being transplanted into fetal rat brain, where they et al., 2006). While there is great contention in the field, the
generate functionally competent neurons and glia (Nunes et al., majority of reports hold that these cells are supportive in repair
2003). However, adult NSCs divide less frequently than their processes, but the evidence that they facilitate regeneration of
embryonic counterparts and therefore may be more difficult to long axonal tracts is limited. In addition, it is not yet clear
expand into large cultures required for clinical applications whether they can be expanded in sufficient numbers for use in
(Doetsch et al., 1999; Morshead et al., 1998). Besides their human cell replacement strategies.
limited replication potential, there is evidence that the
differentiation potential of NSCs decreases with time in culture Mesenchymal stem cells
(Wright et al., 2006). In addition, differentiation of NSCs into
high purity populations has not been demonstrated, although Mesenchymal stem cells (MSC), bone marrow mononuclear
progress has been made to increase the percentage of either cells and umbilical cord blood are potentially rich sources of
neurons or astrocytes during in vitro differentiation using stem cells and a number of studies have used them to treat CNS
different combinations of growth factors (Caldwell et al., 2001; damage. Some of these studies have shown promising results,
Han et al., 2002) or alternate growth conditions (Yan et al., however, basic knowledge concerning their mechanism of
2007). Nonetheless, transplanted multipotent NSCs have been action and therapeutic potential is lacking. Despite the lack of
shown to reach regions of tissue damage, differentiate into understanding of the underling mechanisms, clinical trials for
myelinating oligodendrocytes, and cause clinical improvement SCI treatments are beginning (Yoon et al., 2007; Callera and do
following intraventricular, intravenous, intraspinal, or intra- Nascimento, 2006).
peritoneal delivery to various demyelinating or dysmyelinating MSC and bone marrow-derived cells have epitomized a
animal models (Einstein et al., 2003; Pluchino et al., 2003) central question in stem cell biology: whether stem cells from
(Ben-Hur et al., 2003; Bulte et al., 2003). one tissue can generate cells of another. The discovery of Y-
Approximately 35–40 reports have described neural stem chromosome-labeled neurons (Mezey et al., 2003), Purkinje
cell treatments for SCI (reviewed in Enzmann et al., 2006); most cells (Weimann et al., 2003) or hippocampal cells (Cogle et al.,
have used brain-derived NSC. Many of these studies showed 2004) within the brains of women who had received bone
that transplanted NSCs are able to generate astrocytes and marrow transplants from men ignited controversy of whether
oligodendrocytes very effectively. Generally, the production of this was a rare fusion event, or evidence of stem cell plasticity
neurons was low or not detectable. It has been suggested that the that could lead to useful therapies.
adult SCI environment is conducive for the differentiation of The application of these types of stem cells for CNS repair
NSCs to oligodendrocytes or astrocytes, but for undefined has been reviewed recently (Parr et al., 2007; Enzmann et al.,
M. Coutts, H.S. Keirstead / Experimental Neurology 209 (2008) 368–377 371

2006). These reviews take on the difficult task of comparing In contrast to the lack of neuronal regeneration within the adult
studies with greatly heterogeneous starting materials and dif- CNS, glial regeneration is successful following some insults to the
ferences in injury models, immunosuppression regimes, meth- CNS. Endogenous oligodendrocyte precursors can proliferate and
ods of transplantation, etc. The effort to evaluate these studies is differentiate in response to various types of injuries (Keirstead
well warranted, given the practical advantages of MSC and bone et al., 1998; McTigue et al., 2001; Wolswijk, 2000). Cell division
marrow-derived cells: they are easily obtainable, autologous is a prerequisite for remyelination (Keirstead et al., 1998),
transplantation is possible, they may be immuno-privileged, and which is characterized by thin and short myelin sheaths (Totoiu
they have the ability to migrate to areas of damage and in- and Keirstead, 2005) (Prineas et al., 1993). Interestingly, remy-
flammation. Most of these studies report improved function as a elination is dependent on the re-expression of developmentally-
result of implantation (usually determined by a subjectively- regulated genes (Capello et al., 1997), though it differs in some
scored locomotor test, though some use field potential recording respects from transcription in development (Ibanez et al., 2003).
(Akiyama et al., 2002a,b), and differentiation to oligodendrocytes, While oligodendrocytes proliferate and differentiate in response
or less frequently neurons, weeks to months after transplantation. to SCI, there is a net loss of myelin. Demyelination as a chron-
Some of the most convincing studies demonstrated that LacZ or ic, progressive problem in SCI (Guest et al., 2005; Totoiu and
GFP pre-labeled cells localized to Schwann- and oligodendro- Keirstead, 2005).
cyte-like cells following transplantation (Akiyama et al., 2002a,b) Remyelination is less efficient in old animals than in young
(Akiyama et al., 2002a,b; Sasaki et al., 2001). Opposite results animals (Shields et al., 2000) and less efficient after repeated
were obtained in other studies: there was no detection of trans- episodes of demyelination (Mason et al., 2004). It is possible
differentiation in the transplanted cells, even though functional that depletion of myelinogenic progenitors contributes to
improvement was noted (Koda et al., 2005). remyelination failure. Mature oligodendrocytes are incapable
How can these disparate results be reconciled? A broad of remyelinating axons (Keirstead and Blakemore, 1997; Crang
survey of the literature indicates that differentiation of et al., 1998) and there is no convincing evidence that diffe-
transplanted MSC and bone marrow stem cells is dictated by rentiated oligodendrocytes are able to revert to a progenitor
the environment, a concern that is made more relevant by the fact state. In addition, astrogliosis could contribute to the failure of
that means of in vivo differentiation to high-purity neural remyelination, forming a physical barrier and blocking access of
populations are lacking. Some degree of differentiation may well oligodendrocyte progenitors to demyelinated axons (Keirstead
be possible, especially given that a subset of ex vivo bone et al., 2005; Ibanez et al., 2003). Astrocytes can also express
marrow-derived cells express neuronal and oligodendroglial inhibitory molecules such as Jagged1 (which inhibits oligo-
markers (Goolsby et al., 2003; Steidl et al., 2002). Besides dendrocyte differentiation and process outgrowth) (John et al.,
directly replacing damaged oligodendrocytes and neurons, bone 2002). Failure of remyelination is probably due to a com-
marrow cells and MSC could play an important supportive role bination of environmental factors and innate characteristics of
in SCI therapies. They could create a more favorable envi- endogenous oligodendrocyte progenitors.
ronment for limiting damage and promoting regeneration, via In addition to directly replacing damaged neurons and
immunoregulation (Aggarwal and Pittenger, 2005; Noel et al., oligodendrocytes, stem cell therapies could also play an indirect
2007), expression of growth factors and cytokines (Song et al., role by supporting endogenous stem cells. Transplanted cells
2004), improved vascularization, providing a permissive growth could provide trophic factors (Zhang et al., 2006) or serve as a
substrate, and/or suppressing cavity formation (Hofstetter et al., substrate permissive for growth, differentiation, elongation or
2002). These different mechanisms are not mutually exclusive connection to other cells. Because the glial scar takes weeks to
and a number of them could contribute to improved outcomes. form a thick, rubbery obstruction, there is likely a ‘window of
Indeed, naive and genetically-modified MSC have been used in opportunity’ following injury during which the impediments to
combinatorial therapies in animal models of SCI (Lu et al., 2005; endogenous or transplant-mediated regeneration are fewer. This
Lu et al., 2004). period of time presents an opportunity for stem cell-derived
therapies to repair SCI.
Endogenous stem cells and progenitors
Clinical and scientific challenges
Neural stem cells (NSC) are present in the adult spinal cord,
however, it is clear that the capacity of endogenous NSCs to Potential stem cell therapies have a number of clinical and
replace lost cells after SCI is poor. Axonal regeneration from technical issues that need to be resolved before human clinical
pre-existing neurons is also poor, and it is likely that many of trials can begin. It will not be necessary to answer all of the
the same factors that prevent axonal regeneration also inhibit the questions regarding underlying molecular mechanisms of
function of endogenous NSC, neural progenitors and mature action, but there must be clear evidence that stem cell therapies
neurons. These factors include the formation of the glial scar, are safe, provide improved function and that the benefits
the lack of neurotrophic factors, inhibitory sulfated proteogly- outweigh the risks. Most important is an understanding of the
cans, and inhibitory myelin-associated molecules (reviewed in potential dangers of stem cell-based therapy. Preclinical studies
(Ramer et al., 2005) and (Fitch and Silver, 2008)). Falling will be essential in answering these questions; however, there
cAMP levels may also be inhibitory to regenerating cells will always be some uncertainties when translating preclinical
(Pearse et al., 2004) and differentiating progenitors. animal studies to the human condition. Hence, collective ‘buy-
372 M. Coutts, H.S. Keirstead / Experimental Neurology 209 (2008) 368–377

in’ from the scientific and medical communities is a critical efits and dangers of cell-based therapies for SCI (Hofstetter
prerequisite to a clinical trial. et al., 2005). In these studies, NSCs were transplanted into the
low-thoracic spinal cord of rats 1 week after injury. Functional
Do no harm recovery was noted in the affected hind limbs, but abnormal,
painful sensitivity developed in the forepaws (which had been
A number potential adverse effects must be evaluated before unaffected by the injury). Histology indicated that the transplant
a stem cell-based therapeutic can be applied to humans. The had differentiated in situ to a predominantly astrocytic pheno-
risks include tumorigenesis, immunological complications, type, and that these astrocytes promoted sprouting of sensory
allodynia (pain), or complications associated with an unexpect- fibers within the spinal cord that were associated with allodynia.
ed change in phenotype of the transplanted cells (for example, When the NSCs were directed to produce oligodendrocytes,
dedifferentiation or excess proliferation). further functional gains were attained in the hind limbs and
Tumor formation is a significant concern for transplant allodynia was avoided. These results have been verified by other
strategies involving embryonic stem cells; however, this risk workers (Hendricks et al., 2006). Careful studies such as these
decreases as the cells become more highly differentiated (i.e. less are critical to the preclinical development of stem cell-based
multipotent). Transplanted ESCs by definition form teratomas, therapies. Nonetheless, it should be appreciated that unexpected
which consist of endodermal, mesodermal and ectodermal adverse effects do occur in human clinical trials, despite exhaus-
lineages (Reubinoff et al., 2001). However, no rational medical tive preclinical development.
researcher would consider using undifferentiated ESCs in a Differentiation to undesirable cell types is a risk inherent to
human therapeutic strategy. Somatic stem cells are less pro- all multipotent cells. The NSCs used by Hoffstetter and col-
liferative and less multipotent; hence they are considered less leagues had the potential to produce neurons or oligodendro-
tumorigenic. cytes in vitro, but in the complex environment of the injured
Another important concern is immune rejection of the spinal cord they produced mostly astrocytes (Hofstetter et al.,
transplant. The host's immune system could destroy the 2005). This underscores the importance of evaluating the phe-
implanted cells and thereby eliminate any benefit that they notype of the cells after transplant. In addition to evaluating
conferred. Furthermore, the inflammatory response associated phenotype, preclinical studies must also evaluate proliferation
with cell rejection could cause additional harm to the patient. and migration of the transplanted cells. Uncontrolled prolifer-
HLA matching of the stem cell transplant to the host is one ation and migration is obviously undesirable; spreading beyond
method to avoid immune rejection. Unfortunately, the number of the implant site would increase the risk of adverse events such
available stem cell lines is limited, and it is likely that life-long as cerebrospinal fluid occlusion or emboli causing stroke.
immunosuppression will be necessary to prevent rejection of the
transplanted cells. The side-effects and risks of immunosup- The nature of the transplant
pression are significant and include nausea, vomiting, diarrhea,
liver and kidney toxicity, lowered counts of leukocytes and Preclinical studies will guide decisions on the best transplant
platelets, and increased susceptibility to infections and malig- position within the spinal cord, the number of implantation
nancies (Habwe, 2006). Also, it is not clear what effect im- sites, the appropriate cell number to transplant, and the optimal
munosuppression will have on the transplanted cells. This is not timing of transplants relative to the onset of injury. The optimal
a trivial issue, as evidenced by the fact that pancreatic islet time for transplantation of cells into the injured spinal cord is
transplantation was not successful until immunosuppression likely after acute inflammation and excitotoxicity has subsided,
regimes were developed that were less toxic to the transplanted and prior to the formation of extensive glial scar. Other practical
cells (Nanji and Shapiro, 2004). It is intriguing that ESC and matters concerning the transplant include efficient and safe
ESC-derived cells may not generate the same degree of immune methods to expand and differentiate stem cells. It is imperative
response associated with other transplanted tissues (Drukker that the cells maintain genetic and epigenetic stability, and do
et al., 2006; Utermohlen and Kronke, 2007). Such studies should not become senescent. The methods to induce differentiation
be repeated with the differentiated cells of interest. must also be efficient, so a high percentage of cells attain the
It is conceivable that transplant rejection may be overcome by desired phenotype.
tolerizing recipients prior to transplantation (Salama et al., 2001; A central question concerning the transplant is the optimal
Rosengard and Turka, 2001). Other potential means of avoiding degree of differentiation. While a less differentiated cell may
transplant rejection include generating stem cells that are better respond to environmental cues and show enhanced capa-
immunologically compatible with recipients using somatic cell city for migration and growth, a more differentiated cell may
nuclear transfer (SCNT—deriving a stem cell line by trans- mitigate the risks of inappropriate differentiation and neoplasia. A
ferring nuclei of recipient into donor stem cell or egg) or by broad review of the literature clearly indicates that lineage
developing a “universal donor” ESC line (Lengerke et al., 2007). restriction to the progenitor stage is essential to limit tumor
It remains to be seen if these later approaches will be successful formation and differentiation of undesirable cell types, and
or commercially viable. enhance integration. Lineage restriction prior to transplantation
Allodynia is a significant concern for stem cell-based overcomes limitations of the environment, which may not
strategies to treat SCI. In a thorough and thoughtful investiga- provide appropriate cues for targeted differentiation (for
tion, Hoffstetter and colleagues recently underscored the ben- example, efficient generation of neurons is limited in non-
M. Coutts, H.S. Keirstead / Experimental Neurology 209 (2008) 368–377 373

neurogenic regions of the CNS (Song et al., 2002; Shihabuddin combined cell transplantation with minocycline treatment and
et al., 2000)). The directed differentiation of stem cells to high growth factor (PDGF-AA, bFGF and EGF) delivery to enhance
purity lineages has recently been achieved (Nistor et al., 2005; cell survival (Karimi-Abdolrezaee et al., 2006). Findings from
Keirstead et al., 2005). This is a critical advance in stem cell the laboratory of Aileen Anderson indicated that multipotent
biology since it allows researchers to generate a potentially human CNS stem cells derived from fetal brain could be
inexhaustible supply of relatively pure, committed, lineage- maintained as neurospheres, and that their transplantation into
specific cells for transplantation. sites of moderate spinal cord contusion in adult NOD-scid mice
Stem cells are particularly amenable to genetic modification, resulted in differentiation to neurons and oligodendrocytes,
offering the potential to combine cell replacement therapy with remyelination by transplanted cells and improved locomotion
small molecule approaches to repair. Modifying the transplant (Cummings et al., 2005, 2006). Findings from the laboratory of
population to secrete growth factors or functional blockers of Lars Olson indicated that transduction of neural stem cells
endogenous inhibitors may augment the effect of cell replace- isolated from adult rat spinal cords with neurogenin 2 depressed
ment on injury pathogenesis or repair. The use of a reporter astroglial differentiation and increased oligodendroglial differ-
genes permit sorting of differentiated cells from residual entiation, and that their transplantation into sites of weight-drop
undifferentiated ES cells, mitigating the risk of tumor formation injury in adult rats resulted in remyelination and improved
(Tang et al., 2002). However, there are inherently unpredictable locomotion; notably, in the absence of neurogenin 2 transduc-
consequences when modifying the genome, and regulatory tion, neural stem cells differentiated primarily into astrocytes
agencies are understandably cautious with these methods. A after transplantation, and caused aberrant axonal sprouting
poignant example of this risk was realized in a gene therapy trial to associated with allodynia-like hypersensitivity of the forepaws
treat severe combined immunodeficiency, when the replacement (Hofstetter et al., 2005). These studies clearly indicate that
gene and its vector integrated into the patient genome near a myelinogenic transplants elicit functional recovery following
proto-oncogene, resulting in leukemia (Buckley, 2002). SCI. However, in all instances the mechanism of action is
unknown.
Evaluating success
Regulatory concerns
Ideally, stem cell-based therapies should be assessed using
cellular, physiological and behavioral assays. Many published Regulatory bodies such as the Food and Drug Administration
studies document functional improvements, but do not (FDA) and Institutional Review Boards (IRBs) will guide the
characterize the cells post-transplant nor demonstrate how the translation of stem cell therapies for SCI in animals to human
cells contributed to the outcome. Conversely, some publications clinical trials. Researchers, biotechnology companies, patients
provide evidence of successful differentiation using molecular and patient advocates should be aware of the regulatory issues
and immunological methods, but do not report behavioral out- that must be addressed before human trials can begin. These new
come. Safety studies are rarely undertaken by the scientific experimental therapies will have special concerns. First, the
community, which prioritizes discovery over development. spinal cord is a particularly sensitive and risky site to treat.
While this work is scientifically significant, it will be important Secondly, stem cell therapy is novel and entails a number of
to have a more complete picture before human clinical trials can complex issues that have never been addressed before. The
begin. Ideally, promising findings should be replicated in regulatory issues span categories as diverse as pharmacology
independent laboratories prior to the initiation of clinical trials. and toxicology, rules concerning the manufacture of cell and
A recent concurrence of data from multiple laboratories has tissue based products, xeno- and allogenenic transplantation,
led to the conclusion that myelinogenic transplants elicit histo- and surgical methods. At a minimum, a stem cell-based trans-
logic repair and functional recovery following SCI. Findings plant must be well defined in terms of sterility, purity, potency,
from our laboratory indicated that hESCs can be manipulated in identity, stability, safety and efficacy.
vitro to generate high purity human oligodendrocyte progenitor One regulatory issue common to many different types of
cells (OPCs) in large quantities (Nistor et al., 2005), and that clinical trials is the concept of “process control”. The stem cells
their transplantation into sites of moderate spinal cord contusion and all of the materials that come in contact with them through-
in adult rats results in differentiation to mature oligodendrocytes, out their preparation must have a completely traceable identity
remyelination by transplanted cells and improved locomotion and source. Further, the reagents must have documented quality
(Keirstead et al., 2005). Follow-up studies indicated that the assurances and quality controls. The stem cells themselves must
procedure was safe, in that the transplant was not associated with be screened for the presence of retroviruses and infectious
tumor formation, scarring, tissue pathogenesis, or behavioral pathogens. Karyotypic analysis must be performed to ensure
decline (Cloutier et al., 2006). Findings from the laboratory of there are no gross genetic changes. There can be no undif-
Michael Fehlings indicated that multipotent adult neural ferentiated embryonic stem cells in the transplant population,
precursor cells can be isolated from the subventricular zone of which may have the capacity to form tumors. This last qua-
the forebrain, and that their transplantation into sites of lification can be accomplished by checking for predetermined
aneurysm clip-induced spinal cord compression in adult rats markers such as OCT4, SSEA4, and TRA81.
resulted in differentiation to mature oligodendrocytes, remyeli- Other critical considerations include four toxicological
nation by transplanted cells and improved locomotion; this study parameters: migration of the cell transplant, inappropriate
374 M. Coutts, H.S. Keirstead / Experimental Neurology 209 (2008) 368–377

cellular differentiation, the possibility of tumor formation, and models before risking treatments in human patients. Patients,
host immune responses. Cell migration (bio-distribution) from clinicians, scientists and regulatory agencies must also consider
the implant site can be evaluated in preclinical testing, using longer-term safety issues of stem cell therapies, including the
reverse transcription polymerase chain reaction (RT-PCR) for likelihood of life-long immunosuppressive regimes. While
specific markers on isolated organs following transplantation. attaining a cure will be difficult, the spinal cord research field
Inappropriate cellular differentiation must also be defined in should acknowledge the complexity of the challenge and not be
preclinical testing. Clinical deficits could be worsened if, for deterred from pursuing it.
example, stem cells transplanted into the spinal cord of a patient
inappropriately differentiated into cartilage-producing cells. References
Inappropriate differentiation could also include de-differentiation
to a pluripotent state, which would be a risk for tumor formation. Aggarwal, S., Pittenger, M.F., 2005. Human mesenchymal stem cells modulate
Tumor formation is of particular concern because the cells allogeneic immune cell responses. Blood 105, 1815–1822.
implanted in the spinal cord would be difficult or impossible to Akiyama, Y., Radtke, C., Honmou, O., Kocsis, J.D., 2002a. Remyelination of
the spinal cord following intravenous delivery of bone marrow cells. Glia
remove. Host immune responses must be evaluated in terms of
39, 229–236.
potential harm both to the host and to the implant. Since the cells Akiyama, Y., Radtke, C., Kocsis, J.D., 2002b. Remyelination of the rat spinal
are foreign to the body, some immune response is expected. cord by transplantation of identified bone marrow stromal cells. J. Neurosci. 22,
Immunosuppression will probably be required, as will histologic 6623–6630.
studies to define any immune activation. Anderson, A.J., Robert, S., Huang, W., Young, W., Cotman, C.W., 2004.
Activation of complement pathways after contusion-induced spinal cord
Before clinical trials can begin, standard operating proce-
injury. J. Neurotrauma 21, 1831–1846.
dures must be compiled to standardize the methods used in Baptiste, D.C., Fehlings, M.G., 2007. Update on the treatment of spinal cord
preparing and delivering the cell therapy (including cell injury. Prog. Brain Res. 161, 217–233.
cultivation and differentiation methods, transplantation surgery, Ben-Hur, T., Einstein, O., Mizrachi-Kol, R., Ben-Menachem, O., Reinhartz, E.,
etc.). Surgical teams will need to be assembled and well trained Karussis, D., Abramsky, O., 2003. Transplanted multipotential neural
precursor cells migrate into the inflamed white matter in response to
in every aspect of the transplantation. Equipment may be
experimental autoimmune encephalomyelitis. Glia 41, 73–80.
subject to additional regulatory requirements prior to conduct- Billon, N., Jolicoeur, C., Raff, M., 2006. Generation and characterization of
ing human trials. While much of the equipment is used routinely oligodendrocytes from lineage-selectable embryonic stem cells in vitro.
for clinical neurosurgical practices (e.g. syringes, infusion Methods Mol. Biol. 330, 15–32.
pumps, stereotaxic equipment), the application to stem cell Buckley, R.H., 2002. Gene therapy for SCID—a complication after remarkable
progress. Lancet 360, 1185–1186.
clinical trials will require additional study. Preclinical studies
Bulte, J.W., Ben-Hur, T., Miller, B.R., Mizrachi-Kol, R., Einstein, O., Reinhartz, E.,
using animals have developed standard operating procedures, Zywicke, H.A., Douglas, T., Frank, J.A., 2003. MR microscopy of magnetically
however some aspects must be translated to human scale, such labeled neurospheres transplanted into the Lewis EAE rat brain. Magn. Reson.
as the maximum volume and number of cells that can be Med. 50, 201–205.
transplanted, or the number of injection sites. Caldwell, M.A., He, X., Wilkie, N., Pollack, S., Marshall, G., Wafford, K.A.,
Svendsen, C.N., 2001. Growth factors regulate the survival and fate of cells
derived from human neurospheres. Nat. Biotechnol. 19, 475–479.
Conclusions and summary Callera, F., do Nascimento, R.X., 2006. Delivery of autologous bone marrow
precursor cells into the spinal cord via lumbar puncture technique in patients
The literature over the past few decades clearly documents a with spinal cord injury: a preliminary safety study. Exp. Hematol. 34,
growing appreciation of the very complicated pathophysiology 130–131.
Capello, E., Voskuhl, R.R., McFarland, H.F., Raine, C.S., 1997. Multiple
of SCI. During this time, the field of stem cell biology has
sclerosis: re-expression of a developmental gene in chronic lesions correlates
emerged. This new field has the potential to bring therapies to with remyelination. Ann. Neurol. 41, 797–805.
previously untreatable diseases and injuries. Carpenter, M.K., Inokuma, M.S., Denham, J., Mujtaba, T., Chiu, C.P., Rao, M.S.,
Animal models have shown some positive results, validating 2001. Enrichment of neurons and neural precursors from human embryonic
scientific principles and strategies. In addition, progress has been stem cells. Exp. Neurol. 172, 383–397.
Cibelli, J.B., Grant, K.A., Chapman, K.B., Cunniff, K., Worst, T., Green, H.L.,
made with a number of practical concerns. Commercial-scale
Walker, S.J., Gutin, P.H., Vilner, L., Tabar, V., Dominko, T., Kane, J.,
production of NSC and ESC cultures is progressing, a necessary Wettstein, P.J., Lanza, R.P., Studer, L., Vrana, K.E., West, M.D., 2002.
step in creating a sufficient volume of cells for human trials. Parthenogenetic stem cells in nonhuman primates. Science 295, 819.
Great improvements have been made towards creating “defined” Cloutier, F., Siegenthaler, M.M., Nistor, G., Keirstead, H.S., 2006. Transplantation
growth conditions to yield safe, transplantable cells, free from of human embryonic stem cell-derived oligodendrocyte progenitors into rat
spinal cord injuries does not cause harm. Regen. Med. 1, 469–479.
human and zoonotic pathogens. Differentiation protocols are
Cogle, C.R., Yachnis, A.T., Laywell, E.D., Zander, D.S., Wingard, J.R.,
improving, yielding cells of higher purity and better function. Steindler, D.A., Scott, E.W., 2004. Bone marrow transdifferentiation in brain
Spinal cord researchers are appreciating the complexity of SCI, after transplantation: a retrospective study. Lancet 363, 1432–1437.
and appreciate that a multifaceted approach will be needed (for Crang, A.J., Gilson, J., Blakemore, W.F., 1998. The demonstration by
example, differentiated stem cells, plus trophic factors and tissue transplantation of the very restricted remyelinating potential of post-mitotic
oligodendrocytes. J. Neurocytol. 27, 541–553.
engineering of a permissive environment). These experiments
Cummings, B.J., Uchida, N., Tamaki, S.J., Salazar, D.L., Hooshmand, M.,
will be difficult to design and execute such that meaningful Summers, R., Gage, F.H., Anderson, A.J., 2005. Human neural stem cells
results can be obtained. Unquestionably, there must be an differentiate and promote locomotor recovery in spinal cord-injured mice.
accumulation of studies that show functional recovery in animal Proc. Natl. Acad. Sci. U. S. A. 102, 14069–14074.
M. Coutts, H.S. Keirstead / Experimental Neurology 209 (2008) 368–377 375

Cummings, B.J., Uchida, N., Tamaki, S.J., Anderson, A.J., 2006. Human neural Nomura, T., Toyama, Y., Okano, H., 2005. Transplantation of human
stem cell differentiation following transplantation into spinal cord injured mice: neural stem cells for spinal cord injury in primates. J. Neurosci. Res. 80,
association with recovery of locomotor function. Neurol. Res. 28, 474–481. 182–190.
Deshpande, D.M., Kim, Y.S., Martinez, T., Carmen, J., Dike, S., Shats, I., Rubin, John, G.R., Shankar, S.L., Shafit-Zagardo, B., Massimi, A., Lee, S.C., Raine, C.S.,
L.L., Drummond, J., Krishnan, C., Hoke, A., Maragakis, N., Shefner, J., Brosnan, C.F., 2002. Multiple sclerosis: re-expression of a develop-
Rothstein, J.D., Kerr, D.A., 2006. Recovery from paralysis in adult rats mental pathway that restricts oligodendrocyte maturation. Nat. Med. 8,
using embryonic stem cells. Ann. Neurol. 60, 32–44. 1115–1121.
Doetsch, F., Caille, I., Lim, D.A., Garcia-Verdugo, J.M., Alvarez-Buylla, A., Karimi-Abdolrezaee, S., Eftekharpour, E., Wang, J., Morshead, C.M., Fehlings,
1999. Subventricular zone astrocytes are neural stem cells in the adult M.G., 2006. Delayed transplantation of adult neural precursor cells promotes
mammalian brain. Cell 97, 703–716. remyelination and functional neurological recovery after spinal cord injury.
Donnelly, D.J., Popovich, P.G., 2008. Inflammation and its role in neuroprotec- J. Neurosci. 26, 3377–3389.
tion, axonal regeneration and functional recovery after spinal cord injury. Keirstead, H.S., Blakemore, W.F., 1997. Identification of post-mitotic
Exp. Neurol. 209, 378–388. oligodendrocytes incapable of remyelination within the demyelinated
Drukker, M., Katchman, H., Katz, G., Even-Tov Friedman, S., Shezen, E., adult spinal cord. J. Neuropathol. Exp. Neurol. 56, 1191–1201.
Hornstein, E., Mandelboim, O., Reisner, Y., Benvenisty, N., 2006. Human Keirstead, H.S., Levine, J.M., Blakemore, W.F., 1998. Response of the
embryonic stem cells and their differentiated derivatives are less susceptible oligodendrocyte progenitor cell population (defined by NG2 labelling) to
to immune rejection than adult cells. Stem Cells 24, 221–229. demyelination of the adult spinal cord. Glia 22, 161–170.
Einstein, O., Karussis, D., Grigoriadis, N., Mizrachi-Kol, R., Reinhartz, E., Keirstead, H.S., Nistor, G., Bernal, G., Totoiu, M., Cloutier, F., Sharp, K.,
Abramsky, O., Ben-Hur, T., 2003. Intraventricular transplantation of neural Steward, O., 2005. Human embryonic stem cell-derived oligodendrocyte
precursor cell spheres attenuates acute experimental allergic encephalomy- progenitor cell transplants remyelinate and restore locomotion after spinal
elitis. Mol. Cell. Neurosci. 24, 1074–1082. cord injury. J. Neurosci. 25, 4694–4705.
Enzmann, G.U., Benton, R.L., Talbott, J.F., Cao, Q., Whittemore, S.R., 2006. Kleinsmith, L.J., Pierce Jr., G.B., 1964. Multipotentiality of single embryonal
Functional considerations of stem cell transplantation therapy for spinal cord carcinoma cells. Cancer Res. 24, 1544–1551.
repair. J. Neurotrauma 23, 479–495. Koda, M., Okada, S., Nakayama, T., Koshizuka, S., Kamada, T., Nishio, Y.,
Evans, M.J., Kaufman, M.H., 1981. Establishment in culture of pluripotential Someya, Y., Yoshinaga, K., Okawa, A., Moriya, H., Yamazaki, M., 2005.
cells from mouse embryos. Nature 292, 154–156. Hematopoietic stem cell and marrow stromal cell for spinal cord injury in
Finley, M.F., Kulkarni, N., Huettner, J.E., 1996. Synapse formation and mice. Neuroreport 16, 1763–1767.
establishment of neuronal polarity by P19 embryonic carcinoma cells and Lang, K.J., Rathjen, J., Vassilieva, S., Rathjen, P.D., 2004. Differentiation of
embryonic stem cells. J. Neurosci. 16, 1056–1065. embryonic stem cells to a neural fate: a route to re-building the nervous
Fitch, M.T., Silver, J., 2008. CNS injury, glial scars, and inflammation: system? J. Neurosci. Res. 76, 184–192.
inhibitory extracellular matrices and regeneration failure. Exp. Neurol. 209, Lee, J.B., Lee, J.E., Park, J.H., Kim, S.J., Kim, M.K., Roh, S.I., Yoon, H.S.,
294–301. 2005. Establishment and maintenance of human embryonic stem cell lines
Fleming, J.C., Norenberg, M.D., Ramsay, D.A., Dekaban, G.A., Marcillo, A.E., on human feeder cells derived from uterine endometrium under serum-free
Saenz, A.D., Pasquale-Styles, M., Dietrich, W.D., Weaver, L.C., 2006. The condition. Biol. Reprod. 72, 42–49.
cellular inflammatory response in human spinal cords after injury. Brain 129, Lengerke, C., Kim, K., Lerou, P., Daley, G.Q., 2007. Differentiation potential
3249–3269. of histocompatible parthenogenetic embryonic stem cells. Ann. N.Y. Acad.
Goolsby, J., Marty, M.C., Heletz, D., Chiappelli, J., Tashko, G., Yarnell, D., Fishman, Sci. 1106, 209–218.
P.S., Dhib-Jalbut, S., Bever Jr., C.T., Pessac, B., Trisler, D., 2003. Hematopoietic Li, X.J., Du, Z.W., Zarnowska, E.D., Pankratz, M., Hansen, L.O., Pearce, R.A.,
progenitors express neural genes. Proc. Natl. Acad. Sci. U. S. A. 100, Zhang, S.C., 2005. Specification of motoneurons from human embryonic
14926–14931. stem cells. Nat. Biotechnol. 23, 215–221.
Guest, J.D., Hiester, E.D., Bunge, R.P., 2005. Demyelination and Schwann cell Liu, S., Qu, Y., Stewart, T.J., Howard, M.J., Chakrabortty, S., Holekamp, T.F.,
responses adjacent to injury epicenter cavities following chronic human McDonald, J.W., 2000. Embryonic stem cells differentiate into oligoden-
spinal cord injury. Exp. Neurol. 192, 384–393. drocytes and myelinate in culture and after spinal cord transplantation. Proc.
Habwe, V.Q., 2006. Posttransplantation quality of life: more than graft function. Natl. Acad. Sci. U. S. A. 97, 6126–6131.
Am. J. Kidney Dis. 47, S98–S110. Liu, X.Z., Xu, X.M., Hu, R., Du, C., Zhang, S.X., McDonald, J.W., Dong, H.X.,
Han, S.S., Kang, D.Y., Mujtaba, T., Rao, M.S., Fischer, I., 2002. Grafted Wu, Y.J., Fan, G.S., Jacquin, M.F., Hsu, C.Y., Choi, D.W., 1997. Neuronal
lineage-restricted precursors differentiate exclusively into neurons in the and glial apoptosis after traumatic spinal cord injury. J. Neurosci. 17,
adult spinal cord. Exp. Neurol. 177, 360–375. 5395–5406.
Hausmann, O.N., 2003. Post-traumatic inflammation following spinal cord Llado, J., Haenggeli, C., Maragakis, N.J., Snyder, E.Y., Rothstein, J.D., 2004.
injury. Spinal Cord 41, 369–378. Neural stem cells protect against glutamate-induced excitotoxicity and
Hendricks, W.A., Pak, E.S., Owensby, J.P., Menta, K.J., Glazova, M., Moretto, promote survival of injured motor neurons through the secretion of
J., Hollis, S., Brewer, K.L., Murashov, A.K., 2006. Predifferentiated neurotrophic factors. Mol. Cell. Neurosci. 27, 322–331.
embryonic stem cells prevent chronic pain behaviors and restore sensory Lois, C., Alvarez-Buylla, A., 1993. Proliferating subventricular zone cells in the
function following spinal cord injury in mice. Mol. Med. 12, 34–46. adult mammalian forebrain can differentiate into neurons and glia. Proc.
Hofstetter, C.P., Holmstrom, N.A., Lilja, J.A., Schweinhardt, P., Hao, J., Natl. Acad. Sci. U. S. A. 90, 2074–2077.
Spenger, C., Wiesenfeld-Hallin, Z., Kurpad, S.N., Frisen, J., Olson, L., 2005. Lu, P., Jones, L.L., Snyder, E.Y., Tuszynski, M.H., 2003. Neural stem cells
Allodynia limits the usefulness of intraspinal neural stem cell grafts; directed constitutively secrete neurotrophic factors and promote extensive host
differentiation improves outcome. Nat. Neurosci. 8, 346–353. axonal growth after spinal cord injury. Exp. Neurol. 181, 115–129.
Hofstetter, C.P., Schwarz, E.J., Hess, D., Widenfalk, J., El Manira, A., Prockop, Lu, P., Yang, H., Jones, L.L., Filbin, M.T., Tuszynski, M.H., 2004.
D.J., Olson, L., 2002. Marrow stromal cells form guiding strands in the Combinatorial therapy with neurotrophins and cAMP promotes ax-
injured spinal cord and promote recovery. Proc. Natl. Acad. Sci. U. S. A. 99, onal regeneration beyond sites of spinal cord injury. J. Neurosci. 24,
2199–2204. 6402–6409.
Ibanez, C., Shields, S.A., El-Etr, M., Leonelli, E., Magnaghi, V., Li, W.W., Sim, Lu, P., Jones, L.L., Tuszynski, M.H., 2005. BDNF-expressing marrow stromal
F.J., Baulieu, E.E., Melcangi, R.C., Schumacher, M., Franklin, R.J., 2003. cells support extensive axonal growth at sites of spinal cord injury. Exp.
Steroids and the reversal of age-associated changes in myelination and Neurol. 191, 344–360.
remyelination. Prog. Neurobiol. 71, 49–56. Martin, G.R., 1981. Isolation of a pluripotent cell line from early mouse embryos
Iwanami, A., Kaneko, S., Nakamura, M., Kanemura, Y., Mori, H., Kobayashi, S., cultured in medium conditioned by teratocarcinoma stem cells. Proc. Natl.
Yamasaki, M., Momoshima, S., Ishii, H., Ando, K., Tanioka, Y., Tamaoki, N., Acad. Sci. U. S. A. 78, 7634–7638.
376 M. Coutts, H.S. Keirstead / Experimental Neurology 209 (2008) 368–377

Mason, J.L., Toews, A., Hostettler, J.D., Morell, P., Suzuki, K., Goldman, J.E., Richter, M.W., Roskams, A.J., 2008. Olfactory ensheathing cell transplantation
Matsushima, G.K., 2004. Oligodendrocytes and progenitors become progres- following spinal cord injury: hype or hope? Exp. Neurol. 209, 353–367.
sively depleted within chronically demyelinated lesions. Am. J. Pathol. 164, Rosengard, B.R., Turka, L.A., 2001. The tolerant recipient: looking great in
1673–1682. someone else's genes. J. Clin. Invest. 107, 33–34.
Mayer-Proschel, M., Kalyani, A.J., Mujtaba, T., Rao, M.S., 1997. Isolation of Roy, N.S., Nakano, T., Keyoung, H.M., Windrem, M., Rashbaum, W.K., Alonso,
lineage-restricted neuronal precursors from multipotent neuroepithelial stem M.L., Kang, J., Peng, W., Carpenter, M.K., Lin, J., Nedergaard, M.,
cells. Neuron 19, 773–785. Goldman, S.A., 2004. Telomerase immortalization of neuronally restricted
McTigue, D.M., Wei, P., Stokes, B.T., 2001. Proliferation of NG2-positive cells progenitor cells derived from the human fetal spinal cord. Nat. Biotechnol. 22,
and altered oligodendrocyte numbers in the contused rat spinal cord. 297–305.
J. Neurosci. 21, 3392–3400. Salama, A.D., Remuzzi, G., Harmon, W.E., Sayegh, M.H., 2001. Challenges to
Mezey, E., Key, S., Vogelsang, G., Szalayova, I., Lange, G.D., Crain, B., 2003. achieving clinical transplantation tolerance. J. Clin. Invest. 108, 943–948.
Transplanted bone marrow generates new neurons in human brains. Proc. Sasaki, M., Honmou, O., Akiyama, Y., Uede, T., Hashi, K., Kocsis, J.D., 2001.
Natl. Acad. Sci. U. S. A. 100, 1364–1369. Transplantation of an acutely isolated bone marrow fraction repairs
Morshead, C.M., Craig, C.G., van der Kooy, D., 1998. In vivo clonal analyses demyelinated adult rat spinal cord axons. Glia 35, 26–34.
reveal the properties of endogenous neural stem cell proliferation in the adult Shi, J., Marinovich, A., Barres, B.A., 1998. Purification and characterization of
mammalian forebrain. Development 125, 2251–2261. adult oligodendrocyte precursor cells from the rat optic nerve. J. Neurosci. 18,
Nagy, A., Rossant, J., Nagy, R., Abramow-Newerly, W., Roder, J.C., 1993. 4627–4636.
Derivation of completely cell culture-derived mice from early-passage Shields, S., Gilson, J., Blakemore, W., Franklin, R., 2000. Remyelination occurs
embryonic stem cells. Proc. Natl. Acad. Sci. U. S. A. 90, 8424–8428. as extensively but more slowly in old rats compared to young rats following
Nanji, S.A., Shapiro, A.M., 2004. Islet transplantation in patients with diabetes fliotoxin-induced CNS demyelination. Glia 29, 102.
mellitus: choice of immunosuppression. BioDrugs 18, 315–328. Shihabuddin, L.S., Horner, P.J., Ray, J., Gage, F.H., 2000. Adult spinal cord
Nistor, G.I., Totoiu, M.O., Haque, N., Carpenter, M.K., Keirstead, H.S., 2005. stem cells generate neurons after transplantation in the adult dentate gyrus.
Human embryonic stem cells differentiate into oligodendrocytes in high J. Neurosci. 20, 8727–8735.
purity and myelinate after spinal cord transplantation. Glia 49, 385–396. Silver, J., Miller, J.H., 2004. Regeneration beyond the glial scar. Nat. Rev.
Noel, D., Djouad, F., Bouffi, C., Mrugala, D., Jorgensen, C., 2007. Multipotent Neurosci. 5, 146–151.
mesenchymal stromal cells and immune tolerance. Leuk. Lymphoma 48, Snyder, E.Y., Deitcher, D.L., Walsh, C., Arnold-Aldea, S., Hartwieg, E.A.,
1283–1289. Cepko, C.L., 1992. Multipotent neural cell lines can engraft and participate
Nunes, M.C., Roy, N.S., Keyoung, H.M., Goodman, R.R., McKhann II, G., in development of mouse cerebellum. Cell 68, 33–51.
Jiang, L., Kang, J., Nedergaard, M., Goldman, S.A., 2003. Identification and Song, H., Stevens, C.F., Gage, F.H., 2002. Astroglia induce neurogenesis from
isolation of multipotential neural progenitor cells from the subcortical white adult neural stem cells. Nature 417, 39–44.
matter of the adult human brain. Nat. Med. 9, 439–447. Song, S., Kamath, S., Mosquera, D., Zigova, T., Sanberg, P., Vesely, D.L.,
Ogawa, Y., Sawamoto, K., Miyata, T., Miyao, S., Watanabe, M., Nakamura, M., Sanchez-Ramos, J., 2004. Expression of brain natriuretic peptide by human
Bregman, B.S., Koike, M., Uchiyama, Y., Toyama, Y., Okano, H., 2002. bone marrow stromal cells. Exp. Neurol. 185, 191–197.
Transplantation of in vitro-expanded fetal neural progenitor cells results in Steidl, U., Kronenwett, R., Rohr, U.P., Fenk, R., Kliszewski, S., Maercker, C.,
neurogenesis and functional recovery after spinal cord contusion injury in Neubert, P., Aivado, M., Koch, J., Modlich, O., Bojar, H., Gattermann, N.,
adult rats. J. Neurosci. Res. 69, 925–933. Haas, R., 2002. Gene expression profiling identifies significant dif-
Okano, H., Ogawa, Y., Nakamura, M., Kaneko, S., Iwanami, A., Toyama, Y., ferences between the molecular phenotypes of bone marrow-derived
2003. Transplantation of neural stem cells into the spinal cord after injury. and circulating human CD34+ hematopoietic stem cells. Blood 99, 2037–2044.
Semin. Cell Dev. Biol. 14, 191–198. Tang, F., Shang, K., Wang, X., Gu, J., 2002. Differentiation of embryonic stem
Parr, A.M., Tator, C.H., Keating, A., 2007. Bone marrow-derived mesenchymal cell to astrocytes visualized by green fluorescent protein. Cell. Mol.
stromal cells for the repair of central nervous system injury. Bone Marrow Neurobiol. 22, 95–101.
Transplant. 40, 609–619. Thomson, J.A., Itskovitz-Eldor, J., Shapiro, S.S., Waknitz, M.A., Swiergiel, J.J.,
Pastrana, E., Moreno-Flores, M.T., Gurzov, E.N., Avila, J., Wandosell, F., Diaz- Marshall, V.S., Jones, J.M., 1998. Embryonic stem cell lines derived from
Nido, J., 2006. Genes associated with adult axon regeneration promoted by human blastocysts. Science 282, 1145–1147.
olfactory ensheathing cells: a new role for matrix metalloproteinase 2. Totoiu, M.O., Keirstead, H.S., 2005. Spinal cord injury is accompanied by
J. Neurosci. 26, 5347–5359. chronic progressive demyelination. J. Comp. Neurol. 486, 373–383.
Pearse, D.D., Pereira, F.C., Marcillo, A.E., Bates, M.L., Berrocal, Y.A., Filbin, Uchida, N., Buck, D.W., He, D., Reitsma, M.J., Masek, M., Phan, T.V., Tsukamoto,
M.T., Bunge, M.B., 2004. cAMP and Schwann cells promote axonal growth A.S., Gage, F.H., Weissman, I.L., 2000. Direct isolation of human central
and functional recovery after spinal cord injury. Nat. Med. 10, 610–616. nervous system stem cells. Proc. Natl. Acad. Sci. U. S. A. 97, 14720–14725.
Pluchino, S., Quattrini, A., Brambilla, E., Gritti, A., Salani, G., Dina, G., Galli, Utermohlen, O., Kronke, M., 2007. Survival of priceless cells: active and
R., Del Carro, U., Amadio, S., Bergami, A., Furlan, R., Comi, G., Vescovi, passive protection of embryonic stem cells against immune destruction.
A.L., Martino, G., 2003. Injection of adult neurospheres induces recovery in Arch. Biochem. Biophys. 462, 273–277.
a chronic model of multiple sclerosis. Nature 422, 688–694. Vrana, K.E., Hipp, J.D., Goss, A.M., McCool, B.A., Riddle, D.R., Walker, S.J.,
Prineas, J.W., Barnard, R.O., Kwon, E.E., Sharer, L.R., Cho, E.S., 1993. Multiple Wettstein, P.J., Studer, L.P., Tabar, V., Cunniff, K., Chapman, K., Vilner, L.,
sclerosis: remyelination of nascent lesions. Ann. Neurol. 33, 137–151. West, M.D., Grant, K.A., Cibelli, J.B., 2003. Nonhuman primate partheno-
Raisman, G., Li, Y., 2007. Repair of neural pathways by olfactory ensheathing genetic stem cells. Proc. Natl. Acad. Sci. U. S. A. 100 (Suppl 1), 11911–11916.
cells. Nat. Rev., Neurosci. 8, 312–319. Weimann, J.M., Charlton, C.A., Brazelton, T.R., Hackman, R.C., Blau, H.M.,
Ramer, L.M., Ramer, M.S., Steeves, J.D., 2005. Setting the stage for functional 2003. Contribution of transplanted bone marrow cells to Purkinje neurons in
repair of spinal cord injuries: a cast of thousands. Spinal Cord 43, 134–161. human adult brains. Proc. Natl. Acad. Sci. U. S. A. 100, 2088–2093.
Reubinoff, B.E., Itsykson, P., Turetsky, T., Pera, M.F., Reinhartz, E., Itzik, A., Wolswijk, G., 2000. Oligodendrocyte survival, loss and birth in lesions of
Ben-Hur, T., 2001. Neural progenitors from human embryonic stem cells. chronic-stage multiple sclerosis. Brain 123 (Pt 1), 105–115.
Nat. Biotechnol. 19, 1134–1140. Wright, L.S., Prowse, K.R., Wallace, K., Linskens, M.H., Svendsen, C.N., 2006.
Reynolds, B.A., Weiss, S., 1992. Generation of neurons and astrocytes from Human progenitor cells isolated from the developing cortex undergo
isolated cells of the adult mammalian central nervous system. Science 255, decreased neurogenesis and eventual senescence following expansion in
1707–1710. vitro. Exp. Cell Res. 312, 2107–2120.
Richards, M., Fong, C.Y., Chan, W.K., Wong, P.C., Bongso, A., 2002. Human Yan, J., Xu, L., Welsh, A.M., Hatfield, G., Hazel, T., Johe, K., Koliatsos, V.E.,
feeders support prolonged undifferentiated growth of human inner cell 2007. Extensive neuronal differentiation of human neural stem cell grafts in
masses and embryonic stem cells. Nat. Biotechnol. 20, 933–936. adult rat spinal cord. PLoS. Med. 4, e39.
M. Coutts, H.S. Keirstead / Experimental Neurology 209 (2008) 368–377 377

Yoon, S.H., Shim, Y.S., Park, Y.H., Chung, J.K., Nam, J.H., Kim, M.O., Park, Zeng, X., Rao, M.S., 2007. Human embryonic stem cells: long term stability,
H.C., Park, S.R., Min, B.H., Kim, E.Y., Choi, B.H., Park, H., Ha, Y., 2007. absence of senescence and a potential cell source for neural replacement.
Complete spinal cord injury treatment using autologous bone marrow cell Neuroscience 145, 1348–1358.
transplantation and bone marrow stimulation with granulocyte macro- Zhang, Y.W., Denham, J., Thies, R.S., 2006. Oligodendrocyte progenitor cells
phage-colony stimulating factor: phase I/II clinical trial. Stem Cells 25, derived from human embryonic stem cells express neurotrophic factors.
2066–2073. Stem Cells Dev. 15, 943–952.

Você também pode gostar