<<

Stem Rev DOIIO.I007/s12015-007-9006-9

From Stem Cells to : Prospects for Therapy

Cui P. Chen· Mary E. Kiel • Dorota Sadowski • Randall D. McKinnon

Abstract is an autoimmune disease that brain and injury, to later onset diseases including destroys -fonning oligodendrocytes of the CNS. Parkinson's and Alzheimer's. Individuals stricken face a While the damage can be partially controlled using anti- long and difficult clinical management with treatment op- inflammatory cytokines and steroids, endogenous repair is tions that at best delay but do not stop progression. We are insufficient to replace lost cells. Until now cell replenish- left with a pressing need for novel approaches to promote ment (transplant therapy) has been viewed as unlikely to brain repair, and amongst those being pursued are cell re- succeed due to allograft rejection in this sensitized immune plenishment and replacement strategies employing stem environment. However, advances in stem cell give cells. This article will address the potential of stem cell thera- new hope for deriving patient-specific, autologous oligo- peutics for regeneration in the demyelinating disease MS. dendrocytes which may tip the balance to favor repair. The The lay public and legislative politic have become quite challenge will be to engineer these cells to respond to cues engaged in many debates over the promised miracles of that can target their migration into lesions for brain and stem cell regenerative medicine. Issues adqressedalmost spinal cord repair. daily range from theological to budgetary and unfortunately exaggerated claims of research progress. This noisy brouha- Keywords Embryonic stem cell· Migration' ha tends to supplant the focus on largely untested principles Multiple sclerosis· Myelin· · Transplant of the potential for stem cells to fix that which is broken. Politicians speak of cures, patients grasp for incremental benefits, and biologists often seem left wondering whether this might be a repeat of the ruckus over gene therapy and cold fusion. Often lost in the debate is the clinical perspective The inability of our central (CNS) to self of just what it is that stem cells are projected to fix. In repair exacerbates many neuro-degenerative conditions and neuronal degenerative diseases, for example, it is not entirely extracts a tremendous toll on individuals and society. These clear that the therapeutic objective is to replace lost range from those which affect the young including autoim- and their complex interconnections [I, 2]. MS in contrast mune pathology of multiple sclerosis (MS) and traumatic presents a reasonable challenge for proof of principal, since the pathology of the chronic lesion deficit is constant and C. P. Chen' M. E. Kiel . D. Sadowski' R. D. McKinnon well defined [3] and the objectives for repair are clear [4]. Department of Surgery (Neurosurgery), Unlike neuronal degeneration or neurotrauma, stem cell UMDNJ-Robert Wood Johnson Medical School, therapeutics for MS has a defined agenda. 675 Hoes Lane, S-225, Piscataway, NJ 08854, USA

R. D. McKinnon (121) The Cancer Institute of New Jersey, UMDNJ-Robert Wood Johnson Medical School, Piscataway, NJ. USA Oligodendrocytes produce myelin sheaths which insulate e-mail: [email protected] neuronal .MyeJin is critical for fast (saltatory) axonal conduction, for the survival of these axons [5, 6], and to leads to intervention strategies such as grafting exogenous suppress neogenesis and thus quench inappropliate cells for replenishment. In preclinical models, transplanted rewiring of the brain and spinal cord [7, 8]. Decades of stem and progenitor cells which generate oligodendrocytes inquiry into MS have failed to identify cause [9, 10] but promote repair and recovery in myelin mutant rodents [28- succeeded in defining a mechanism [11], its consequences on 32], from demyelinating lesions [33, 34] and after SCI [35- axon degeneration [12] and intervention therapy (interferon- 38]. Stem cells have thus emerged as a great hope for beta inunune suppression). While promising, this is not a therapeutic reconstitution and brain repair, and autologous cure, its efficacy remains controversial [13, 14], and en- histocompatible stem cells are the reagent of choice. dogenous repair is insufficient to overcome the damage [3]. In relapse remitting MS, regions with myelin loss (plaques) are self repaired (shadow plaques) by oligoden- drocytes which are presumed to be generated de novo from an endogenous pool of precursors [15-17], termed 'adult' The inner cell mass of pre-implantation blastocysts contains progenitor cells [18, 19]. While there can be substantial cells which can be c10nally expanded in culture as embry- repair of local demyelination in experimental models [4], onic stem (ES) cells [39-41]. Individually they are pluri- the local pools of these cells are insufficient for significant potential, generating all three germ layer cells in culture repair of MS lesions [20]. Recruitment of more distal and, when injected into a blastocyst, they fully particpate in progenitors into MS lesions appears to fail, as they migrate organogenesis (chimerism) including germ cells to produce to the edge but not into demyelinated plaques [21], and the a monoclonal . ES technology was developed for center of clu'onic lesions are completely devoid of oligo- mouse transgenics and has extended to a number of species dendrocytes [3]. Adult progenitors thus appear designed for including human ES cells [42,43]. If, the logic goes, bone local homeostatic maintenance rather than as a mobile de- marrow stem cells can regenerate the hematopoietic system fense for acute damage control. Thus in addition to immune of a cancer patient, then ES cells hold the promise and suppression, effective MS therapy requires a mechanism to potential to regenerate any organ system for any disease. both supplement the precursor pool and target these cells Fueling the optimism for this strategy are estimates which into a lesion. suggest that restoring only a fraction (10%) of normal The myelin inhibitors that suppress inappropriate axon activity will promote functional recovery for many diseases, sprouting also interfere with repair after traumatic spinal although this number remains an enigmatic estimate. cord injury (SCI). Compression of the cord results in There are limitations to this logic for clinical therapy. severed projection axons and the loss of intemeurons due to First, cell replacement does not address the underlying a breakdown of the blood-brain barrier and toxicity of disease, and for infectious or autoimmune diseases exoge- released and inflammatory cytokines nous grafts may only 'feed the fire' with limited therapeutic [22]. Cellular hypertrophy (the glial scar) then contributes benefit. Second, allograft donor cells are susceptible to to an environment through which damaged axons cannot immune rejection, and both the costs and complications of regrow [8]. A second wave of damage extends beyond this immune suppression overrides potential benefits for chronic wound due to oligodendrocyte death and demyelination of progressive diseases. Third is the pressing concern of ES adjacent, otherwise intact axons. Thus myelin in the acute cell tumorigenicity [44]. Grafted ES cells generate terato- wound prevents axonal regeneration while its subsequent carcinomas [44, 45] and teratomas (embryoid bodies) loss in the sub-acute wound exacerbates the deficits. which in themselves present an ethical dilemma [46]. MS and SCI are good examples of why therapeutic ap- While differentiated progeny of ES cells are not tumori- proaches to CNS repair are in desperate need of a paradigm genic, there is an absolute need to assess the tumorigenic shift. Regenerating tissues such as blood and epithelial potential of a pre-differentiated ES culture in immune- surfaces employ a seed of multipotential cells in an un- compromised pre-clinical models before they can. be detemlined "stem" state for self replenishment [23]. The considered clinically safe. Finally, a fourth problem is the best studied of these are stem cells in bone marrow, and ethics of blastocyst manipulations to generate ES cells. To hematopoietic reconstitution is the gold standard for Stem avoid debates over when a fertilized egg becomes a person Cell Therapeutics [23]. The brain and spinal cord also the field has aggressively explored ethically neutral contain uncommitted stem cells which can generate neurons alternative sources including multipotent 'adult' stem cells. and in vitro [24--27]. While these appear to serve as a After birth sources (umbilical cord, placenta) or amniotic resource to rejuvenate neural cells throughout life, their fluid [47] may provide patient-specific stem cells which apparent inability to repair an acute wound may reflect the could be banked at birth for autologous grafting. Stromal unique architecture of the.brain which, unlike skin or blood, mesenchyme may also provide multipotential cells [48], is perhaps too complex for eXlensive self repair. This then although their apparent neuroectoderm potential is under intense debate [49). Indeed the ability of any genn layer OLs are generated from neuroepithelial stem cells [60, specific 'adult' stem cell to trans-differentiate remains contro- 61] in the gem1inal zone of developing brain and spinal versial [23). A promising alternative to trans-differentiation cord [62, 63). They first emerge as transient amplifYing is the reprogramming of somatic cells by nuclear transfer glioblasts (OPCs) and their specification in vivo, and from [50, 51] or gene conversion [52] to generate 'induced' stem ES cells in vitro, requires sequential instructive cues cells with properties similar to pluripotential ES cells [53, (Fig. 1a). The neural ectodenn is first specified by a set 54]. Thus there remains great hope that once such of cross-regulating inhibitory signaling pathways during approaches are feasible the ethics of blastocyst manipula- gastrulation. Bone morphogenic protein (BMP) promotes tions will be moot, the limitations of allografts eliminated, epithelial differentiation from ectodenn via inhibitory (ld) and clinical stem cell therapeutics using autologous cells can regulators that block 'default' neurogenesis [64, 65). be programmatic. There then will remain the challenge to Instructive cues including fibroblast growth factor (FGF) program the differentiation of stem cells to efficiently then promote neuroepithelial differentiation by inducing generate tissue-specific cells for repair. BMP antagonists (Noggin, Chordin, Follistatin) from Hensen's node (Spemanns' organizer in amphibians) to locally block BMP [66). Dorsal-ventral specification cues including Sonic hedgehog (Shh) then instruct neural lineage detennination [67, 68). In the embryonic spinal cord OPCs A prerequisite for ES cell therapeutics is to instruct lineage are first generated in the ventral lateral domain pMN of the differentiation prior to grafting. For ES-derived oligoden- neural ectodenn [69-71). This domain also generates motor drocytes (OLs) this involves the sequential presentation of neurons, and both lineages are specified by the ventralizing signaling cues which direct ES to OL development in vivo effects of Shh [72] via the basic helix-loop-helix factor (Fig. I). Since mature OLs are post-migratory, the focal cell Olig2 [73-75). for grafting is their immediate progenitor the oligodendro- The inhibition ofBMP signaling is also important for the cyte precursor cell (OPC) (Fig. Ib). OPCs migrate through differentiation of ES cells into neural stem cells in vitro the complex architecture of the brain during early develop- [76). Mouse ES cells self renew in the presence ofleukemia ment, and when grafted into a neonatal recipient they can inhibitory factor (LlF) and BMP or serum factors [64, 65). both migrate and differentiate into OLs [55, 56). Whether Withdrawing these in non-adherent conditions promotes the the adult brain can also provide appropriate signals for the aggregation of disorganized embryonic bodies with a migration and maturation of ES-derived OPCs is largely un- mixture of differentiated cell types [77], and in adherent explored. Thus the cues involved in developmental myelina- cultures generates a less complex mixture of largely neural tion (Fig. 1) are prime candidates for manipulating the adult progenitors. Protocols have been developed for the sequen- CNS to favor ES-derived myelin regeneration and repair. tial instruction of ES cells into neural precursors [78] then

PDGF FGF

~0.00 blastocyst ~ ·~GF~~~'k:

=:~~5=_::,,~::..':;:~::':::;~:·:.=::~=:':!··OC··"'G"iC,'CNP .,..,.', = ".."~"' eIlGER..NG2 , • VimentliF,GD3

04 Fig. 1 oL development. a OPC specification; embryonic stem (ES) this lineage is shown below. Monoclonal A2B5 detect gangliosides cells self-renew in leukemia inhibitory factor plus BMP or sera and [57]; 04 sulfatide and glycolipids [58], and 01 galactocerebroside sequentially generate EB (embryoid bodies) NS (neural stem) cells and (GalC) [59]. Other markers include platelet-derived growth factor 0(. oPCs via factors that first promote neural induction (FGF induced receptor (PDGFR), proteoglycan NG2, ganglioside GD3, cyclic BMP antagonists) then NT (neural tube) ventralization (Shh). b OPC nucleotide phosphohydrolase (eNP), myelin associated glycoprotein maturation in vitro, depicting the sequential emergence of antigens in (MAG), myelin oligodendrocyte glycoprotein (MOG), myelin basic migratolY OPCs and postmitotic Ols. l'vlaturation is driven by PDGF protein (MBP) and proteolipid protein (PLP) and inhibited by FGF. The sequential emergence of antigens marking into glia [32, 61, 79, 80]. First, ES cells in monolayer induction and patterning [66, 67, 101]. However the full culture or dissociated embryoid bodies are cultured in repertoire of OPC fate potential in vitro is not evident in serum-free conditions with retinoic acid (RA) [81], which vivo, where their actual fates may be more restricted [102]. interacts with FGF and Shh to pattern homeodomain and Thus it is important to determine whether in vitro derived bHLH factor expression in the ventral spinal cord [82]. ES progeny generated via distinct methods represent stable Neural differentiation is induced by autocrine signals such phenotypes with therapeutic benefit. as FGF [83] and is enhanced by activated Notch [84]. Second, neural stem cells are amplified by mitogens including EGF [85] and FGF [86]. Third, gliogenesis is instructed by the ventralizing factor Shh [79, 87, 88]. Finally, the cultures are switched into media containing Pre-clinical studies on transplantation to rescue myelin PDGF to promote oPC proliferation and survival [89, 90] pathology started with the work of Madeline Gumpel and and FGF to block terminal differentiation [91]. In addition colleagues some 25 years ago [103, 104]. Early studies to growth factors for induction and selection, genetic used neural tissue grafts including CNS fragments or oLs modifications have been devised to select OPCs and isolated fi'om newborns rodents [105-107]. These and eliminate other cells in order to generate highly purified subsequent studies demonstrated myelin sheath formation OPC populations from· ES cells [79, 80]. These cultures and the potential of exogenous tissue for remyelination in thus approach the appropriate reagent for therapeutic model systems including genetic dysmyelination (Shiverer intervention in demyelinating diseases. mice, myelin deficient rats) and in experimental demyelin- The current explosion of studies on embryonic and adult ation induced by chemicals (cuprizone, ethidium bromide), stem cells in vitro is reminiscent of the expansion of tumor immune-mediated (expelimental allergic encephalomyeli- virology in the 1980's and neurobiology in the 1990's. tis), after viral attack (mouse hepatitis virus), and after Lessons learned from these prior studies are relevant to traumatic CNS injury [34, 36, 37, 56, 108-111]. interpretations of stem cell fates in vitro and their therapeutic Studies to date have examined grafts of mitotic, mobile potential. General precautions associated with manipulating progenitor cells including oPCs isolated from neonatal rat cells in vitro include the pitfalls of cell line cross contami- brain [33, 55], an immortal rat OPC line CG4 [112], oPCs nation [92] and the concern that cells in culture can undergo from human embryonic brain [113] and ES-derived oPCs alterations that can lead to malignant transformation. fi'om both mouse [32, 35] and human [36]. The ability of Unique concerns for stem cells include misinterpreting the oPCs to both proliferate and migrate is key to their efficacy stability of chemical induced phenotypes in vitro [49] and [29]. While it is impractical to consider xenograft neural in vivo artifacts due to cell fusion [93]. Thus the stability of tissue as a resource for clinical transplants, stem cells now ES-derived OPCs is an important issue. oLs in vitro can provide new prospects for generating autologous oLs for revert to 04 progenitors [94], from 04 to o2A [95, 96], remyelination. The potential of in vitro differentiated ES and from o2A into neural stem cells [97]. oPCs can also progeny still requires parallel comparisons of the efficacy differentiate into other glial phenotypes in vitro [98, 99]. of distinct ES-derived cell populations to generate oLs, Such plasticity underscores the potential of culture con- manipulations to guarantee their clinical safety, and studies ditions to influence fate [100]. Plasticity is not simply an on how to manipulate the signaling environment in the artifact of culture, as neural fate can also be reprogrammed adult brain to recapitulate the cues which direct oPC in vivo by surgical reposition at many stages of neural migration and oL maturation in development.

C -25 ,-L -L -20 U QJ -15 p<.OOI ~ ---.I.- Q) E -10 i= slow fast - 5 (.64) (.36)

Shi ShiES ShiES Fig. 2 ES-to-OL in Shiverer chimeras. a Adult Shil+ (normal) and and exit time recorded. Shi mutants lack balance and coordination, Shi/Shi (mutant) mice. b confocal ofES-derived MBP+ OLs in Shi-ES while 36% of Shi-ES chimeras showed improved locomotor function chimeric mouse spinal cord. ShiiShi mutants are devoid of MBP" relative to age matched controls (p