Abstract
We here propose an updated concept of stem cells (SCs), with an emphasis on neural stem cells (NSCs). The conventional view, which has touched principally on the essential property of lineage multipotency (e.g., the ability of NSCs to differentiate into all neural cells), should be broadened to include the emerging recognition of biofunctional multipotency of SCs to mediate systemic homeostasis, evidenced in NSCs in particular by the secretion of neurotrophic factors. Under this new conceptual context and taking the NSC as a leading example, one may begin to appreciate and seek the “logic” behind the wide range of molecular tactics the NSC appears to serve at successive developmental stages as it integrates into and prepares, modifies, and guides the surrounding CNS micro- and macro-environment towards the formation and self-maintenance of a functioning adult nervous system. We suggest that embracing this view of the “multipotency” of the SCs is pivotal for correctly, efficiently, and optimally exploiting stem cell biology for therapeutic applications, including reconstitution of a dysfunctional CNS.
Keywords: Stem cells, Neural stem cells, Multipotency, Neurotrophic factors, Neural Repair, Spinal Cord Injury.
BACKGROUND
Along with refinement of our understanding on the biology and translational potential of neural stem cells (NSCs) [1-6], there has been an increasingly appreciated paradigm shift regarding how the adult mammalian central nervous system (CNS) could be repaired for functional restoration. The long-standing dogma that the adult CNS was immutable to anatomical and functional repair was challenged by an unambiguous demonstration that cells derived from the CNS from a variety of structures and at different developmental stages, including adulthood, possess stem-like properties. NSCs are primordial and uncommitted cells that have been believed to give rise to the vast array of more specialized cells of the CNS. They are operationally defined by their abilities (1) to differentiate into cells of all neural lineages (i.e. neurons of multiple subtypes, oligodendroglia, and astroglia) in multiple regional and developmental contexts (i.e., to be multipotent); (2) to self-renew (i.e., to give rise also to new NSCs with similar potential); (3) to migrate and populate developing and/or degenerating CNS regions; and (4) to have biofunctional multipotency to mediate systemic homeostasis through capacities such as production of trophic factors, formation of gap junctions, etc. [7,8]. Since monoclonal derivation of progeny is obligatory to the definition of NSC: that is, a single cell must possess all the aforementioned attributes, which suggests the ready availability of NSCs, many studies, including ours, in the past two decades, provided hope that the use of NSCs might circumvent some limitations of presently available graft material and gene transfer vehicles and make feasible a broader range of therapeutic strategies. This significant advance has led neurobiologists to speculate how such phenomena might be further harnessed for both therapeutic advantage and for better understanding of developmental neurobiology mechanisms.
Most studies, to date, are focused principally on exploring the biologically most apparent features of NSCs in comparison to existing gene therapy and cell transplantation strategies. For this purpose, NSCs, with their homogeneous and well-defined neural differentiation capacity, have been pursued primarily as a modality that could replace dead or degenerating neural cells in a wide variety of neurological diseases and trauma. For certain pathological models of the brain, NSCs and their progeny not only could integrate with host tissue at their site of implantation [9-11], but also could interact with distant brain tissue via migration in response to chemical cues in the host [12-15]. The responsiveness of NSCs to microenvironmental cues in the surrounding CNS tissue provides them with a unique trait distinguishing them from fetal brain/spinal cord tissue and non-neural cells (e.g., fibroblasts or mesenchymal stromal stem cells [MSCs]), as well as from most viral vectors and protein infusion devices for gene delivery. For example, haematopoietic cells could not efficiently cross the blood–brain barrier and integrate throughout the CNS as effectively as NSCs. In addition, NSCs are able to “sense” the presence of degenerating neural tissue [13,14,16-18]. Conversely, effective replacement of dying or dysfunctional neurons by NSCs in the adult CNS, an ideal hypothesis the field initially entertained, has been proven to be a much more challenging endeavor, though in a few experimental models of neurodegeneration such NSC-based cell substitution appeared to be feasible in replacing dead oligodendrocytes [9,19]. We therefore have discussed here the biological perspectives of epigenetic and genetic features of rodent (in particular, murine) and human neural stem cells regarding their capacity to produce neurotrophic factors (NTFs). Also included are views of investigators in the field on how to modulate and/or augment the homeostatic function of NSCs. Finally we summarize our current thoughts about the critical roles that NSCs may play in designs for improved regimens of NTF delivery regarding neural repair and pathology correction.
NEUROTROPHIC FACTORS AND NEURAL STEM CELLS
Because NSCs are dynamic developmental entities their therapeutic potential has been studied primarily via strategies devised to realize desirable neural phenotypic differentiations. Much less recognized and appreciated is the fact that these developmental cells, by innate biology, produce neurotrophic factors that influence the growth and well-being of their own and surrounding tissues via autocrine and paracrine mechanisms. Efforts to understand this more recently recognized perspective of stem cell biology were first triggered by the observation that undifferentiated donor NSCs could exert marked neuroprotection to the spinal cord following traumatic injury, engendering a speculation that NSCs and other stem cells may have inherent capability to synthesize trophic factors [20]. Considerable research endeavors have since taken place to seek beneficial impact of stem cells (e.g., NSCs, MSCs, and embryonic stem cells [ESCs]) that are mediated mainly through soluble trophic factors and other cytokines that enable the body to reestablish homeostasis after pathologic and traumatic insults, inflammation, and tissue infarction or degeneration [21-24]. In this article, based on data mainly derived from studies on traumatic spinal cord injury (SCI), we seek to establish conceptually the biological principles of trophic factor delivery by stem cells as a novel approach to new therapeutic strategies based on developmental mechanisms [7]. For this goal, we have expanded the conceptual scope of tropic factors to include the following categories of molecules.
By our definition, neurotrophic factors, here including classic neurotrophins, are groups of naturally occurring substances that support neural cell survival, proliferation, migration, differentiation, growth, and function [25]. They are usually proteins or simpler peptides, and are essential regulating and signaling molecules for cell development and function. Neurotrophic factors contribute to neurogenesis, and are critical in shaping neural network structure and physiological processes such as learning, memory formation, and sensorimotor function [26]. Some neurotrophic factors, conversely, can be directly involved in pathological processes [25], via effects on synaptic biology, dynamics of neurotransmitter release and synaptic efficacy, whereas others act through secondary messengers and various kinases [27].
Listed below are the six biological families of neurotrophic factors that directly impact stem cell development, and are expressed by natural or bioengineered stem cells. 1. Classic neurotrophins that comprise nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), and neurotrophin-4 (NT-4); 2. The transforming growth factor (TGF)-beta family, represented by glial cell line-derived neurotrophic factor (GDNF) and the bone morphogenic proteins (BMPs); other members are neurturin, artemin, and persephin; 3. The cytokine growth factor family, including ciliary neurotrophic factor (CNTF), leukemia inhibitory factor (LIF), and cardiotropin-1; 4. The epidermal growth factor (EGF) family, consisting of EGF, transforming growth factor-alpha (TGF-alpha), the neuregulins, and neural and thymus-derived activator for ErbB kinases (NTAK); 5. The insulin-like growth factors (IGFs), consisting of insulin-like growth factor I (IGF-I) and IGF-II; and 6. The fibroblast growth factor (FGF) family, consisting of at least 24 different proteins including acidic FGF (FGF 1) and basic FGF (FGF 2) [28,29].
For the purpose of this review, we focus our discussion only on the family of classic neurotrophins since all its members (i.e., NGF, BDNF, NT-3, and NT4/5) have been demonstrated to play critical roles in neuronal survival, differentiation, and/or function in neurotrauma settings [30]. The neurotrophins act by binding to receptor tyrosine kinases of the Trk families. NGF binds for tropomyosin-receptor-kinase-A (TrkA), BDNF and NT-4/5 bind TrkB, and NT-3 preferentially binds TrkC. Importantly, NT-3 up-regulates TrkC expression in NSCs and induces them to differentiate into neurons [31-33]. For example, Ad-TrkC can be constructed to express TrkC effectively in NSCs, enhancing NT-3-mediated neuronal differentiation, an approach that may offer additional strategies for treating neurotrauma [33]. Overexpression of TrkC in rat neural progenitor cells improves survival and reduces glial differentiation of donor cells in the intact spinal cord [34]. Neurotrophin-mediated Trk signaling plays an essential, cell-autonomous role in regulating the proliferation and differentiation of embryonic cortical precursors and thus controls cortical development at early stages [35]. Multiple alternatively spliced isoforms have been observed for TrkA, TrkB, and TrkC, especially in non-neuronal cells [31,36-38]. Some of these isoforms lack the cytoplasmic tyrosine kinase domain, but retain selective signaling and may inhibit neurite outgrowth [37,39-41]. However, brain-derived neurotrophic factor (BDNF) has proliferative effects on NSCs through the truncated TRK-B receptor [42]. Neurotrophins also bind to p75NTR. Activation of this receptor may cause cell death rather than survival, as p75NTR-/- mice show reductions in neuronal cell death after pilocarpine-induced seizures compared to wild-type [43,44]. Interestingly, the p75 neurotrophin receptor is also involved in proliferation of undifferentiated mouse ESCs, and becomes down-regulated upon ES cell differentiation [45]. NT-3 improves the neural differentiation of skin-derived precursors (SKPs) induced by retinoic acid (RA) through a p75NTR-dependent signaling pathway [46,47]. In the following sections, we will discuss how a novel concept of using prototype or engineered NSCs to deliver therapeutic neurotrophic factors was developed, based on data collected from SCI investigations, and how this concept has subsequently been used to guide administration of neurotrophic factors to treat or investigate representative neurological disorders such as SCI, Alzheimer's disease (AD) and amyotrophic lateral sclerosis (ALS).
ADMINISTRATION OF NEUROTROPHIC FACTORS FOR TREATING SCI
The pharmacological use of neurotrophic factors for SCI treatment has been tested primarily with two neurotrophins: BDNF and NT-3. Unlike peripheral nerve axons, which regenerate well, the adult mammalian spinal cord appears to have an unsuccessful capacity for axonal regrowth [48,49]. Neurotrophic factors, when administered in appropriate physical and temporal gradients, have been shown to support axonal growth after peripheral nerve injury. In contrast, antagonizing these growth factors in the extracellular matrix impedes neurite outgrowth [50,51]. Due to these findings, plus the fact that molecules have been identified that can suppress axonal sprouting and regeneration in the CNS (e.g., the chondroitin sulfate proteoglycan [CSPG] molecules) [52,53], substantial experimental efforts have been given to enhance regeneration of the supraspinal descending axons through activating neurotrophic factor-mediated mechanisms that may overcome the growth inhibitory environment in the CNS [54-60]. Additional approaches include combinatorial tactics that comprise transplantation of progenitor cells into the lesion cavity with simultaneous administration of neurotrophic factors [61], or direct augmentation of key molecules of trophic factor-triggered signal transduction, such as raising intracellular cAMP levels pharmacologically [62,63]. Many of these studies report incremental improvement in axonal growth and/or motor behavior, projecting hope for advancing towards the ultimate goal of restoring function following clinical SCI.
As examples, several groups reported that BDNF can enhance growth of supraspinal motor axons into permissive growth milieus placed at sites of SCI. Both rubrospinal axons [64,65] and reticulospinal axons [66], descending networks that regulate spinal cord motor neural activities, regenerate in response to BDNF stimulation. Reversal of atrophy of neurons in the red nucleus, even one year after SCI, can occur after BDNF stimulation [67]. However, corticospinal tract (CST) axons did not respond to BDNF post-SCI, even though BDNF prevents the death of corticospinal neuronal cell bodies when directly applied to the cerebral cortex. This outcome has been attributed to the lack of BDNF receptor TrkB on CST fibers [68].
The ability of NT-3 treatment to promote regeneration of severed CST axons was also reported [69]. This triggered additional bioengineering initiatives to enhance the delivery efficiency of NT3. Subsequently, Grill and colleagues, adapting a gene therapy strategy, demonstrated that autologous fibroblasts genetically modified to secrete NT-3 could be transplanted into the epicenter of experimental SCI to improve neural repair [70]. More recently, Tuszynski’s group at UCSD reported beneficial effects of artificially established NT-3 gradients using cell-based gene delivery, increasing axonal regeneration in experimental SCI [71]. Importantly, studies comparing the therapeutic effect on axonal growth of three different methods of replenishing neurotrophic factors (direct infusion, transplantation of unmodified cells, and transplantation of genetically engineered cells with enhanced secretory function) have reported favorable results with genetically modified cells [72]. These data support the notion that using cells to administer neurotrophic factors may offer clinically favorable pharmacodynamic properties due to the interactive capability of “these functional basic units of life” [71,72]. Therefore, donor cells such as NSCs, in particular, have become highly promising candidates for delivering NTFs due to their innate biology, which regulates molecule secretion in response to environmental changes in the nervous system [7].
This viewpoint is further strengthened by observation of a therapeutic impact of endogenous NSCs on experimental SCI under the enhanced physical activity paradigm of rehabilitation that induced functional recovery. In some cases, there was a positive correlation between degrees of functional improvement and the number of nestin-expressing NSCs present in the post-lesion spinal cord [73-76], suggesting that NSC proliferation enhanced by exercise may help augment NTF production (see below for more details). Additional support comes from the facts that severe side effects of neurotrophic factors could be caused by conventional drug delivery regimens that lack biological feedback regulations, and by barriers imposed by the peptidergic structure of these trophic factors, which impair their penetration into the brain or spinal cord parenchyma, and therefore make their pharmaco-therapeutic properties difficult to evaluate [77].
A NOVEL CONCEPT OF USING PROTOTYPE OR ENGINEERED NSCS TO DELIVER NEUROTROPHIC FACTORS: INSIGHTS FROM STUDIES OF ENDOGENOUS NSCS AND ACTIVITY-DEPENDENT RECOVERY FOLLOWING SCI
Endogenous NSCs in the spinal cord and their response to trauma: Mitotic activity has been known to persist in the adult mammalian spinal cord [78]. Weiss and colleagues found that the lumbosacral spinal cord normally produces the greatest number of multipotent cells, and the cervical cord the least [79]. Under most conditions, adult NSCs have astroglial characteristics and express glia fibrillary-associated protein (GFAP) [78], but retain the ability to undergo neurogenesis [80,81], and can give rise to functional neurons and glia in vitro and in vivo [82]. It has been shown that neurotrauma significantly increases the proliferative capacity of spinal cord NSCs. Ependymal zone cells that express low levels of nestin were found to be upregulated, increasing mitotic activity and generating glia, shortly after an injury to the dorsal funiculus of the spinal cord. If implanted in a pro-neurogenesis environment [74,75,83], they are also capable of migrating and undergoing neurogenesis [81,83,84,85].
Ependymal and parenchymal neural progenitors were also reported to be activated in response to spinal cord transaction [86,87]. When isolated in vitro, they generate astrocytes and oligodendrocytes but few neurons. A fraction of ependymal zone cells possessing stem cell properties or oligodendrocyte progenitor cells in the parenchyma proliferate following compression and contusion injuries [84,88], and appeared to participate in repopulation of the injured areas, suggesting their potential participation in lesion repair [89,90]. This response of NSCs to the injury occurs rather rapidly; for instance, nestin was seen in ependymal cells as early as 1 day after minimal sharp instrument injury to the lateral columns [91]. This injury, however, did not supply a sufficient stimulus for ependymal zone cells to migrate to the lesion site. A relatively larger injury to the dorsal funiculus, by contrast, led to ependymal zone NSC migration from the central canal to the lesion site [82].
Since brain NSCs have been reported to respond to external stimuli such as enhanced physical activity and exercises, by increasing their proliferation, differentiation and survival [92], an extension of that rationale is to ascertain whether physical activity and exercise could serve as an independent stimulus for endogenous NSC proliferation also in the adult spinal cord. We earlier hypothesized that a major function of proliferating NSCs in the adult mammalian CNS is the production of trophic factors and other homeostasis-maintaining molecules that are essential for endogenous healing and plasticity processes in the spinal cord [93]. Our published data suggest that physical activity may be beneficial partially by increasing proliferation of endogenous NSCs that produce neurotrophic factors such as GDNF and BDNF, which in turn mediate neuroplasticity and improve sensorimotor functions [74]. Conversely, the contribution of neurotrophic factors to exercise-induced functional recovery per se has been investigated extensively. Voluntary exercise induces an up-regulation of BDNF and NT3 mRNA and protein levels in the spinal cord [94]. Increased levels of BDNF-associated mRNA and protein levels near the injury site have also been reported following treadmill training or running [94]. Additionally, direct stimulation of hindlimb stepping was noted after intrathecal administration of BDNF [95]. Other factors related to BDNF activity, such as synapsin 1, NT-3 and cyclic AMP (cAMP) response element binding protein (CREB), are also elevated in response to post-injury exercise [96].
Although injury alone may induce an increase in the levels of these neurotrophic agents [97], these astrocyte-based increases were transient (< 2 days after SCI), and thus, unlikely to facilitate later phase neuroplasticity or functional recovery. In contrast, animals that were exercised in the post-injury period demonstrated prolonged elevation of these factors, which may offer a new therapeutic window for other interventions. There is currently no agreed-upon conclusion regarding the origin of exercise-enhanced BDNF expression despite numerous studies that implemented NSCs, astrocytes, neurons and blood-born cells as neurotrophin providers under varied situations [98]. Finally, the spinal cord's inherent regenerative potentials can also be directly stimulated by post-injury physical activity. Exercise has been shown to double the number of proliferative cells in the CNS [74,99] and to have additional benefits, such as enhanced long-term potentiation of post-synaptic efficiency [100].
Overall, experimental outcomes suggest that synapse formation and maintenance can be activity-dependent (e.g., exercise-driven), largely operated by neurotrophins [101]. Such neurotrophic drives may not only promote birth of new neurons, but also facilitate oligodendrogenesis [102]. We therefore speculate that the amplitude of NSC-based release of neurotrophic factors could be further modified by physical activity-related rehabilitation modalities and specific pharmacological treatments augmenting NSC participation and their trophic factor production that, by mimicking developmental processes, augment reciprocal therapeutic relationships with surrounding cells and cue molecules [7].
MECHANISMS THAT ENABLE THERAPEUTIC DELIVERY OF NEUROTROPHIC FACTORS FROM PROTOTYPE OR GENETICALLY ENGINEERED NSCS FOR SPINAL CORD REPAIR
It has been well recognized that NSCs have an innate ability to detect pathologic targets for trophic actions. We and other investigators previously described a critical feature of prototype NSCs that is distinct from non-NSC carriers of neurotrophin transgenes – the ability to detect pathological loci and adopt appropriate developmental initiatives toward customized trophic support or cell replacement [103,104]. These abilities were first suggested by experimental data that was derived from a clone of murine NSCs after their implantation into an adult rat spinal cord after segmental hemisection (i.e., open wound lesion), where the NSCs contibuted markedly to functional restoration [20]. Moreover, when NSCs of the same lineage were transplanted into adult mouse neocortex in which pyramidal neurons of a circumscribed region were induced experimentally to undergo apoptosis, they differentiated differently than when grafted into uninjured neocortex: they preferentially differentiated into pyramidal neurons, whereas these same NSCs yielded mostly glia in normally developed neocortex [103]. These data indicate that donor NSCs can respond to biological cues in normal and abnormal microenvironments, suggesting that NSCs spontaneously have mechanisms compensating for genetic [17,105] or acquired deficiencies [14]. Though the exact mechanisms regulating the capacity of NSCs to exert developmental functions in an adult microenvironment are still unknown, it is clear that when confronted with injury or other pathologic conditions, NSCs recapitulate certain of their developmental mechanisms [24,93,103].
It is currently believed that the signals modifying exogenous and endogenous NSC production of trophic factors affect a complex group of mitogens and chemokines, trophic and tropic agents, plus adhesion and extracellular matrix molecules (i.e., common developmental signaling molecules), as well as chemotactic and angiogenic factors, activated microglia, inflammatory cells, invading macrophages and damaged neurons and glia (i.e., general inflammation-mediating components). Optimization of therapy for tissue protection and repair in the post-mitotic environment of the adult CNS may require overcoming an insufficient supply of NSCs and endogenous neurotrophic factors. Hence, donor NSCs become attractive to consider as therapeutic supplements. However, exogenous prototype NSCs without engineered enhancements are vulnerable to same insults from secondary injury molecules as endogenous cells [20,106]. Therefore, we and our colleagues at the Langer Lab at MIT recently devised a novel chemical engineering approach to protect donor NSCs by applying antioxidant drug-releasing polymer film to alter the implantation milieu against neuroinflammation mediated by reactive nitrogen species (RNS). Our technology has been fruitful not only in enhancing the survival of exogenous NSCs but also in maintaining their capacity to produce neurotrophic factors such as GDNF and BDNF [107].
APPLICATION OF NTFS OR NSCS TO TREAT NEURODEGENERATIVE DISEASES
As a consideration of translational initiatives beyond SCI, the following sections will briefly discuss the possibility of using NTFs and NSCs in treatment of neurodegenerative diseases, citing studies on Alzheimer’s Disease (AD) and amyotrophic lateral sclerosis (ALS) as representative examples.
The potential use of NGF, a member of the neurotrophin family, for AD therapy has been reviewed in detail [29]. NGF emerged as a candidate therapeutic molecule for neuronal degeneration soon after its discovery [111], due to its role as a vital survival factor for sensory and sympathetic neurons during development [112]. NGF also prevents both lesion-induced and spontaneous atrophy of basal forebrain cholinergic neurons, as well as ameliorating memory deficits in aged rats; both are hallmark therapeutic targets for AD [113-115]. In AD animal models NGF treatment reduced neuronal death, and simultaneously stimulated cholinergic neurons, despite exposure to a cohort of deleterious mechanisms that cause neuronal degeneration, including excitotoxicity, aging, and amyloid overproduction [116-122]. NGF treatment improved cognition in a variety of models of rodent memory dysfunction, and its neuroprotective actions have been also confirmed in non-human primate models [123-128]. Mechanistically, NGF activates Erk/MAP kinase to influence a number of growth and function-related intracellular pathways via TrkA receptors, and activates pro-survival gene expression and inhibits apoptosis via the Akt pathways [127]. The studies to date validate the essential trophic impact of NGF on adult cholinergic neurons, and corroborate that NGF is needed (and produced) throughout life in the neocortex and hippocampus [128,129].
Therefore, if the expressions of p75 and TrkA receptors remain sufficient for NGF responsiveness, NGF delivery near degenerating cholinergic neurons appears to be an appealing clinical strategy [130-133]. However, effectively delivering NGF to the brain in humans remains a challenge, primarily because the NGF protein is too large to cross the blood brain barrier (BBB). Intra-ventricular infusion can bypass the BBB in animal models, but then, NGF spreads non-selectively in the CNS, resulting in unwanted side effects such as weight loss [134], sympathetic axon sprouting [135] and neuropathic pain related to Schwann cell invasion [136,137], despite effectiveness in the rescue of degenerating cholinergic neurons. The adverse effects were also observed in human trials [138]. Similar effects occurred in trials of patients with Parkinson’s disease when GDNF or BDNF were infused intraventricularly [139,140]. These side effects observed in clinical trials highlight the necessity to establish new approaches that can overcome certain ineffective aspects of direct NTF delivery that lacks feedback regulatory mechanisms.
ALS is a fatal neurodegenerative disease with a signature pathology of progressive motor neuron death in both the spinal cord and brain, which culminates in rapid loss of muscle function and eventual respiratory failure [141,142]. The only clinically available therapy to date is riluzole (Rilutek), which marginally extends survival by limiting excitotoxicity [143] and increasing neurotrophin release from astrocytes [144]. The majority of ALS cases are sporadic, whereas only 10% are familial (FALS) [145], among which 15-20% can be traced to point mutations in cytosolic Cu2+/Zn2+ superoxide dismutase 1 (SOD1) [146].
The rapid neuronal degeneration in ALS deems neuronal replacement and neuroprotection as valuable clinical approaches to treatment. Therefore, different types of progenitor or stem cells with capacity to produce NTFs have been tested in rodent models of ALS [147-151]; these include hNT neurons derived from the human teratocarcinoma cell line, mouse Sertoli cells [147,149], human umbilical cord blood cells [147,151], human [151], or mouse [148] bone marrow transplants, [152] and mouse or human NSCs [153,154].
Data in published studies suggests, at first glance, that the lineage or site of origin of NSCs may be responsible for some of the drastic differences in therapeutic efficacy. For instance, transplantation of human NSCs isolated from fetal forebrain (i.e., HFB2050) [14] into multiple loci of the spinal cord resulted in marked increase of life span in SOD1 (G93A) mice [154], whereas neural progenitor cells (NPCs) derived from human cortex and spinal cord produced lesser results [155]. However, further analysis suggests that the NTF profiles of donor cells might have played a key role determining the therapeutic effect in rodent ALS models [154,156]. Prototype human NSCs (HFB2050) secrete a spectrum of NTFs, including BDNF and GDNF [14], whereas genetically engineered NPCs may produce high levels of one specific NTF only, which may not be adequate to counteract ALS pathophysiology that is triggered by multiple pathogenic pathways [141]. Likewise, direct expression of GDNF delivered by lentiviral vector transfection to the lumbar spinal cord showed no beneficial effects on motor neuron survival [157], nor did robust GDNF production in the spinal cord by genetically modified NPCs prevent muscle atrophy in SOD1 rats [158]. These results underscore the need to define the range of signaling mechanisms governing the interaction between stem cells and the surrounding microenvironment to reach homeostasis by regulative mechanisms such as NTFs, other secreted molecules, and direct cell-to-cell communication (e.g., via gap junctions). Better understanding of these processes will ultimately enable us to design combinatorial approaches with stem cell-mediated NTF therapy to tackle complex systemic pathologies such as ALS.
To the best of our knowledge, the only controlled ALS clinical trial with stem cells reported so far was performed in Italy [159], though there had been a few uncontrolled clinical studies with other growth factors, including peripheral injection, intraventricular infusion, or intrathecal injection of CNTF and BDNF [160,161]. In the phase 1 trial the effects of autologous bone marrow derived mesenchymal stem cells (MSCs) were transplanted safely in the thoracic spinal cord of 10 ALS patients [159], and no adverse effects were seen in the 24-month post-transplantation observation period. However, no significant changes in disease progression were observed. Based on recent findings in experimental animals, glial pathology in ALS should targeted in future trials [162]. For example, ALS model mice with conditionally deleted astrocytes expressing mutant hSOD1 showed delayed disease progression [163]. By introduction of stem cells with diversified capacities to express NTFs, such as GDNF, BDNF, IGF-I or VEGF, as well as forming direct cell-to-cell communications (e.g., via gap junctions) [8], motor neuron survival and function may be improved through modificaion of the tissue microenvironment [109,154].
FUTURE DIRECTIONS
Engineering NSCs as Transgene Carriers or Biopumps
It has been well established that NSCs constitutively produce a broad range of appropriately functioning peptidergic neurotrophic and neurite outgrowth-promoting factors [104]. To augment such capabilities further, NSCs have been engineered to express various transgenes, including NT-4/5, GDNF, BDNF, NGF, L1, sonic hedgehog, wnt-1, wnt-3a, as well as an assortment of biosynthetic and metabolic enzymes. Customized implantation of genetically engineered NSCs has been used to enhance neuronal differentiation, neurite outgrowth and connectivity within diseased tissue loci [108,109]. Also, NSCs have been manipulated to express the specific TrkC receptor of a neuron-inducing neurotrophin, NT-3 [33,77]. The engineered NSCs respond to NT-3 in an autocrine or paracrine fashion [33], which appears to trigger a significantly higher percentage of differentiating neurons in the TrkC-expressing NSCs group exposed to NT-3 than in the rAd-LacZ control cell group similarly exposed to NT-3 [33].
However, attempting to intervene in the natural expression of the various neurotrophic factors in their various proportions through genetic manipulation actually appeared to cause profile changes in trophic factor expression by the engineered cells [110]. For example, enhancing expression of NT-3 in a given clone of NSCs actually extinguished the clone’s expression of GDNF, replacing the promotion of motor axon ingrowth with an enhanced ingrowth of sensory axons [110]. Caution is called for when testing a therapeutic strategy based on transgene-mediated NTF overexpression in NSCs, for unintended effects may result from such (unanticipated) “disturbance” in post-engineering biological programs of the donor stem cells. The aforementioned facts, overall, suggest the feasibility of using bona fide NSCs or genetically engineered NSCs to serve as biopumps for a broad selection of biofactors when one is aiming to repair damaged or dysfunctional neural tissues.
Induced Pluripotent Stem Cells (iPS Cells)
The past few years have seen the swift development in pluripotent stem cell technology led by the initiating report from the Yamanaka laboratory [165], in which a group of stemness- or mitosis-related transcription factor genes (Oct4, Sox2, Klf4 and c-Myc) were introduced to transform somatic cells dramatically into stem cell-like primitive entities with pluripotency and differentiation flexibility that are, to certain degrees, similar to ESCs. However, there are serious concerns regarding the safety of such iPSCs, especially the tumorigenic consequence, which currently prevents practical clinical use of iPS cells. Significant intrinsic variability in the derived iPSCs, abnormal expression of imprinted genes due to the random integration of transcription factors, persistent donor gene expression, and difficulty in obtaining a sufficiently rapid cell multiplication rate in vitro to build up a cell population sufficient for use in human patients further limit the clinical use of these cells [165-167]. Therefore, additional molecular strategies using alterative gene targets [168,169], fewer targets [170-173], or viral vector-free transfection technologies have been developed [175-178], to circumvent the safety deficits of iPS cells before their therapeutic potential can be systematically examined. However, recent reports showed that patient-specific iPSCs can be generated for investigative studies on drug screening or for experimental therapy trials [179-181]. In addition, direct trans-differentiation of adult cell types has been presented as a further therapeutic option for reversing disease progression or transforming neighboring healthy cells to take over functions lost by diseased or dying cells [182]. Despite these advances, there appear to be no reports to date on NTF production by iPS cells, a pivotal parameter of stem cell biology [7], albeit there is recent important progress in the use of synthetic modified mRNA to increase efficiency of cell reprogramming and directed differentiation, as well as analyses of the impact of epigenetic memory on the molecular and functional properties of iPS cells [183-186].
A SUMMARY NOTE
Academia and the public have, in general, accepted the concept that stem cells provide an enormous opportunity to advance the understanding of developmental biology, and especially to develop novel clinical therapies. However, we still need refined knowledge on how NSCs can be crafted to deliver the range of NTFs that would meet the micro-environmental requirements to establish homeostasis that would offset an ongoing disease process. This ability of stem cells, buoyed by their innate developmental biological properties, may teach us how to mitigate the adverse side effects resulting from direct administration of NTFs one at a time. Complex diseases such as ALS, AD and Parkinson’s disease are especially likely to require complex solutions. NSCs, as described in this review, can interface and work synergistically with gene and growth factor therapy, gap junction-mediated homeostasis, anti-apoptotic and neuroprotective strategies, stimulation of neurogenesis, anti-inflammatory and anti-scarring approaches, material science and tissue engineering, and at least in the case of SCI, with physical activity and exercise. Therefore, we propose an updated concept of NTF application. The traditional approach based on the pharmacological principles of trophic factor delivery (i.e., the dose and duration of administering a particular NTF), needs be revised to incorporate the newly appreciated importance of feedback-controlled production of trophic factors by the NSCs to mediate systemic homeostasis. Under this new conceptual context, we can begin to understand the wide spectrum of molecular tactics the NSCs deploy to regulate their trophic factor secretion at each normal developmental stage as they integrate into and prepare, modify, and guide the surrounding CNS environment towards the formation and homeostatic maintenance of a physiologically functioning adult nervous system. We believe that effectively adopting this conceptual view of “functional multipotency” of stem cells is essential for correct, efficient, and optimal use of trophic factors toward therapeutic goals (Fig. 1) [7,151].
Fig. (1) .
Schematic summary of stem cell-based therapeutic strategies. a, Stem cells possess intrinsic trophic factor producing abilities, and are able to respond to environmental cues to customize the profile of trophic factor production to stage homeostasis, which, as a capacity, can be further augmented by genetically engineering the cells with extra copies of transgenes of desirable molecules. b, Donor stem cells, prototype or genetically modified, may provide therapeutic benefits through at least three distinct mechanisms that could cast synergistic impacts: (1) replacement of the dysfunctional or dead host cells; (2) homeostatic regulation through delivery of trophic factors in targeted manners that are biologically regulated (e.g., in response to particular environmental cues, etc.) as well as establishment of gap junctions (b: upper inset), etc.; and (3) recruitment of and nourishment for host endogenous stem cells. The aforementioned therapeutic mechanism No. 2, apparently, carries a wide spectrum of regulatory tactics that can be further explored to refine the trophic factor and/or other cytokine secretion at each developmental stage or neural disorder status as NSCs integrate into and prepare, modify, and guide the surrounding CNS environment towards the formation and homeostatic maintenance of a physiologically functioning adult nervous system. We believe that effectively adopting this conceptual view of the stem cell-based NTF application is essential for correct, efficient, and optimal use of trophic factors to reach their designated therapeutic goals.
ACKNOWLEDGMENTS
Support was received from the VA Rehabilitation R&D, VA Biomedical Laboratory R&D, NIH, DOD, CIMIT, HeadNorth Foundation, Department of Neurosurgery, Brigham & Women’s Hospital and Massachusetts SCI cure research grant (Teng Lab).
REFERENCES
- 1.Gage FH. Stem cells of the central nervous system. Curr. Opin. Neurobiol. 1998;8(5):671–676. doi: 10.1016/s0959-4388(98)80098-6. [DOI] [PubMed] [Google Scholar]
- 2.Ryder EF, Snyder EY, Cepko CL. Establishment and characterization of multipotent neural cell lines using retrovirus vector-mediated oncogene transfer. J. Neurobiol. 1990;21(2 ):356–375. doi: 10.1002/neu.480210209. [DOI] [PubMed] [Google Scholar]
- 3.Reynolds BA, Weiss S. Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system. Science. 1992;255(5052 ):1707–1710. doi: 10.1126/science.1553558. [DOI] [PubMed] [Google Scholar]
- 4.Snyder EY, Deitcher DL, Walsh C, Arnold-Aldea S, Hartwieg EA, Cepko CL. Multipotent neural cell lines can engraft and participate in development of mouse cerebellum. Cell. 1992;68(1 ):33–51. doi: 10.1016/0092-8674(92)90204-p. [DOI] [PubMed] [Google Scholar]
- 5.Angevine JB Jr, Sidman RL. Autoradiographic study of cell migration during histogenesis of cerebral cortex in the mouse. Nature. 1961;192:766–768. doi: 10.1038/192766b0. [DOI] [PubMed] [Google Scholar]
- 6.Altman J. Are new neurons formed in the brains of adult mammals? Science. 1962;135:1127–1128. doi: 10.1126/science.135.3509.1127. [DOI] [PubMed] [Google Scholar]
- 7.Teng YD, Kabatas S, Wakeman DR, Li J, Snyder EY, Sidman RL. Functional multipotency of neural stem cells and its therapeutic implications. In: Ulrich H, editor. Perspectives of Stem Cells: From tools for studying mechanisms of neuronal differentiation towards therapy. Chapter 16. San Diego: Springer-Verlag Inc; 2009. pp. 255–270. [Google Scholar]
- 8.Jäderstad J, Jäderstad LM, Li J, Chintawar S, Salto C, Pandolfo M, Ourednik V, Teng YD, Sidman RL, Arenas E, Snyder EY, Herlenius E. Communication via gap junctions underlies early functional and beneficial interactions between grafted neural stem cells and the host. Proc. Natl. Acad. Sci. USA. 2010;107(11 ):5184–5189. doi: 10.1073/pnas.0915134107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Rosario CM, Yandava BD, Kosaras B, Zurakowski D, Sidman RL, Snyder EY. Differentiation of engrafted multipotent neural progenitors towards replacement of missing granule neurons in meander tail cerebellum may help determine the locus of mutant gene action. Development. 1997;124(21 ):4213–4224. doi: 10.1242/dev.124.21.4213. [DOI] [PubMed] [Google Scholar]
- 10.Auerbach JM, Eiden MV, McKay RD. Transplanted CNS stem cells form functional synapses in vivo. Eur. J. Neurosci. 2000;12(5 ):1696–1704. doi: 10.1046/j.1460-9568.2000.00067.x. [DOI] [PubMed] [Google Scholar]
- 11.Chung S, Shin BS, Hwang M, Lardaro T, Kang UJ, Isacson O, Kim KS. Neural precursors derived from embryonic stem cells, but not those from fetal ventral mesencephalon, maintain the potential to differentiate into dopaminergic neurons after expansion in vitro. Stem Cells. 2006;24(6 ):1583–1593. doi: 10.1634/stemcells.2005-0558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Yandava BD, Billinghurst LL, Snyder EY. "Global" cell replacement is feasible via neural stem Cell Transplant.ation: evidence from the dysmyelinated shiverer mouse brain. Proc. Natl. Acad. Sci. USA. 1999;96(12 ):7029–7034. doi: 10.1073/pnas.96.12.7029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Imitola J, Raddassi K, Park KI, Mueller FJ, Nieto M, Teng YD, Frenkel D, Li J, Sidman RL, Walsh CA, Snyder EY, Khoury SJ. Directed migration of neural stem cells to sites of CNS injury by the stromal cell-derived factor 1alpha/CXC chemokine receptor 4 pathway. Proc. Natl. Acad. Sci. USA. 2004;101(52 ):18117–18122. doi: 10.1073/pnas.0408258102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Redmond DE, Jr, Bjugstad KB, Teng YD, Ourednik V, Ourednik J, Wakeman DR, Parsons XH, Gonzalez R, Blanchard BC, Kim SU, Gu Z, Lipton SA, Markakis EA, Roth RH, Elsworth JD, Sladek JR, Jr, Sidman RL, Snyder EY. Behavioral improvement in a primate Parkinson's model is associated with multiple homeostatic effects of human neural stem cells. Proc. Natl. Acad. Sci. USA. 2007;104(29 ):12175–12180. doi: 10.1073/pnas.0704091104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Behrstock S, Ebert AD, Klein S, Schmitt M, Moore JM, Svendsen CN. Lesion-induced increase in survival and migration of human neural progenitor cells releasing GDNF. Cell Transplant. 2008;17(7 ):753–762. doi: 10.3727/096368908786516819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Aboody KS, Brown A, Rainov NG, Bower KA, Liu S, Yang W, Small JE, Herrlinger U, Ourednik V, Black PM, Breakefield XO, Snyder EY. Neural stem cells display extensive tropism for pathology in adult brain: evidence from intracranial gliomas. Proc. Natl. Acad. Sci. USA. 2000;97(23 ):12846–12851. doi: 10.1073/pnas.97.23.12846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Park KI, Teng YD, Snyder EY. The injured brain interacts reciprocally with neural stem cells supported by scaffolds to reconstitute lost tissue. Nat. Biotechnol. 2002;20(11 ):1111–1117. doi: 10.1038/nbt751. [DOI] [PubMed] [Google Scholar]
- 18.Ma DK, Ming GL, Song H. Glial influences on neural stem cell development: cellular niches for adult neurogenesis. Curr. Opin. Neurobiol. 2005;15(5 ):514–520. doi: 10.1016/j.conb.2005.08.003. [DOI] [PubMed] [Google Scholar]
- 19.Cao QL, Zhang YP, Howard RM, Walters WM, Tsoulfas P, Whittemore SR. Pluripotent stem cells engrafted into the normal or lesioned adult rat spinal cord are restricted to a glial lineage. Exp. Neurol. 2001;167(1 ):48–58. doi: 10.1006/exnr.2000.7536. [DOI] [PubMed] [Google Scholar]
- 20.Teng YD, Lavik EB, Qu X, Park KI, Ourednik J, Zura-kowski D, Langer R, Snyder EY. Functional recovery following traumatic spinal cord injury mediated by a unique polymer scaffold seeded with neural stem cells. Proc. Natl. Acad. Sci. USA. 2002;99(5 ):3024–3029. doi: 10.1073/pnas.052678899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Pluchino S, Zanotti L, Rossi B, Brambilla E, Ottoboni L, Salani G, Martinello M, Cattalini A, Bergami A, Furlan R, Comi G, Constantin G, Martino G. Neurosphere-derived multipotent precursors promote neuroprotection by an immunomodulatory mechanism. Nature. 2005;436(7048 ):266–271. doi: 10.1038/nature03889. [DOI] [PubMed] [Google Scholar]
- 22.Gersh BJ, Simari RD, Behfar A, Terzic CM, Terzic A. Cardiac cell repair therapy: a clinical perspective. Mayo Clin. Proc. 2009;84(10 ):876–892. doi: 10.4065/84.10.876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Redmond DE Jr, Elsworth JD, Roth RH, Leranth C, Collier TJ, Blanchard B, Bjugstad KB, Samulski RJ, Aebischer P, Sladek J.R., Jr. Embryonic substantia nigra grafts in the mesencephalon send neurites to the host striatum in non-human primate after overexpression of GDNF. J. Comp. Neurol. 2009;515(1 ):31–40. doi: 10.1002/cne.22028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Mocchetti I, Bachis A, Masliah E. Chemokine receptors and neurotrophic factors: potential therapy against aids dementia? J. Neurosci. Res. 2008;86(2 ):243–255. doi: 10.1002/jnr.21492. [DOI] [PubMed] [Google Scholar]
- 25.Ventura C, Cavallini C, Bianchi F, Cantoni S. Stem cells and cardiovascular repair: a role for natural and synthetic molecules harboring differentiating and paracrine logics. Cardiovasc. Hematol. Agents Med. Chem. 2008;6(1 ):60–68. doi: 10.2174/187152508783329975. [DOI] [PubMed] [Google Scholar]
- 26.Kaplan DR, Miller FD. Neurotrophin signal transduction in the nervous system. Curr. Opin. Neurobiol. 2000;10(3 ):381–391. doi: 10.1016/s0959-4388(00)00092-1. [DOI] [PubMed] [Google Scholar]
- 27.Aicardi G, Argilli E, Cappello S, Santi S, Riccio M, Thoenen H, Canossa M. Induction of long-term potentiation and depression is reflected by corresponding changes in secretion of endogenous brain-derived neurotrophic factor. Proc. Natl. Acad. Sci. USA. 2004;101(44 ):15788–15792. doi: 10.1073/pnas.0406960101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Tuszynski MH. Neurotrophic factors. In: Tuszynski JH, editor. CNS Regeneration: Basic Science and Clinical Advances. San Diego: Academic Press; 1999. pp. 109–158. [Google Scholar]
- 29.Tuszynski MH. Nerve growth factor gene delivery: animal models to clinical trials. Dev. Neurobiol. 2007;67(9 ):1204–1215. doi: 10.1002/dneu.20510. [DOI] [PubMed] [Google Scholar]
- 30.Bibel M, Barde YA. Neurotrophins: key regulators of cell fate and cell shape in the vertebrate nervous system. Genes Dev. 2000;14(23 ):2919–2937. doi: 10.1101/gad.841400. [DOI] [PubMed] [Google Scholar]
- 31.Hapner SJ, Boeshore KL, Large TH, Lefcort F. Neural differentiation promoted by truncated trkC receptors in collaboration with p75(NTR) Dev. Biol. 1998;201(1 ):90–100. doi: 10.1006/dbio.1998.8970. [DOI] [PubMed] [Google Scholar]
- 32.Guo JS, Zeng YS, Li HB, Huang WL, Liu RY, Li XB, Ding Y, Wu LZ, Cai DZ. Cotransplant of neural stem cells and NT-3 gene modified Schwann cells promote the recovery of transected spinal cord injury. Spinal Cord. 2007;45(1 ):15–24. doi: 10.1038/sj.sc.3101943. [DOI] [PubMed] [Google Scholar]
- 33.Wang JM, Zeng YS, Liu RY, Huang WL, Xiong Y, Wang YH, Chen SJ, Teng YD. Recombinant adenovirus vector-mediated functional expression of neurotropin-3 receptor (TrkC) in neural stem cells. Exp. Neurol. 2007;203(1 ):123–127. doi: 10.1016/j.expneurol.2006.07.028. [DOI] [PubMed] [Google Scholar]
- 34.Castellanos DA, Tsoulfas P, Frydel BR, Gajavelli S, Bes JC, Sagen J. TrkC overexpression enhances survival and migration of neural stem Cell Transplant.s in the rat spinal cord. Cell Transplant. 2002;11(3 ):297–307. [PubMed] [Google Scholar]
- 35.Bartkowska K, Paquin A, Gauthier AS, Kaplan DR, Miller FD. Trk signaling regulates neural precursor cell proliferation and differentiation during cortical development. Development. 2007;134(24 ):4369–4380. doi: 10.1242/dev.008227. [DOI] [PubMed] [Google Scholar]
- 36.Barker PA, Lomen-Hoerth C, Gensch EM, Meakin SO, Glass DJ, Shooter EM. Tissue-specific alternative splicing generates two isoforms of the trkA receptor. J. Biol. Chem. 1993;268(20 ):15150–15157. [PubMed] [Google Scholar]
- 37.Tacconelli A, Farina AR, Cappabianca L, Desantis G, Tessitore A, Vetuschi A, Sferra R, Rucci N, Argenti B, Screpanti I, Gulino A, Mackay AR. TrkA alternative splicing: a regulated tumor-promoting switch in human neuroblastoma. Cancer Cell. 2004;6(4 ):347–360. doi: 10.1016/j.ccr.2004.09.011. [DOI] [PubMed] [Google Scholar]
- 38.Narumiya S, Ohno M, Tanaka N, Yamano T, Shimada M. Enhanced expression of full-length TrkB receptors in young rat brain with hypoxic/ischemic injury. Brain Res. 1998;797(2 ):278–286. doi: 10.1016/s0006-8993(98)00385-0. [DOI] [PubMed] [Google Scholar]
- 39.Fryer RH, Kaplan DR, Kromer LF. Truncated trkB receptors on nonneuronal cells inhibit BDNF-induced neurite outgrowth in vitro. Exp. Neurol. 1997;148(2 ):616–627. doi: 10.1006/exnr.1997.6699. [DOI] [PubMed] [Google Scholar]
- 40.Bartkowska K, Paquin A, Gauthier AS, Kaplan DR, Miller FD. Trk signaling regulates neural precursor cell proliferation and differentiation during cortical development. Development. 2007;134(24 ):4369–4380. doi: 10.1242/dev.008227. [DOI] [PubMed] [Google Scholar]
- 41.Hempstead BL. The many faces of p75NTR. Curr. Opin. Neurobiol. 2002;12(3 ):260–267. doi: 10.1016/s0959-4388(02)00321-5. [DOI] [PubMed] [Google Scholar]
- 42.Islam O, Loo TX, Heese K. Brain-derived neurotrophic factor (BDNF) has proliferative effects on neural stem cells through the truncated TRK-B receptor, MAP kinase, AKT, and STAT-3 signaling pathways. Curr. Neurovasc. Res. 2009;6(1 ):42–53. doi: 10.2174/156720209787466028. [DOI] [PubMed] [Google Scholar]
- 43.Naumann T, Casademunt E, Hollerbach E, Hofmann J, Dechant G, Frotscher M, Barde YA. Complete deletion of the neurotrophin receptor p75NTR leads to long-lasting increases in the number of basal forebrain cholinergic neurons. J. Neurosci. 2002;22(7 ):2409–2418. doi: 10.1523/JNEUROSCI.22-07-02409.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Troy CM, Friedman JE, Friedman WJ. Mechanisms of p75-mediated death of hippocampal neurons. Role of caspases. J. Biol. Chem. 2002;277(37 ):34295–34302. doi: 10.1074/jbc.M205167200. [DOI] [PubMed] [Google Scholar]
- 45.Moscatelli I, Pierantozzi E, Camaioni A, Siracusa G, Campag-nolo L. p75 neurotrophin receptor is involved in proliferation of undifferentiated mouse embryonic stem cells. Exp. Cell Res. 2009;315(18 ):3220–3232. doi: 10.1016/j.yexcr.2009.08.014. [DOI] [PubMed] [Google Scholar]
- 46.Zhang W, Zeng YS, Wang JM, Ding Y, Li Y, Wu W. Neurotrophin-3 improves retinoic acid-induced neural differentiation of skin-derived precursors through a p75NTR-dependent signaling pathway. Neurosci. Res. 2009;64(2 ):170–176. doi: 10.1016/j.neures.2009.02.010. [DOI] [PubMed] [Google Scholar]
- 47.Morris ZS, McClatchey AI. The neurofibroma cell of origin: SKPs expand the playing field. Cell Stem Cell. 2009;4(5 ):371–372. doi: 10.1016/j.stem.2009.04.010. [DOI] [PubMed] [Google Scholar]
- 48.Reier PJ. Cellular transplantation strategies for spinal cord injury and translational neurobiology. NeuroRx. 2004;1(4 ):424–451. doi: 10.1602/neurorx.1.4.424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Fitch MT, Silver J. CNS injury, glial scars, and inflammation: Inhibitory extracellular matrices and regeneration failure. Exp. Neurol. 2008;209(2 ):294–301. doi: 10.1016/j.expneurol.2007.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Terenghi G. Peripheral nerve regeneration and neurotrophic factors. J. Anat. 1999;194( Pt 1 ):1–14. doi: 10.1046/j.1469-7580.1999.19410001.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Boyd JG, Gordon T. Neurotrophic factors and their receptors in axonal regeneration and functional recovery after peripheral nerve injury. Mol. Neurobiol. 2003;27(3 ):277–324. doi: 10.1385/MN:27:3:277. [DOI] [PubMed] [Google Scholar]
- 52.Silver J, Miller JH. Regeneration beyond the glial scar. Nat. Rev. Neurosci. 2004;5(2 ):146–156. doi: 10.1038/nrn1326. [DOI] [PubMed] [Google Scholar]
- 53.Fawcett JW. Overcoming inhibition in the damaged spinal cord. J. Neurotrauma. 2006;23(3-4 ):371–383. doi: 10.1089/neu.2006.23.371. [DOI] [PubMed] [Google Scholar]
- 54.Neumann S, Woolf CJ. Regeneration of dorsal column fibers into and beyond the lesion site following adult spinal cord injury. Neuron. 1999;23(1 ):83–91. doi: 10.1016/s0896-6273(00)80755-2. [DOI] [PubMed] [Google Scholar]
- 55.Seijffers R, Mills CD, Woolf CJ. ATF3 increases the intrinsic growth state of DRG neurons to enhance peripheral nerve regeneration. J. Neurosci. 2007;27(30 ):7911–7920. doi: 10.1523/JNEUROSCI.5313-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Lakatos A, Franklin RJ. Transplant mediated repair of the central nervous system: an imminent solution? Curr. Opin. Neurol. 2002;15(6 ):701–705. doi: 10.1097/01.wco.0000044766.39452.f6. [DOI] [PubMed] [Google Scholar]
- 57.Tuszynski MH. Growth-factor gene therapy for neurodegenerative disorders. Lancet Neurol. 2002;1(1 ):51–57. doi: 10.1016/s1474-4422(02)00006-6. [DOI] [PubMed] [Google Scholar]
- 58.Qiu J, Cai D, Dai H, McAtee M, Hoffman PN, Bregman BS, Filbin MT. Spinal axon regeneration induced by elevation of cyclic AMP. Neuron. 2002;34(6 ):895–903. doi: 10.1016/s0896-6273(02)00730-4. [DOI] [PubMed] [Google Scholar]
- 59.Novikova LN, Novikov LN, Kellerth JO. Biopolymers and biodegradable smart implants for tissue regeneration after spinal cord injury. Curr. Opin. Neurol. 2003;16(6 ):711–715. doi: 10.1097/01.wco.0000102620.38669.3e. [DOI] [PubMed] [Google Scholar]
- 60.Rossi F, Gianola S, Corvetti L. Regulation of intrinsic neuronal properties for axon growth and regeneration. Prog. Neurobiol. 2007;81(1 ):1–28. doi: 10.1016/j.pneurobio.2006.12.001. [DOI] [PubMed] [Google Scholar]
- 61.Lu P, Yang H, Jones LL, Filbin MT, Tuszynski MH. Combinatorial therapy with neurotrophins and cAMP promotes axonal regeneration beyond sites of spinal cord injury. J. Neurosci. 2004;24(28 ):6402–6409. doi: 10.1523/JNEUROSCI.1492-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Nikulina E, Tidwell JL, Dai HN, Bregman BS, Filbin MT. The phosphodiesterase inhibitor rolipram delivered after a spinal cord lesion promotes axonal regeneration and functional recovery. Proc. Natl. Acad. Sci. USA. 2004;101(23 ):8786–8790. doi: 10.1073/pnas.0402595101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Pearse DD, Pereira FC, Marcillo AE, Bates ML, Berrocal YA, Filbin MT, Bunge MB. cAMP and Schwann cells promote axonal growth and functional recovery after spinal cord injury. Nat. Med. 2004;10(6 ):610–616. doi: 10.1038/nm1056. [DOI] [PubMed] [Google Scholar]
- 64.Kobayashi NR, Fan DP, Giehl KM, Bedard AM, Wiegand SJ, Tetzlaff W. BDNF and NT-4/5 prevent atrophy of rat rubrospinal neurons after cervical axotomy, stimulate GAP-43 and Talpha1-tubulin mRNA expression, and promote axonal regeneration. J. Neurosci. 1997;17(24 ):9583–9595. doi: 10.1523/JNEUROSCI.17-24-09583.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Liu Y, Kim D, Himes BT, Chow SY, Schallert T, Murray M, Tessler A, Fischer I. Transplants of fibroblasts genetically modified to express BDNF promote regeneration of adult rat rubrospinal axons and recovery of forelimb function. J. Neurosci. 1999;19(11 ):4370–4387. doi: 10.1523/JNEUROSCI.19-11-04370.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Jin Y, Fischer I, Tessler A, Houle JD. Transplants of fibroblasts genetically modified to express BDNF promote axonal regeneration from supraspinal neurons following chronic spinal cord injury. Exp. Neurol. 2002;177(1 ):265–275. doi: 10.1006/exnr.2002.7980. [DOI] [PubMed] [Google Scholar]
- 67.Kwon BK, Liu J, Messerer C, Kobayashi NR, McGraw J, Oschipok L, Tetzlaff W. Survival and regeneration of rubrospinal neurons 1 year after spinal cord injury. Proc. Natl. Acad. Sci. USA. 2002;99(5 ):3246–3251. doi: 10.1073/pnas.052308899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Lu P, Blesch A, Tuszynski MH. Neurotrophism without neurotropism: BDNF promotes survival but not growth of lesioned corticospinal neurons. J. Comp. Neurol. 2001;436(4 ):456–470. doi: 10.1002/cne.1080. [DOI] [PubMed] [Google Scholar]
- 69.Schnell L, Schneider R, Kolbeck R, Barde YA, Schwab ME. Neurotrophin-3 enhances sprouting of corticospinal tract during development and after adult spinal cord lesion. Nature. 1994;367(6459 ):170–173. doi: 10.1038/367170a0. [DOI] [PubMed] [Google Scholar]
- 70.Grill R, Murai K, Blesch A, Gage FH, Tuszynski MH. Cellular delivery of neurotrophin-3 promotes corticospinal axonal growth and partial functional recovery after spinal cord injury. J. Neurosci. 1997;17(14 ):5560–5572. doi: 10.1523/JNEUROSCI.17-14-05560.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Kadoya K, Tsukada S, Lu P, Coppola G, Geschwind D, Filbin MT, Blesch A, Tuszynski MH. Combined intrinsic and extrinsic neuronal mechanisms facilitate bridging axonal regeneration one year after spinal cord injury. Neuron. 2009;64(2 ):165–172. doi: 10.1016/j.neuron.2009.09.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Hendriks WT, Ruitenberg MJ, Blits B, Boer GJ, Verhaagen J. Viral vector-mediated gene transfer of neurotrophins to promote regeneration of the injured spinal cord. Prog. Brain Res. 2004;146:451–476. doi: 10.1016/S0079-6123(03)46029-9. [DOI] [PubMed] [Google Scholar]
- 73.Ying Z, Roy RR, Zhong H, Zdunowski S, Edgerton VR, Gomez-Pinilla F. BDNF-exercise interactions in the recovery of symmetrical stepping after a cervical hemisection in rats. NeuroScience. 2008;155(4):1070–1078. doi: 10.1016/j.neuroscience.2008.06.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Konya D, Liao WL, Choi H, Yu D, Woodard MC, Newton KM, King AM, Pamir NM, Black PM, Frontera WR, Sab-harwal S, Teng YD. Functional recovery in T13-L1 hemisected rats resulting from peripheral nerve rerouting: role of central neuroplasticity. Regen. Med. 2008;3(3 ):309–327. doi: 10.2217/17460751.3.3.309. [DOI] [PubMed] [Google Scholar]
- 75.Foret A, Quertainmont R, Botman O, Bouhy D, Amabili P, Brook G, Schoenen J, Franzen R. Stem cells in the adult rat spinal cord: plasticity after injury and treadmill training exercise. J. Neurochem. 2010;112(3 ):762–772. doi: 10.1111/j.1471-4159.2009.06500.x. [DOI] [PubMed] [Google Scholar]
- 76.Behrman AL, Bowden MG, Nair PM. Neuroplasticity after spinal cord injury and training: an emerging paradigm shift in rehabilitation and walking recovery. Phys. Ther. 2006;86(10 ):1406–1425. doi: 10.2522/ptj.20050212. [DOI] [PubMed] [Google Scholar]
- 77.Mocchetti I, Brown M. Targeting neurotrophin receptors in the central nervous system. CNS Neurol. Disord. Drug Targets. 2008;7(1 ):71–82. doi: 10.2174/187152708783885138. [DOI] [PubMed] [Google Scholar]
- 78.Adrian EK, Jr., Walker BE. Incorporation of thymidine-H3 by cells in normal and injured mouse spinal cord. J. Neuropathol. Exp. Neurol. 1962;21:597–609. doi: 10.1097/00005072-196210000-00007. [DOI] [PubMed] [Google Scholar]
- 79.Weiss S, Dunne C, Hewson J, Wohl C, Wheatley M, Peterson AC, Reynolds BA. Multipotent CNS stem cells are present in the adult mammalian spinal cord and ventricular neuroaxis. J. Neurosci. 1996;16(23 ):7599–7609. doi: 10.1523/JNEUROSCI.16-23-07599.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Tramontin AD, Garcia-Verdugo JM, Lim DA, Alvarez-Buylla A. Postnatal development of radial glia and the ventricular zone (VZ): a continuum of the neural stem cell compartment. Cereb. Cortex. 2003;13(6 ):580–587. doi: 10.1093/cercor/13.6.580. [DOI] [PubMed] [Google Scholar]
- 81.Shihabuddin LS, Horner PJ, Ray J, Gage FH. Adult spinal cord stem cells generate neurons after transplantation in the adult dentate gyrus. J. Neurosci. 2000;20(23 ):8727–8735. doi: 10.1523/JNEUROSCI.20-23-08727.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Horner PJ, Power AE, Kempermann G, Kuhn HG, Palmer TD, Winkler J, Thal LJ, Gage FH. Proliferation and differentiation of progenitor cells throughout the intact adult rat spinal cord. J. Neurosci. 2000;20(6 ):2218–2228. doi: 10.1523/JNEUROSCI.20-06-02218.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Song HJ, Stevens CF, Gage FH. Neural stem cells from adult hippocampus develop essential properties of functional CNS neurons. Nat. Neurosci. 2002;5(5 ):438–445. doi: 10.1038/nn844. [DOI] [PubMed] [Google Scholar]
- 84.Frisen J, Johansson CB, Torok C, Risling M, Lendahl U. Rapid, widespread, and longlasting induction of nestin contributes to the generation of glial scar tissue after CNS injury. J. Cell Biol. 1995;131(2 ):453–464. doi: 10.1083/jcb.131.2.453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Shihabuddin LS, Palmer TD, Gage FH. The search for neural progenitor cells: prospects for the therapy of neurodegenerative disease. Mol. Med. Today. 1999;5(11 ):474–480. doi: 10.1016/s1357-4310(99)01596-8. [DOI] [PubMed] [Google Scholar]
- 86.Yamamoto S, Yamamoto N, Kitamura T, Nakamura K, Nakafuku M. Proliferation of parenchymal neural progenitors in response to injury in the adult rat spinal cord. Exp. Neurol. 2001;172(1 ):115–127. doi: 10.1006/exnr.2001.7798. [DOI] [PubMed] [Google Scholar]
- 87.Matthews MA, St Onge MF, Faciane CL. An electron microscopic analysis of abnormal ependymal cell proliferation and envelopment of sprouting axons following spinal cord transection in the rat. Acta Neuropathol. 1979;45(1 ):27–36. doi: 10.1007/BF00691801. [DOI] [PubMed] [Google Scholar]
- 88.Tripathi RB, McTigue DM. Chronically increased ciliary neurotrophic factor and fibroblast growth factor-2 expression after spinal contusion in rats. J. Comp. Neurol. 2008;510(2 ):129–144. doi: 10.1002/cne.21787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Takahashi M, Arai Y, Kurosawa H, Sueyoshi N, Shirai S. Ependymal cell reactions in spinal cord segments after compression injury in adult rat. J. Neuropathol. Exp. Neurol. 2003;62(2 ):185–194. doi: 10.1093/jnen/62.2.185. [DOI] [PubMed] [Google Scholar]
- 90.McTigue DM, Horner PJ, Stokes BT, Gage FH. Neurotrophin-3 and brain-derived neurotrophic factor induce oligodendrocyte proliferation and myelination of regenerating axons in the contused adult rat spinal cord. J. Neurosci. 1998;18(14 ):5354–5365. doi: 10.1523/JNEUROSCI.18-14-05354.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Mothe AJ, Tator CH. Proliferation, migration, and differentiation of endogenous ependymal region stem/progenitor cells following minimal spinal cord injury in the adult rat. NeuroScience. 2005;131(1 ):177–187. doi: 10.1016/j.neuroscience.2004.10.011. [DOI] [PubMed] [Google Scholar]
- 92.Kempermann G, Kuhn HG, Gage FH. More hippocampal neurons in adult mice living in an enriched environment. Nature. 1997;386(6624 ):493–495. doi: 10.1038/386493a0. [DOI] [PubMed] [Google Scholar]
- 93.Teng YD, Liao WL, Choi H, Konya D, Sabharwal S, Langer R, Sidman RL, Snyder EY, Frontera WR. Physical activity-mediated functional recovery after spinal cord injury: potential roles of neural stem cells. Regen. Med. 2006;1(6 ):763–776. doi: 10.2217/17460751.1.6.763. [DOI] [PubMed] [Google Scholar]
- 94.Gomez-Pinilla F, Ying Z, Roy RR, Molteni R, Edgerton VR. Voluntary exercise induces a BDNF-mediated mechanism that promotes neuroplasticity. J. Neurophysiol. 2002;88(5 ):2187–2195. doi: 10.1152/jn.00152.2002. [DOI] [PubMed] [Google Scholar]
- 95.Jakeman LB, Wei P, Guan Z, Stokes BT. Brain-derived neurotrophic factor stimulates hindlimb stepping and sprouting of cholinergic fibers after spinal cord injury. Exp. Neurol. 1998;154(1 ):170–184. doi: 10.1006/exnr.1998.6924. [DOI] [PubMed] [Google Scholar]
- 96.Ying Z, Roy RR, Edgerton VR, Gomez-Pinilla F. Voluntary exercise increases neurotrophin-3 and its receptor TrkC in the spinal cord. Brain Res. 2003;987(1 ):93–99. doi: 10.1016/s0006-8993(03)03258-x. [DOI] [PubMed] [Google Scholar]
- 97.Danzer SC, He X, McNamara JO. Ontogeny of seizure-induced increases in BDNF immunoreactivity and TrkB receptor activation in rat hippocampus. Hippocampus. 2004;14(3 ):345–355. doi: 10.1002/hipo.10190. [DOI] [PubMed] [Google Scholar]
- 98.Osaka M, Honmou O, Murakami T, Nonaka T, Houkin K, Hamada H, Kocsis JD. Intravenous administration of mesenchymal stem cells derived from bone marrow after contusive spinal cord injury improves functional outcome. Brain Res. 2010;1343:226–235. doi: 10.1016/j.brainres.2010.05.011. [DOI] [PubMed] [Google Scholar]
- 99.van Praag H, Kempermann G, Gage FH. Running increases cell proliferation and neurogenesis in the adult mouse dentate gyrus. Nat. Neurosci. 1999;2(3 ):266–270. doi: 10.1038/6368. [DOI] [PubMed] [Google Scholar]
- 100.van Praag H, Christie BR, Sejnowski TJ, Gage FH. Running enhances neurogenesis, learning, and long-term potentiation in mice. Proc. Natl. Acad. Sci. USA. 1999;96(23 ):13427–13431. doi: 10.1073/pnas.96.23.13427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Vaynman S, Gomez-Pinilla F. License to run: exercise impacts functional plasticity in the intact and injured central nervous system by using neurotrophins. Neurorehabil. Neural Repair. 2005;19(4 ):283–295. doi: 10.1177/1545968305280753. [DOI] [PubMed] [Google Scholar]
- 102.Krityakiarana W, Espinosa-Jeffrey A, Ghiani CA, Zhao PM, Topaldjikian N, Gomez-Pinilla F, Yamaguchi M, Kotchabhakdi N, de Vellis J. Voluntary exercise increases oligodendrogenesis in spinal cord. Int. J. Neurosci. 2010;120(4 ):280–290. doi: 10.3109/00207450903222741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Snyder EY, Taylor RM, Wolfe JH. Neural progenitor cell engraftment corrects lysosomal storage throughout the MPS VII mouse brain. Nature. 1995;374(6520 ):367–370. doi: 10.1038/374367a0. [DOI] [PubMed] [Google Scholar]
- 104.Imitola J, Park KI, Teng YD, Nisim S, Lachyankar M, Ourednik J, Mueller FJ, Yiou R, Atala A, Sidman RL, Tuszynski M, Khoury SJ, Snyder EY. Stem cells: cross-talk and developmental programs. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2004;359(1445 ):823–837. doi: 10.1098/rstb.2004.1474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Flax JD, Aurora S, Yang C, Simonin C, Wills AM, Bill-inghurst LL, Jendoubi M, Sidman RL, Wolfe JH, Kim SU, Snyder EY. Engraftable human neural stem cells respond to developmental cues, replace neurons, and express foreign genes. Nat. Biotechnol. 1998;16(11 ):1033–1039. doi: 10.1038/3473. [DOI] [PubMed] [Google Scholar]
- 106.Hill CE, Guller Y, Raffa SJ, Hurtado A, Bunge M. A calpain inhibitor enhances the survival of Schwann cells in vitro and after transplantation into the injured spinal cord. J. Neurotrauma. 2010;27(9 ):1685–1695. doi: 10.1089/neu.2010.1272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Yu D, Neeley WL, Pritchard CD, Slotkin JR, Woodard EJ, Langer R, Teng YD. Blockade of peroxynitrite-induced neural stem cell death in the acutely injured spinal cord by drug-releasing polymer. Stem Cells. 2009;27(5 ):1212–1222. doi: 10.1002/stem.26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Hedlund E, Hefferan MP, Marsala M, Isacson O. Cell therapy and stem cells in animal models of motor neuron disorders. Eur. J. Neurosci. 2007;26(7 ):1721–1737. doi: 10.1111/j.1460-9568.2007.05780.x. [DOI] [PubMed] [Google Scholar]
- 109.Suzuki M, Svendsen CN. Combining growth factor and stem cell therapy for amyotrophic lateral sclerosis. Trends Neurosci. 2008;31(4 ):192–198. doi: 10.1016/j.tins.2008.01.006. [DOI] [PubMed] [Google Scholar]
- 110.Lu P, Jones LL, Snyder EY, Tuszynski MH. Neural stem cells constitutively secrete neurotrophic factors and promote extensive host axonal growth after spinal cord injury. Exp. Neurol. 2003;181(2 ):115–129. doi: 10.1016/s0014-4886(03)00037-2. [DOI] [PubMed] [Google Scholar]
- 111.Levi-Montalcini R. The nerve growth factor 35 years later. Science. 1987;237(4819 ):1154–1162. doi: 10.1126/science.3306916. [DOI] [PubMed] [Google Scholar]
- 112.Levi-Montalcini R, Hamburger V. Selective growth stimulating effects of mouse sarcoma on the sensory and sympathetic nervous system of the chick embryo. J. Exp. Zool. 1951;116(2 ):321–361. doi: 10.1002/jez.1401160206. [DOI] [PubMed] [Google Scholar]
- 113.Fischer W, Wictorin K, Bjorklund A, Williams LR, Varon S, Gage FH. Amelioration of cholinergic neuron atrophy and spatial memory impairment in aged rats by nerve growth factor. Nature. 1987;329(6134 ):65–68. doi: 10.1038/329065a0. [DOI] [PubMed] [Google Scholar]
- 114.Markowska AL, Koliatsos VE, Breckler SJ, Price DL, Olton DS. Human nerve growth factor improves spatial memory in aged but not in young rats. J. Neurosci. 1994;14(8 ):4815–4824. doi: 10.1523/JNEUROSCI.14-08-04815.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Bezprozvanny I, Mattson MP. Neuronal calcium mishandling and the pathogenesis of Alzheimer's disease. Trends Neurosci. 2008;31(9 ):454–463. doi: 10.1016/j.tins.2008.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Koliatsos VE, Clatterbuck RE, Nauta HJ, Knusel B, Burton LE, Hefti FF, Mobley WC, Price DL. Human nerve growth factor prevents degeneration of basal forebrain cholinergic neurons in primates. Ann. Neurol. 1991;30(6 ):831–840. doi: 10.1002/ana.410300613. [DOI] [PubMed] [Google Scholar]
- 117.Emerich DF, Winn SR, Harper J, Hammang JP, Baetge EE, Kordower JH. Implants of polymer-encapsulated human NGF-secreting cells in the nonhuman primate: rescue and sprouting of degenerating cholinergic basal forebrain neurons. J. Comp. Neurol. 1994;349(1 ):148–164. doi: 10.1002/cne.903490110. [DOI] [PubMed] [Google Scholar]
- 118.Kordower JH, Winn SR, Liu YT, Mufson EJ, Sladek JR, Jr, Hammang JP, Baetge EE, Emerich DF. The aged monkey basal forebrain: rescue and sprouting of axotomized basal forebrain neurons after grafts of encapsulated cells secreting human nerve growth factor. Proc. Natl. Acad. Sci. USA. 1994;91(23 ):10898–10902. doi: 10.1073/pnas.91.23.10898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Holtzman DM, Li Y, Parada LF, Kinsman S, Chen CK, Valletta JS, Zhou J, Long JB, Mobley WC. p140trk mRNA marks NGF-responsive forebrain neurons: evidence that trk gene expression is induced by NGF. Neuron. 1992;9(3 ):465–478. doi: 10.1016/0896-6273(92)90184-f. [DOI] [PubMed] [Google Scholar]
- 120.Tuszynski MH, Gage FH. Bridging grafts and transient nerve growth factor infusions promote long-term central nervous system neuronal rescue and partial functional recovery. Proc. Natl. Acad. Sci. USA. 1995;92(10 ):4621–4625. doi: 10.1073/pnas.92.10.4621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Cooper JD, Salehi A, Delcroix JD, Howe CL, Belichenko PV, Chua-Couzens J, Kilbridge JF, Carlson EJ, Epstein CJ, Mobley WC. Failed retrograde transport of NGF in a mouse model of Down's syndrome: reversal of cholinergic neurodegenerative phenotypes following NGF infusion. Proc. Natl. Acad. Sci. USA. 2001;98(18 ):10439–10444. doi: 10.1073/pnas.181219298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Tuszynski MH, Roberts J, Senut MC, U HS, Gage FH. Gene therapy in the adult primate brain: intraparenchymal grafts of cells genetically modified to produce nerve growth factor prevent cholinergic neuronal degeneration. Gene Ther. 1996;3(4 ):305–314. [PubMed] [Google Scholar]
- 123.Perry EK, Curtis M, Dick DJ, Candy JM, Atack JR, Blox-ham CA, Blessed G, Fairbairn A, Tomlinson BE, Perry RH. Cholinergic correlates of cognitive impairment in Parkinson's disease: comparisons with Alzheimer's disease. J. Neurol. Neurosurg. Psychiatry. 1985;48(5 ):413–421. doi: 10.1136/jnnp.48.5.413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Perry EK, Tomlinson BE, Blessed G, Bergmann K, Gibson PH, Perry RH. Correlation of cholinergic abnormalities with senile plaques and mental test scores in senile dementia. Br. Med. J. 1978;2(6150 ):1457–1459. doi: 10.1136/bmj.2.6150.1457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Candy JM, Perry RH, Perry EK, Irving D, Blessed G, Fair-bairn AF, Tomlinson BE. Pathological changes in the nucleus of Meynert in Alzheimer's and Parkinson's diseases. J. Neurol. Sci. 1983;59(2 ):277–289. doi: 10.1016/0022-510x(83)90045-x. [DOI] [PubMed] [Google Scholar]
- 126.Masliah E, Terry RD, Alford M, DeTeresa R, Hansen LA. Cortical and subcortical patterns of synaptophysinlike immunoreactivity in Alzheimer's disease. Am. J. Pathol. 1991;138(1 ):235–246. [PMC free article] [PubMed] [Google Scholar]
- 127.Mufson EJ, Ikonomovic MD, Styren SD, Counts SE, Wuu J, Leurgans S, Bennett DA, Cochran EJ, DeKosky ST. Preservation of brain nerve growth factor in mild cognitive impairment and Alzheimer disease. Arch. Neurol. 2003;60(8 ):1143–1148. doi: 10.1001/archneur.60.8.1143. [DOI] [PubMed] [Google Scholar]
- 128.Korsching S, Auburger G, Heumann R, Scott J, Thoenen H. Levels of nerve growth factor and its mRNA in the central nervous system of the rat correlate with cholinergic innervation. EMBO J. 1985;4(6 ):1389–1393. doi: 10.1002/j.1460-2075.1985.tb03791.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Whittemore SR, Ebendal T, Larkfors L, Olson L, Seiger A, Stromberg I, Persson H. Development and regional expression of beta nerve growth factor messenger RNA and protein in the rat central nervous system. Proc. Natl. Acad. Sci. USA. 1986;83(3 ):817–821. doi: 10.1073/pnas.83.3.817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Savaskan E, Muller-Spahn F, Olivieri G, Bruttel S, Otten U, Rosenberg C, Hulette C, Hock C. Alterations in trk A, trk B and trk C receptor immunoreactivities in parietal cortex and cerebellum in Alzheimer's disease. Eur. Neurol. 2000;44(3 ):172–180. doi: 10.1159/000008229. [DOI] [PubMed] [Google Scholar]
- 131.Chu Y, Cochran EJ, Bennett DA, Mufson EJ, Kordower JH. Down-regulation of trkA mRNA within nucleus basalis neurons in individuals with mild cognitive impairment and Alzheimer's disease. J. Comp. Neurol. 2001;437(3 ):296–307. doi: 10.1002/cne.1284. [DOI] [PubMed] [Google Scholar]
- 132.Mufson EJ, Ma SY, Dills J, Cochran EJ, Leurgans S, Wuu J, Bennett DA, Jaffar S, Gilmor ML, Levey AI, Kordower JH. Loss of basal forebrain P75(NTR) immunoreactivity in subjects with mild cognitive impairment and Alzheimer's disease. J. Comp. Neurol. 2002;443(2 ):136–153. doi: 10.1002/cne.10122. [DOI] [PubMed] [Google Scholar]
- 133.Salehi A, Delcroix JD, Belichenko PV, Zhan K, Wu C, Valletta JS, Takimoto-Kimura R, Kleschevnikov AM, Sambamurti K, Chung PP, Xia W, Villar A, Campbell WA, Kulnane LS, Nixon RA, Lamb BT, Epstein CJ, Stokin GB, Goldstein LS, Mobley WC. Increased App expression in a mouse model of Down's syndrome disrupts NGF transport and causes cholinergic neuron degeneration. Neuron. 2006;51(1 ):29–42. doi: 10.1016/j.neuron.2006.05.022. [DOI] [PubMed] [Google Scholar]
- 134.Williams LR. Hypophagia is induced by intracerebroventricular administration of nerve growth factor. Exp. Neurol. 1991;113(1 ):31–37. doi: 10.1016/0014-4886(91)90143-z. [DOI] [PubMed] [Google Scholar]
- 135.Isaacson LG, Saffran BN, Crutcher KA. Intracerebral NGF infusion induces hyperinnervation of cerebral blood vessels. Neurobiol. Aging. 1990;11(1 ):51–55. doi: 10.1016/0197-4580(90)90062-5. [DOI] [PubMed] [Google Scholar]
- 136.Emmett CJ, Stewart GR, Johnson RM, Aswani SP, Chan RL, Jakeman LB. Distribution of radioiodinated recombinant human nerve growth factor in primate brain following intracerebroventricular infusion. Exp. Neurol. 1996;140(2 ):151–160. doi: 10.1006/exnr.1996.0125. [DOI] [PubMed] [Google Scholar]
- 137.Winkler J, Ramirez GA, Kuhn HG, Peterson DA, Day-Lollini PA, Stewart GR, Tuszynski MH, Gage FH, Thal LJ. Reversible Schwann cell hyperplasia and sprouting of sensory and sympathetic neurites after intraventricular administration of nerve growth factor. Ann. Neurol. 1997;41(1 ):82–93. doi: 10.1002/ana.410410114. [DOI] [PubMed] [Google Scholar]
- 138.Eriksdotter Jonhagen M, Nordberg A, Amberla K, Backman L, Ebendal T, Meyerson B, Olson L, Seiger Shigeta M, Theodorsson E, Viitanen M, Winblad B, Wahlund LO. Intracerebroventricular infusion of nerve growth factor in three patients with Alzheimer's disease. Dement. Geriatr. Cogn. Disord. 1998;9(5 ):246–257. doi: 10.1159/000017069. [DOI] [PubMed] [Google Scholar]
- 139.Kordower JH, Palfi S, Chen EY, Ma SY, Sendera T, Coch-ran EJ, Mufson EJ, Penn R, Goetz CG, Comella CD. Clinicopathological findings following intraventricular glial-derived neurotrophic factor treatment in a patient with Parkinson's disease. Ann. Neurol. 1999;46(3 ):419–424. doi: 10.1002/1531-8249(199909)46:3<419::aid-ana21>3.0.co;2-q. [DOI] [PubMed] [Google Scholar]
- 140.Nutt JG, Burchiel KJ, Comella CL, Jankovic J, Lang AE, Laws ER, Jr., Lozano AM, Penn RD, Simpson RK, Jr., Stacy M, Wooten GF. Randomized, double-blind trial of glial cell line-derived neurotrophic factor (GDNF) in PD. Neurology. 2003;60(1 ):69–73. doi: 10.1212/wnl.60.1.69. [DOI] [PubMed] [Google Scholar]
- 141.Cleveland DW, Rothstein JD. From Charcot to Lou Gehrig: deciphering selective motor neuron death in ALS. Nat. Rev. Neurosci. 2001;2(11 ):806–819. doi: 10.1038/35097565. [DOI] [PubMed] [Google Scholar]
- 142.Julien JP. Amyotrophic lateral sclerosis. unfolding the toxicity of the misfolded. Cell. 2001;104(4 ):581–591. doi: 10.1016/s0092-8674(01)00244-6. [DOI] [PubMed] [Google Scholar]
- 143.Doble A. The pharmacology and mechanism of action of riluzole. Neurology. 1996;47(6 Suppl 4 ):S233–241. doi: 10.1212/wnl.47.6_suppl_4.233s. [DOI] [PubMed] [Google Scholar]
- 144.Peluffo H, Estevez A, Barbeito L, Stutzmann JM. Riluzole promotes survival of rat motoneurons in vitro by stimulating trophic activity produced by spinal astrocyte monolayers. Neurosci. Lett. 1997;228(3 ):207–211. doi: 10.1016/s0304-3940(97)00384-4. [DOI] [PubMed] [Google Scholar]
- 145.Brown RH Jr. Amyotrophic lateral sclerosis: recent insights from genetics and transgenic mice. Cell. 1995;80(5 ):687–692. doi: 10.1016/0092-8674(95)90346-1. [DOI] [PubMed] [Google Scholar]
- 146.Andersen PM, Sims KB, Xin WW, Kiely R, O'Neill G, Ravits J, Pioro E, Harati Y, Brower RD, Levine JS, Heinicke HU, Seltzer W, Boss M, Brown RH., Jr Sixteen novel mutations in the Cu/Zn superoxide dismutase gene in amyotrophic lateral sclerosis: a decade of discoveries, defects and disputes. Amyotroph. Latera. Scler. Other Motor Neuron Disord. 2003;4(2 ):62–73. doi: 10.1080/14660820310011700. [DOI] [PubMed] [Google Scholar]
- 147.Garbuzova-Davis S, Willing AE, Milliken M, Saporta S, Zigova T, Cahill DW, Sanberg PR. Positive effect of transplantation of hNT neurons (NTera 2/D1 cell-line) in a model of familial amyotrophic lateral sclerosis. Exp. Neurol. 2002;174(2 ):169–180. doi: 10.1006/exnr.2002.7860. [DOI] [PubMed] [Google Scholar]
- 148.Corti S, Locatelli F, Donadoni C, Guglieri M, Papadimitriou D, Strazzer S, Del Bo R, Comi GP. Wild-type bone marrow cells ameliorate the phenotype of SOD1-G93A ALS mice and contribute to CNS, heart and skeletal muscle tissues. Brain. 2004;127(Pt 11 ):2518–2532. doi: 10.1093/brain/awh273. [DOI] [PubMed] [Google Scholar]
- 149.Hemendinger R, Wang J, Malik S, Persinski R, Copeland J, Emerich D, Gores P, Halberstadt C, Rosenfeld J. Sertoli cells improve survival of motor neurons in SOD1 transgenic mice, a model of amyotrophic lateral sclerosis. Exp. Neurol. 2005;196(2 ):235–243. doi: 10.1016/j.expneurol.2005.07.025. [DOI] [PubMed] [Google Scholar]
- 150.Yan J, Xu L, Welsh AM, Chen D, Hazel T, Johe K, Koliatsos VE. Combined immunosuppressive agents or CD4 antibodies prolong survival of human neural stem cell grafts and improve disease outcomes in amyotrophic lateral sclerosis transgenic mice. Stem Cells. 2006;24(8 ):1976–1985. doi: 10.1634/stemcells.2005-0518. [DOI] [PubMed] [Google Scholar]
- 151.Habisch HJ, Janowski M, Binder D, Kuzma-Kozakiewicz M, Widmann A, Habich A, Schwalenstocker B, Hermann A, Brenner R, Lukomska B, Domanska-Janik K, Ludolph AC, Storch A. Intrathecal application of neuroectodermally converted stem cells into a mouse model of ALS: limited intraparenchymal migration and survival narrows therapeutic effects. J. Neural. Transm. 2007;114(11 ):1395–1406. doi: 10.1007/s00702-007-0748-y. [DOI] [PubMed] [Google Scholar]
- 152.Garbuzova-Davis S, Klasko SK, Sanberg PR. Intravenous administration of human umbilical cord blood cells in an animal model of MPS III B. J. Comp. Neurol. 2009;515(1 ):93–101. doi: 10.1002/cne.21949. [DOI] [PubMed] [Google Scholar]
- 153.Svendsen CN, ter Borg MG, Armstrong RJ, Rosser AE, Chandran S, Ostenfeld T, Caldwell MA. A new method for the rapid and long term growth of human neural precursor cells. J. Neurosci. Methods. 1998;85(2 ):141–152. doi: 10.1016/s0165-0270(98)00126-5. [DOI] [PubMed] [Google Scholar]
- 154.Teng YD, Benn SC, Kalkanis SN, Shefner JM, Onario RC, Cheng B, Lachyankar MB, Macconi M, Li J, Maragakis NJ, Lládo J, Erkmen K, Redmond DE, Jr, Sidman RL, Przedborski S, Rothstein JD, Brown RH, Jr., Snyder EY. Neural stem cells, acting through multiple mechanisms, extensively prolong life and preserve function in a mouse model of ALS. 2010. to be submitted. [Google Scholar]
- 155.Xu L, Yan J, Chen D, Welsh AM, Hazel T, Johe K, Hatfield G, Koliatsos VE. Human neural stem cell grafts ameliorate motor neuron disease in SOD-1 transgenic rats. Transplantation. 2006;82(7 ):865–875. doi: 10.1097/01.tp.0000235532.00920.7a. [DOI] [PubMed] [Google Scholar]
- 156.Klein SM, Behrstock S, McHugh J, Hoffmann K, Wallace K, Suzuki M, Aebischer P, Svendsen CN. GDNF delivery using human neural progenitor cells in a rat model of ALS. Hum. Gene Ther. 2005;16(4 ):509–521. doi: 10.1089/hum.2005.16.509. [DOI] [PubMed] [Google Scholar]
- 157.Guillot S, Azzouz M, Deglon N, Zurn A, Aebischer P. Local GDNF expression mediated by lentiviral vector protects facial nerve motoneurons but not spinal motoneurons in SOD1(G93A) transgenic mice. Neurobiol. Dis. 2004;16(1 ):139–149. doi: 10.1016/j.nbd.2004.01.017. [DOI] [PubMed] [Google Scholar]
- 158.Suzuki M, McHugh J, Tork C, Shelley B, Klein SM, Aebischer P, Svendsen CN. GDNF secreting human neural progenitor cells protect dying motor neurons, but not their projection to muscle, in a rat model of familial ALS. PLoS One. 2007;2(1 ):e689. doi: 10.1371/journal.pone.0000689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Mazzini L, Ferrero I, Luparello V, Rustichelli D, Gunetti M, Mareschi K, Testa L, Stecco A, Tarletti R, Miglioretti M, Fava E, Nasuelli N, Cisari C, Massara M, Vercelli R, Oggioni GD, Carriero A, Cantello R, Monaco F, Fagioli F. Mesenchymal stem Cell Transplant.ation in amyotrophic lateral sclerosis: A Phase I clinical trial. Exp. Neurol. 2010;223(1 ):229–237. doi: 10.1016/j.expneurol.2009.08.007. [DOI] [PubMed] [Google Scholar]
- 160.Group A-CTS. A double-blind placebo-controlled clinical trial of subcutaneous recombinant human ciliary neurotrophic factor (rHCNTF) in amyotrophic lateral sclerosis. ALS CNTF Treatment Study Group. Neurology. 1996;46(5 ):1244–1249. doi: 10.1212/wnl.46.5.1244. [DOI] [PubMed] [Google Scholar]
- 161.Group TBS. A controlled trial of recombinant methionyl human BDNF in ALS: The BDNF Study Group (Phase III) Neurology. 1999;52(7 ):1427–1433. doi: 10.1212/wnl.52.7.1427. [DOI] [PubMed] [Google Scholar]
- 162.Nagai M, Re DB, Nagata T, Chalazonitis A, Jessell TM, Wichterle H, Przedborski S. Astrocytes expressing ALS-linked mutated SOD1 release factors selectively toxic to motor neurons. Nat. Neurosci. 2007;10(5 ):615–622. doi: 10.1038/nn1876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Yamanaka K, Chun SJ, Boillee S, Fujimori-Tonou N, Yama-shita H, Gutmann DH, Takahashi R, Misawa H, Cleveland DW. Astrocytes as determinants of disease progression in inherited amyotrophic lateral sclerosis. Nat. Neurosci. 2008;11(3 ):251–253. doi: 10.1038/nn2047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4 ):663–676. doi: 10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
- 165.Pick M, Stelzer Y, Bar-Nur O, Mayshar Y, Eden A, Benvenisty N. Clone- and gene-specific aberrations of parental imprinting in human induced pluripotent stem cells. Stem Cells. 2009;27(11 ):2686–2690. doi: 10.1002/stem.205. [DOI] [PubMed] [Google Scholar]
- 166.Marchetto MC, Yeo GW, Kainohana O, Marsala M, Gage FH, Muotri AR. Transcriptional sigNature. and memory retention of human-induced pluripotent stem cells. PLoS One. 2009;4(9 ):e7076. doi: 10.1371/journal.pone.0007076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Ghosh Z, Wilson KD, Wu Y, Hu S, Quertermous T, Wu JC. Persistent donor cell gene expression among human induced pluripotent stem cells contributes to differences with human embryonic stem cells. PLoS One. 2010;5(2 ):e8975. doi: 10.1371/journal.pone.0008975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Heng JC, Feng B, Han J, Jiang J, Kraus P, Ng JH, Orlov YL, Huss M, Yang L, Lufkin T, Lim B, Ng HH. The nuclear receptor Nr5a2 can replace Oct4 in the reprogramming of murine somatic cells to pluripotent cells. Cell Stem Cell. 2010;6(2 ):167–174. doi: 10.1016/j.stem.2009.12.009. [DOI] [PubMed] [Google Scholar]
- 169.O'Malley J, Woltjen K, Kaji K. New strategies to generate induced pluripotent stem cells. Curr. Opin. Biotechnol. 2009;20(5 ):516–521. doi: 10.1016/j.copbio.2009.09.005. [DOI] [PubMed] [Google Scholar]
- 170.Huangfu D, Osafune K, Maehr R, Guo W, Eijkelenboom A, Chen S, Muhlestein W, Melton DA. Induction of pluripotent stem cells from primary human fibroblasts with only Oct4 and Sox2. Nat. Biotechnol. 2008;26(11 ):1269–1275. doi: 10.1038/nbt.1502. [DOI] [PubMed] [Google Scholar]
- 171.Kim JB, Greber B, Arauzo-Bravo MJ, Meyer J, Park KI, Zaehres H, Scholer HR. Direct reprogramming of human neural stem cells by OCT4. Nature. 2009;461(7264 ):649–643. doi: 10.1038/nature08436. [DOI] [PubMed] [Google Scholar]
- 172.Kim JB, Sebastiano V, Wu G, Arauzo-Bravo MJ, Sasse P, Gentile L, Ko K, Ruau D, Ehrich M, van den Boom D, Meyer J, Hubner K, Bernemann C, Ortmeier C, Zenke M, Fleischmann BK, Zaehres H, Scholer HR. Oct4-induced pluripotency in adult neural stem cells. Cell. 2009;136(3 ):411–419. doi: 10.1016/j.cell.2009.01.023. [DOI] [PubMed] [Google Scholar]
- 173.Kim JB, Zaehres H, Wu G, Gentile L, Ko K, Sebastiano V, Arauzo-Bravo MJ, Ruau D, Han DW, Zenke M, Scholer HR. Pluripotent stem cells induced from adult neural stem cells by reprogramming with two factors. Nature. 2008;454(7204 ):646–650. doi: 10.1038/nature07061. [DOI] [PubMed] [Google Scholar]
- 174.Stadtfeld M, Nagaya M, Utikal J, Weir G, Hochedlinger K. Induced pluripotent stem cells generated without viral integration. Science. 2008;322(5903 ):945–949. doi: 10.1126/science.1162494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Lyssiotis CA, Foreman RK, Staerk J, Garcia M, Mathur D, Markoulaki S, Hanna J, Lairson LL, Charette BD, Bouchez LC, Bollong M, Kunick C, Brinker A, Cho CY, Schultz PG, Jaenisch R. Reprogramming of murine fibroblasts to induced pluripotent stem cells with chemical complementation of Klf4. Proc. Natl. Acad. Sci. USA. 2009;106(22 ):8912–8917. doi: 10.1073/pnas.0903860106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Gonzalez F, Barragan Monasterio M, Tiscornia G, Montserrat Pulido N, Vassena R, Batlle Morera L, Rodriguez Piza I, Izpisua Belmonte JC. Generation of mouse-induced pluripotent stem cells by transient expression of a single nonviral polycistronic vector. Proc. Natl. Acad. Sci. USA. 2009;106(22 ):8918–8922. doi: 10.1073/pnas.0901471106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Yu J, Hu K, Smuga-Otto K, Tian S, Stewart R, Slukvin II, Thomson JA. Human induced pluripotent stem cells free of vector and transgene sequences. Science. 2009;324(5928 ):797–801. doi: 10.1126/science.1172482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Okita K, Hong H, Takahashi K, Yamanaka S. Generation of mouse-induced pluripotent stem cells with plasmid vectors. Nat. Protoc. 2010;5(3 ):418–428. doi: 10.1038/nprot.2009.231. [DOI] [PubMed] [Google Scholar]
- 179.Dimos JT, Rodolfa KT, Niakan KK, Weisenthal LM, Mitsumoto H, Chung W, Croft GF, Saphier G, Leibel R, Goland R, Wichterle H, Henderson CE, Eggan K. Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons. Science. 2008;321(5893 ):1218–1221. doi: 10.1126/science.1158799. [DOI] [PubMed] [Google Scholar]
- 180.Maehr R, Chen S, Snitow M, Ludwig T, Yagasaki L, Goland R, Leibel RL, Melton DA. Generation of pluripotent stem cells from patients with type 1 diabetes. Proc. Natl. Acad. Sci. USA. 2009;106(37 ):15768–15773. doi: 10.1073/pnas.0906894106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Miyoshi N, Ishii H, Nagai K, Hoshino H, Mimori K, Tanaka F, Nagano H, Sekimoto M, Doki Y, Mori M. Defined factors induce reprogramming of gastrointestinal cancer cells. Proc. Natl. Acad. Sci. USA. 2010;107(1 ):40–45. doi: 10.1073/pnas.0912407107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Zhou Q, Brown J, Kanarek A, Rajagopal J, Melton DA. In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature. 2008;455(7213 ):627–632. doi: 10.1038/nature07314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Warren L, Manos PD, Ahfeldt T, Loh YH, Li H, Lau F, Ebina W, Mandal PK, Smith ZD, Meissner A, Daley GQ, Brack AS, Collins JJ, Cowan C, Schlaeger TM, Rossi Dc. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell. 2010 Sep 29; doi: 10.1016/j.stem.2010.08.012. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Kim K, Doi A, Wen B, Ng K, Zhao R, Cahan P, Kim J, Aryee MJ, Ji H, Ehrlich LI, Yabuuchi A, Takeuchi A, Cunniff KC, Hongguang H, McKinney-Freeman S, Naveiras O, Yoon TJ, Irizarry RA, Jung N, Seita J, Hanna J, Murakami P, Jaenisch R, Weissleder R, Orkin SH, Weissman IL, Feinberg AP, Daley GQ. Epigenetic memory in induced pluripotent stem cells. Nature. 2010;467(7313 ):285–290. doi: 10.1038/nature09342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Polo JM, Liu S, Figueroa ME, Kulalert W, Eminli S, Tan KY, Apostolou E, Stadtfeld M, Li Y, Shioda T, Natesan S, Wagers AJ, Melnick A, Evans T, Hochedlinger K. Cell type of origin influences the molecular and functional properties of mouse induced pluripotent stem cells. Nat. Biotechnol. 2010;28(8 ):848–855. doi: 10.1038/nbt.1667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Ji H, Ehrlich LI, Seita J, Murakami P, Doi A, Lindau P, Lee H, Aryee MJ, Irizarry RA, Kim K, Rossi DJ, Inlay MA, Serwold T, Karsunky H, Ho L, Daley GQ, Weissman IL, Feinberg AP. Comprehensive methylome map of lineage commitment from haematopoietic progenitors. Nature. 2010;467(7313 ):338–342. doi: 10.1038/nature09367. [DOI] [PMC free article] [PubMed] [Google Scholar]