CC BY-NC-ND 4.0 · J Neuroanaesth Crit Care 2015; 02(01): 015-022
DOI: 10.4103/2348-0548.148379
Review Article
Thieme Medical and Scientific Publishers Private Ltd.

Stem-cell therapy for neurologic diseases

Shilpa Sharma
1   Department of Pediatric Surgery, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
,
Devendra K. Gupta
1   Department of Pediatric Surgery, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
› Author Affiliations
Further Information

Address for correspondence:

Dr. Shilpa Sharma
Department of Pediatric Surgery
All India Institute of Medical Sciences, Ansari Nagar, Room no 4001, PC Block, New Delhi - 110 029
India   

Publication History

Publication Date:
05 May 2018 (online)

 

Abstract

With the advent of research on stem cell therapy for various diseases, an important need was felt in the field of neurological diseases. While congenital lesion may not be amenable to stem cell therapy completely, there is a scope of partial improvement in the lesions and halt in further progression. Neuro degenerative lesions like Parkinson's disease, multiple sclerosis and amyotrophic lateral sclerosis have shown improvement with stem cell therapy. This article reviews the available literature and summarizes the current evidence in the various neurologic diseases amenable to stem cell therapy, the plausible mechanism of action, ethical concerns with insights into the future of stem cell therapy.


#

INTRODUCTION

Ongoing research on stem cell therapy for various diseases has motivated the search for a cure in the arena of neurological diseases. This is one field where medicine has little hope for recovery and the patient might be incapacitated for life, waiting for some miraculous cure. Various basic scientists have been working on the role of stem cells in different neurological diseases hoping to find a breakthrough. Here we review the available literature, current evidence in various neurologic diseases amenable to stem cell therapy, try to explain the plausible mechanism of action and discuss the ethical concerns with insights into the future of stem cell treatment.


#

WHAT CAN STEM CELL OFFER?

The neurological diseases may be grossly classified as congenital or acquired lesions. The acquired lesions may result from hypoxic, traumatic injury, and infective or degenerative lesions. While the neurological deficits from congenital lesions may not be completely amenable to any sort of medical therapy, stem cells may offer a regeneration of neurons from the place where the growth has halted. Neurodegenerative disorders like Parkinson’s disease, multiple sclerosis and amyotrophic lateral sclerosis may be halted or slowed in progression by the use of stem cells. Scientists have been working on the process of utilizing stem cells to replace or replenish the damaged cells.[1] [2] [3] [4] The recent advances in cultivation of human embryonic stem cells and the recognition of plasticity in the human nervous system have spawned new ideations into the possibility of using versatile stem or stem-like cells to treat neurological conditions.[5] Though at present, very little experimental work has the potential to reach the phase 3 clinical trial level, there is substantial evidence to believe that stem cells can act as a support environment for survival or recovery of damaged neurons by reducing inflammation and helping remyelination. Stem cells have the ability to produce neurotrophic or immunosuppressive factors thus forming a favorable environment for brain tissue repair and long-term survival of transplanted cells in the central nervous system.[6] In animal studies, benefits of stem cell-mediated gene transfer of therapeutic genes such as neurotrophic factors and enzymes have been demonstrated.[7]


#

WHAT KIND OF STEM CELLS CAN BE USED?

Neurons and glial cells have successfully been generated from stem cells. There are fundamental and realistic differences between grafts of primary neural cells and the transplantation of in vitro expanded neural stem cells (NSCs).[8] [9] [10] A variety of stem cells have been explored as therapeutic options for treating many neurologic diseases.

These include

  • Pluripotent stem cells like embryonic stem cells and induced pluripotent stem cells

  • Multipotent adult stem cells like fetal brain tissue, fetal spinal cord derived tissue, NSCs or neural progenitor cells, and mesenchymal stem cells from various sources

  • Autologous bone marrow derived mononuclear cells

  • Adipose tissue-derived stem cells

  • Umbilical cord stem cells.

A fully functional neuron cell derived from a stem cell should ideally be electrically excitable, have the ability to release the appropriate neurotransmitter and form neural structures like processes and synapses, before it can be justified in offering a complete therapeutic treatment for neurological diseases. Pluripotent cells possess the capacity to give rise to all three germ layers, while multipotent cells give rise to more restricted lineages. NSCs are capable of differentiating to cell types within a neural lineage and may be derived directly from fetal or adult neural tissue, or differentiation of embryonic stem cells via cell culture manipulation.[11] [12] Mesenchymal stem cells are derived from adult bone marrow and can transdifferentiate to a neural lineage.[13]

Laboratory work has demonstrated that embryonic stem cells induced pluripotent stem cells and mesenchymal stem cells may help to generate neuron-like cells, but these cells are lacking in the functional aspects like proper fiber outgrowth and integration into the host neuronal networks. Induced pluripotent stem cells can be generated from autologous somatic tissue such as fibroblasts and reprogrammed into embryonic stem-like cells by the addition of transcription factors like Oct 3/4, Klf, Sox 2, and c-Myc.[14] Reprogramming of fibroblasts has been accomplished successfully by using various combinations of factors delivered by vector, virus, protein, or RNA-mediated approaches.[15]


#

NEUROLOGICAL DISEASES AMENABLE TO STEM CELL THERAPY

Neurological diseases that are of a degenerative nature characterized by the loss of neurons in the brain or spinal cord may be amenable to stem cell therapy. Alzheimer’s and Huntington’s disease result in widespread loss of neurons in the brain, while Parkinson’s disease involves the specific and localized loss of dopaminergic neurons in the substantia nigra, amyotrophic lateral sclerosis and spinal muscular atrophy involve the degeneration and loss of motor neurons in the brainstem and spinal cord. These are summarized in [Table 1]. The authors have experience of using stem cell therapy for patients of meningomyelocele, poliomyelitis, cerebral palsy, neurogenic bowel, neurogenic bladder, and amyotrophic lateral sclerosis.[16] Stem cell mobilization has benefited our patients of cerebral palsy, spinal muscular atrophy, and Duchenne muscular dystrophy.

Table 1

Neurological diseases amenable to stem cell therapy

Parkinson's disease

Alzheimer's disease

Huntington's disease

Multiple sclerosis

Amyotrophic lateral sclerosis

Stroke

Cerebral palsy

Head injury

Spinal cord injury

Spinal muscular atrophy

Neurological deficits in meningomyelocoele

Neurological deficits in poliomyelitis

Duchenne muscular dystrophy


#

MECHANISM OF ACTION OF CELL-BASED THERAPIES

Neurodegenerative diseases caused by a loss of neurons and glia in the brain or spinal cord have the potential to be benefitted from stem cells. In the past, in vitro approaches like oncogene-induce dimmortalization and growth-factor stimulation of naturally occurring central and peripheral nervous system stem cells have been studied.[8] The initial strategies were designed to replace dead cells in injured tissue, the potential of stem cells to migrate, secrete trophic factors, and immunomodulation allows their therapeutic use as a vehicle for gene therapy, as in Parkinson’s disease, or as immunomodulators and neuroprotectors in diseases such as multiple sclerosis.[17] It has been realized thatwe need to know much more about how to controlstem cell proliferation and differentiation into specific phenotypes, inducetheir integration into existing neural and synaptic circuits, and optimize functional recovery in animal models closely resembling the human disease.[18]

Today, the cell-based therapies are based on direct cell transfer or tissue grafts or providing environmental enrichment. In some diseases, the type of cells used may be more characterized for a particular function. The cell-based therapy for Parkinson’s or Huntington’s diseases is based on the ability of stem cells to replace lost dopaminergic mesencephalic or striatal neurons.[6] On the other hand, amyotrophic lateral sclerosis is benefitted from cellular therapies that enrich the local spinal cord environment to support the remaining motor neurons. The newly transplanted neurons need to integrate, synapse, and recapitulate a neural network similar to the one lost in disease. Alternatively, stem cells may provide environmental enrichment to support host neurons by producing neurotrophic factors, scavenging toxic factors, or creating auxiliary neural networks around affected areas.[19]

There are various strategies for environmental enrichment utilizing stem cells to provide de novo synthesis and delivery of neuroprotective growth factors at the site of disease. Growth factors such as glial-derived neurotrophic factor (GDNF), brain-derived neurotrophic factor (BDNF), insulin-like growth factor-I (IGF-I), and vascular endothelial growth factor (VEGF) are protective in neurodegenerative disease models and provide in situ environmental enrichment at the main locus of disease.[20] [21] [22] [23] [24] Other factorsaffecting the potential efficacy of cellular therapies include integration of grafted neurons, migration of engrafted cells, and the distances that axons need to extend to reach their targets. Bone marrow-derived cells developing neuronal and vascular phenotype sand aiding in repair of injured brain have been reported.[20]

Selecting the appropriate stem cell type and understanding the desired mechanism of support is the initial step in developing and translating cellular therapies to patients. [Table 2] outlines the various neurological diseases where stem cells have been used.

Table 2

Various neurological diseases amenable to stem cell treatment with the plausible mechanism of action

Disease

Symptoms

Pathophysiology

Plausible mechanism of benefit with stem cell therapy

GABA = Gamma-aminobutyric acid, GDNF = Glial-derived neurotrophic factor, BDNF = Brain-derived neurotrophic factor, IGF-I = Insulin-like growth factor-I, and VEGF = Vascular endothelial growth factor, SMA = Spinal muscular atrophy, NGF = Nerve growth factor, iPS = Induced pluripotent stem

Alzheimer's disease[25] [26] [27] [28] [29]

Memory loss and cognitive decline

Widespread loss of neurons and synaptic contacts throughout the cortex, hippocampus, amygdala, and basal forebrain

Regulation of neurotransmitter activity

Targeting the cholinergic system to provide environmental enrichment

Enhance neurogenesis

Replace lost neurons

Enhancing BDNF levels

Nerve growth factor production. Genetically engineered patient fibroblasts that produce NGF. Combination of above

Parkinson's disease[30] [31] [32] [33] [34] [35]

Severe motor deficits, tremors, muscle rigidity, and unstable gait and posture

Progressive loss of dopaminergic neurons in the substantia nigra

Replacement of lost dopaminergic neurons

Foetal ventral midbrain tissue transplant

Embryonic stem cells

Grafting both embryonic and mesenchymal stem cells-derived dopaminergic neurons

Patient-specific dopaminergic neurons using induced pluripotent stem cells.

Environmental enrichment may also support existing dopaminergic neurons and slow or prevent further degeneration.

Growth factor therapy through direct delivery or viral-based systems

Transplantation of mesenchymal and neural stem cells engineered to produce growth factors such as BDNF, VEGF, GDNF and IGF-I in situ

Combination of cellular replacement and environmental enrichment

Huntington's disease[36] [37] [38] [39] [40] [41] [42] [43]

Involuntary motor activity, dementia, personality changes, and cognitive impairment associated with the progressive loss of medium spiny neurons

Autosomal dominant polyglutamine disease caused by the accumulation of CAG repeats in the Huntingingene. Loss of these GABAergic neurons in the striatum is also accompanied by degeneration in the cortex, brain stem and hippocampus

Foetal tissue grafting using the whole ganglionic eminence as an optimal source of mesenchymal stem cells

Transplantation of neural cells and striatal grafts into rodent models demonstrated that mesenchymal stem cells integrate and form circuitry in the host Striatal injections of neural stem cells demonstrate incorporation as well as migration to secondary sites associated with disease

MSC in the brain promote endogenous neuronal growth, encourage synaptic connection from damaged neurons, decrease apoptosis, reduce levels of free radicals, and regulate inflammation

Environmental enrichment with neural stem cells expressing GDNF protected neurons and promoted functional recovery

Amyotrophic lateral sclerosis[44] [45] [46]

Loss of coordination and muscle strength with transition to complete loss of muscle control

Death typically results from respiratory failure within 2-5 years of diagnosis

Involving the degeneration and loss of motor neurons

Mesenchymal and neural stem cells

Environmental enrichment via GDNF, VEGF and IGF-I

Distal production of GDNF in muscle protects neuromuscular junctions and promotes motor neuron protection, likely by retrograde transport

Environmental enrichment to motor neurons and neuromuscular junctions

Foetal spinal cord-derived neural stem cells

Cellular therapies provide both an integrating neural component and environmental enrichment to support and protect motor neuronsfrom degeneration

Spinal muscular atrophy[47] [48]

SMA type I early onset severe muscle weakness and fatality within 2 years

Selective loss of motor neurons (mutation or loss of SMN1gene, alternative splicing variants encoded by SMN2 gene)

Grafting of embryonic stem cells-derived neural stem cells

Multiple sclerosis

Physical, mood disorders, and depression

Autoimmune chronic inflammatory disease of the central nervous system disease, results in the death of oligodendrocytes

Trials with haemopoietic stem cells, mesenchymal stem cells, neural stem cells, and oligodendrocyte precursor cells

Neurologic deficits in Meningomyelocele[16]

Lower limb neurologic deficit

Neurogenic bladder

Neurogenic bowel

A congenital defect in which the nerve roots are entrappedin a meningocele sac

Epidural injection of autologous bone marrow derived mononuclear cells may provide environmental enrichment and help the detethered nerve roots to regenerate

Stroke[49] [50] [51] [52] [53] [54] [55]

Sudden onset of neurological deficits

Usually a result of a vascular event

Foetal neural stem cells

Embryonic stem cells

Adult tissue sources

Bone marrow-derived stem cells, Umbilical cord blood-derived haemopoietic stem cells

Mesenchymal stem cells

Multipotent adult progenitor cells

Spinal cord injury[56] [57]

Neurological deficits following trauma

Trauma to spinal cord

Foetal neural stem cells

Embryonic stem cells

Adult tissue sources

Bone marrow-derived stem cells

Umbilical cord blood-derived stem cells

Mesenchymal stem cells

iPS cells

Head injury[58] [59] [60] [61]

Neurological deficits following trauma

Trauma to brain and skull

Exogenous trophic factor administration

Transplantation of neural progenitor cells

Adult tissue sources

Bone marrow-derived stem cells

Umbilical cord blood-derived stem cells

Mesenchymal stem cells

iPS cells

Perinatal brain injury[62] [63]

Neurological deficits following hypoxic trauma during and after birth

Perinatal hypoxic injury

Neural stem cells

Embryonic stem cells

Mesenchymal stem cells

Umbilical cord stem cells

iPS cells


#

ROUTES OF ADMINISTRATION OF CELL-BASED THERAPIES

It has been realized that an appropriate selection of the type of stem cells based on the kind of neurologic dysfunction is required to achieve optimal therapeutic efficacy.[2] However, there is a paucity of suitable cell types for appropriate cell replacement therapies. The main aim while administering cell-based therapies is to ensure that the cells reach the desired site of action. The various routes that have been used for neurological disorders include:

  • Intracerebral via cell transplants in the brain

  • Intrastriatal, fetal neural transplants

  • Intravenous route

  • Intra-arterial via the cerebral artery

  • Intrathecal

  • Lumbar puncture direct lumbar and cervical intraspinal injections

  • Intracisternal

  • Stem cell mobilization approaches in which endogenous stem and progenitor cells are mobilized by cytokines such as granulocyte colony stimulatory factor (GCSF) or chemokines such as stromal cell-derived factor-1 (SDF-1)

  • Trophic and growth factor support, such as delivering BDNF or GDNF into the brain to support injured neurons.

The various approaches may beused in combinations to maximize the efficiency. Recent advances include research on use of three-dimensional substrate-adherent embryonic stem cell-derived neural aggregates to enhance the efficacy of treatment.[64] The field of nanotechnology has been used to tag markers like superparamagnetic iron oxide nanoparticle with stem cells as a therapy for brain diseases and monitor the migration of stem cell in affected regions of the brain.[65]


#

ETHICAL CONCERNS

During the beginning of the stem cell revolution, animal tissue was widely used to conduct experiments; and thus look for xenotransplants as a possible cure for some diseases.[66] However, this field has not gained momentum particularly due to ethical issues and potential life-threatening complications.[67] [68] [69]

Though clinical improvement has been observed in cases of Parkinson and Huntington disease patients transplanted with freshly isolated fetal brain tissue, but there are ethical issues in this kind of restorative treatment.[6] [70] [71] The first concern is the use of human fetal tissue for treatment purpose and the very fact that this may be misused by planning pregnancy and termination only for the use of such tissue. Also the amounts of tissue obtained may not be adequate for full treatment, thus exposing the patient to tissues from multiple sources. The second concern is the need of use of immunosuppression and potential side effects like spread of viral infections and the risk of development of malignancy in future. Other issues include graft overgrowth and presence of non-neuronal cells within the graft. Side effects like dyskinesias have been reported following cell transplants.

There is no immunosuppression for cellular therapies derived from autologous sources. However, autologous cells may be less desirable when dealing with genetic diseases because the cells may possess the same genetic predisposition to disease.

The ethical concerns for use of human embryonic stem cells are equally significant as the use of the embryo for medicinal purpose is not approved ethically. Though autologous stem cells are less potent and may have a short duration of effect, they are free from ethical concerns. Recently, the induced pluripotent stem cell shave gained importance due to their major potential advantages as patient-specific neuroblasts are suitable for transplantation, avoid immune reactions, and can be produced without the use of embryonic stem cells.[72] [73] [74] [75] [76] The use of trophic factors has also gained momentum, but unless the benefits are shown to outweigh the risks and the scientific basis of mechanism of action is established, their use would continue to be in the trial phases. Further research will definitely establish the utility in times to come.


#

FUTURE DIRECTIONS

Neurodegenerative diseases area burden in the society as they incapacitate the human brain and nervous system to a great extent. As the exact scientific pathophysiology of the various diseases still remains a mystery, we are yet to find a permanent treatment. Though there are few medications for most of the diseases, the treatment mostly remains symptomatic and one has to bear with the side effects of lifelong medications.

The stem cell revolution has been looked upon as a breakthrough miraculous treatment as it may have the potential both to stop the further downhill course of the disease and also to reverse the symptomatology especially if given early in the onset of disease.

Cellular therapies and environmental enrichment offer great promise for the treatment of these diseases, and research progress to date supports the utilization of stem cells to offer cellular replacement and/or provide environmental enrichment to attenuate neurodegeneration.[77] [78] [79] In diseases where specific subpopulations of cells or widespread neuronal loss are present, cellular replacement may reproduce or stabilize neuronal networks. In addition, they may provide neurotrophic support to remaining cells or prevent the production or accumulation of toxic factors that harm neurons. In many cases, cellular therapies provide beneficial effects through both mechanisms.

However, there are still many hurdles in reaching the goal of stem cell therapy for neurological diseases. These include:

  • Lack of knowledge about the true mechanisms of these diseases

  • It is still uncertain what kind of stem cells are an ideal source for cellular grafts for a particular disease

  • The mechanism by which stem cells transplantation leads to an enhanced functional recovery and structural reorganization is not unraveled

  • The molecular mechanisms underlying the critical steps in cell-based repair are still not known

  • Angiogenesis and neurogenesis are coupled in the nervous system. Thus, both mechanisms might need a combination of therapies to be effective

  • The optimal timing for intervention and patient selection has to be identified as the mechanism of action may not be the same in different stages of the disease

  • The optimal delivery route and cell dose has to be evaluated by repeated studies

  • The relevant clinical end points and monitoring for effectiveness have to be evaluated.

The way forward should include sound research on both the basic and preclinical settings for each neurological disease. There is a need to improve standardization and develop safe master celllines derived from human embryonic stem cells, induced pluripotent stem cells, and NSCs. Cell therapy derived from these sources are expected to become available for cell replacementor production of trophic, anti-inflammatory, and restorative factors for therapeutic use within a few years.[80] Apart from cell replacement therapy, other strategies that need to be explored include enhancement of endogenous plasticity and recruitment of endogenous neurogenesis.[81] The most immediate impact on patients will be achieved by making use of the trophic support capability of stem cell therapy.[20]

The stimulation of endogenous adult stem cell-mediated repair mechanisms in the brain might offer new avenues for therapy. The stem cell therapy approach to neurological disease will have to be scalable, easily reproducible, and cost effective so that it is easily available to those in need. The future of stem cell therapy has still many unread chapters that need to be explored by scientists so that the maximum potential of this new emerging field of biology is utilized for the benefit of mankind.


#
#

No conflict of interest has been declared by the author(s).

  • REFERENCES

  • 1 Gore A, Li Z, Fung HL, Young JE, Agarwal S, Antosiewicz-Bourget J. et al. Somatic coding mutations in human induced pluripotent stem cells. Nature 2011; 471: 63-7
  • 2 Yoo J, Kim HS, Hwang DY. Stem cells as promising therapeutic options for neurological disorders. J Cell Biochem 2013; 114: 743-53
  • 3 Kim SU, de Vellis J. Stem cell-based cell therapy in neurological diseases: A review. J Neurosci Res 2009; 87: 2183-200
  • 4 Hou L, Hong T. Stem cells and neurodegenerative diseases. Sci China C Life Sci 2008; 51: 287-94
  • 5 Holden C. Neuroscience. Versatile cells against intractable diseases. Science 2002; 297: 500-2
  • 6 Lescaudron L, Naveilhan P, Neveu I. The use of stem cells in regenerative medicine for Parkinson's and Huntington's diseases. Curr Med Chem 2012; 19: 6018-35
  • 7 Kim SU, Lee HJ, Kim YB. Neural stem cell-based treatment for neurodegenerative diseases. Neuropathology 2013; 33: 491-504
  • 8 Baetge EE. Neural stem cells for CNS transplantation. Ann N Y Acad Sci 1993; 695: 285-91
  • 9 Conti L, Reitano E, Cattaneo E. Neural stem cell systems: Diversities and properties after transplantation in animal models of diseases. Brain Pathol 2006; 16: 143-54
  • 10 Yan J, Xu L, Welsh AM, Hatfield G, Hazel T, Johe K. et al. Extensive neuronal differentiation of human neural stem cell grafts in adult rat spinal cord. PLoS Med 2007; 4: e39
  • 11 Wichterle H, Lieberam I, Porter JA, Jessell TM. Directed differentiation of embryonic stem cells into motor neurons. Cell 2002; 110: 385-97
  • 12 Gaspard N, Vanderhaeghen P. Mechanisms of neural specification from embryonic stem cells. Curr Opin Neurobiol 2010; 20: 37-43
  • 13 Satija NK, Singh VK, Verma YK, Gupta P, Sharma S, Afrin F. et al. Mesenchymal stem cell-based therapy: A new paradigm in regenerative medicine. J Cell Mol Med 2009; 13: 4385-402
  • 14 Yamanaka S. Induction of pluripotent stem cells from mouse fibroblasts by four transcription factors. Cell Prolif 2008; 41 Suppl (Suppl. 01) 51-6
  • 15 Yakubov E, Rechavi G, Rozenblatt S, Givol D. Reprogramming of human fibroblasts to pluripotent stem cells using mRNA of four transcription factors. Biochem Biophys Res Commun 2010; 394: 189-93
  • 16 Sharma S, Gupta DK, Venugopal P, Kumar L, Dattagupta S, Arora MK. Therapeutic use of stem cells in congenital anomalies: A pilot study. J Indian Assoc Pediatr Surg 2006; 11: 211-7
  • 17 Costa C, Comabella M, Montalban X. Stem cell-based treatment of neurologic diseases. Med Clin (Barc) 2012; 139: 208-14
  • 18 Lindvall O, Kokaia Z, Martinez-Serrano A. Stem cell therapy for human neurodegenerative disorders-how to make it work. Nat Med 2004; 10: S42-50
  • 19 Lunn JS, Sakowski SA, Feldman EL. Stem cell technology for neurodegenerative diseases. Ann Neurol 2011; 70: 353-61
  • 20 Hess DC, Borlongan CV. Stem cells and neurological diseases. Cell Prolif 2008; 41 Suppl (Suppl. 01) 94-114
  • 21 Suzuki M, Svendsen CN. Combining growth factor and stem cell therapy for amyotrophic lateral sclerosis. Trends Neurosci 2008; 31: 192-8
  • 22 Lunn JS, Hefferan MP, Marsala M, Feldman EL. Stem cells: Comprehensive treatments for amyotrophic lateral sclerosis in conjunction with growth factor delivery. Growth Factors 2009; 27: 133-40
  • 23 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: 753-62
  • 24 Ebert AD, Barber AE, Heins BM, Svendsen CN. Ex vivo delivery of GDNF maintains motor function and prevents neuronal loss in a transgenic mouse model of Huntington's disease. Exp Neurol 2010; 224: 155-62
  • 25 Tuszynski MH, Thal L, Pay M, Salmon DP, UHS. Bakay R. et al. A phase 1 clinical trial of nerve growth factor gene therapy for Alzheimer disease. Nat Med 2005; 11: 551-5
  • 26 Drapeau E, Nora Abrous D. Stem cell review series: Role of neurogenesis in age-related memory disorders. Aging Cell 2008; 7: 569-89
  • 27 Li G, Peskind ER, Millard SP, Chi P, Sokal I, Yu CE. et al. Cerebrospinal fluid concentration of brain-derived neurotrophic factor and cognitive function in non-demented subjects. PLoS One 2009; 4: e5424
  • 28 Tuszynski MH. Nerve growth factor gene therapy in Alzheimer disease. Alzheimer Dis Assoc Disord 2007; 21: 179-89
  • 29 Shruster A, Melamed E, Offen D. Neurogenesis in the aged and neurodegenerative brain. Apoptosis 2010; 15: 1415-21
  • 30 Morizane A, Darsalia V, Guloglu MO, Hjalt T, Carta M, Li JY. et al. A simple method for large-scale generation of dopamine neurons from human embryonic stem cells. J Neurosci Res 2010; 88: 3467-78
  • 31 Cui YF, Hargus G, Xu JC, Schmid JS, Shen YQ, Glatzel M. et al. Embryonic stem cell-derived L1 overexpressing neural aggregates enhance recovery in Parkinsonian mice. Brain 2010; 133: 189-204
  • 32 Cooper O, Hargus G, Deleidi M, Blak A, Osborn T, Marlow E. et al. Differentiation of human ES and Parkinson's disease iPS cells into ventral midbrain dopaminergic neurons requires a high activity form of SHH, FGF8a and specific regionalization by retinoic acid. Mol Cell Neurosci 2010; 45: 258-66
  • 33 Xiong N, Zhang Z, Huang J, Chen C, Zhang Z, Jia M. et al. VEGF-expressing human umbilical cord mesenchymal stem cells, an improved therapy strategy for Parkinson's disease. Gene Ther 2011; 18: 394-402
  • 34 Garbayo E, Montero-Menei CN, Ansorena E, Lanciego JL, Aymerich MS, Blanco-Prieto MJ. Effective GDNF brain delivery using microspheres--a promising strategy for Parkinson's disease. J Control Release 2009; 135: 119-26
  • 35 Ebert AD, Beres AJ, Barber AE, Svendsen CN. Human neural progenitor cells over-expressing IGF-1 protect dopamine neurons and restore function in a rat model of Parkinson's disease. Exp Neurol 2008; 209: 213-23
  • 36 Clelland CD, Barker RA, Watts C. Cell therapy in Huntington disease. Neurosurg Focus 2008; 24: E9
  • 37 Kelly CM, Dunnett SB, Rosser AE. Medium spiny neurons for transplantation in Huntington's disease. Biochem Soc Trans 2009; 37: 323-8
  • 38 Nakao N, Ogura M, Nakai K, Itakura T. Embryonic striatal grafts restore neuronal activity of the globus pallidus in a rodent model of Huntington's disease. Neuroscience 1999; 88: 469-77
  • 39 Olson SD, Pollock K, Kambal A, Cary W, Mitchell GM, Tempkin J. et al. Genetically engineered mesenchymal stem cells as a proposed therapeutic for Huntington's disease. Mol Neurobiol 2012; 45: 87-98
  • 40 McBride JL, Behrstock SP, Chen EY, Jakel RJ, Siegel I, Svendsen CN. et al. Human neural stem cell transplants improve motor function in a rat model of Huntington's disease. J Comp Neurol 2004; 475: 211-9
  • 41 Maucksch C, Vazey EM, Gordon RJ, Connor B. Stem cell-based therapy for Huntington's disease. J Cell Biochem 2013; 114: 754-63
  • 42 Rosser A, Svendsen CN. Stem cells for cell replacement therapy: A therapeutic strategy for HD?. Mov Disord 2014; 29: 1446-54
  • 43 Nicoleau C, Viegas P, Peschanski M, Perrier AL. Human pluripotent stem cell therapy for Huntington's disease: Technical, immunological, and safety challenges human pluripotent stem cell therapy for Huntington's disease: Technical, immunological, and safety challenges. Neurotherapeutics 2011; 8: 562-76
  • 44 Garbuzova-Davis S, Willing AE, Zigova T, Saporta S, Justen EB, Lane JC. et al. Intravenous administration of human umbilical cord blood cells in a mouse model of amyotrophic lateral sclerosis: Distribution, migration, and differentiation. J Hematother Stem Cell Res 2003; 12: 255-70
  • 45 Suzuki M, McHugh J, Tork C, Shelley B, Hayes A, Bellantuono I. et al. Direct muscle delivery of GDNF with human mesenchymal stem cells improves motor neuron survival and function in a rat model of familial ALS. Mol Ther 2008; 16: 2002-10
  • 46 Lunn JS, Sakowski SA, Federici T, Glass JD, Boulis NM, Feldman EL. Stem cell technology for the study and treatment of motor neuron diseases. Regen Med 2011; 6: 201-13
  • 47 Corti S, Nizzardo M, Nardini M, Donadoni C, Salani S, Ronchi D. et al. Neural stem cell transplantation can ameliorate the phenotype of a mouse model of spinal muscular atrophy. J Clin Invest 2008; 118: 3316-30
  • 48 Corti S, Nizzardo M, Nardini M, Donadoni C, Salani S, Ronchi D. et al. Embryonic stem cell-derived neural stem cells improve spinal muscular atrophy phenotype in mice. Brain 2010; 133: 465-81
  • 49 Shinozuka K, Dailey T, Tajiri N, Ishikawa H, Kaneko Y, Borlongan CV. Stem cell transplantation for neuroprotection in stroke. Brain Sci 2013; 3: 239-61
  • 50 Dailey T, Metcalf C, Mosley YI, Sullivan R, Shinozuka K, Tajiri N. et al. An update on translating stem cell therapy for stroke from bench to bedside. J Clin Med 2013; 2: 220-41
  • 51 Tang Y, Yasuhara T, Hara K, Matsukawa N, Maki M, Yu G. et al. Transplantation of bone marrow-derived stem cells: A promising therapy for stroke. Cell Transplant 2007; 16: 159-69
  • 52 Sinden JD, Vishnubhatla I, Muir KW. Prospects for stem cell-derived therapy in stroke. Prog Brain Res 2012; 201: 119-67
  • 53 Mendez-Otero R, de Freitas GR, André C, de Mendonça ML, Friedrich M, Oliveira-Filho J. Potential roles of bone marrow stem cells in stroke therapy. Regen Med 2007; 2: 417-23
  • 54 Bang OY. Current status of cell therapies in stroke. Int J Stem Cells 2009; 2: 35-44
  • 55 Rosado-de-Castro PH, Pimentel-Coelho PM, da Fonseca LM, de Freitas GR, Mendez-Otero R. The rise of cell therapy trials for stroke: Review of published and registeredstudies. Stem Cells Dev 2013; 22: 2095-111
  • 56 Nandoe Tewarie RS, Hurtado A, Bartels RH, Grotenhuis A, Oudega M. Stem cell–based therapies for spinal cord injury. J Spinal Cord Med 2009; 32: 105-14
  • 57 Ronaghi M, Erceg S, Moreno-Manzano V, Stojkovic M. Challenges of stem cell therapy for spinal cord injury: Human embryonic stem cells, endogenous neural stem cells, or induced pluripotent stem cells?. Stem Cells 2010; 28: 93-9
  • 58 Richardson RM, Singh A, Sun D, Fillmore HL, Dietrich 3rd DW, Bullock MR. Stem cell biology in traumatic brain injury: Effects of injury and strategies for repair. Neurosurg 2010; 112: 1125-38
  • 59 Kim HJ, Lee JH, Kim SH. Therapeutic effects of human mesenchymal stem cells on traumatic brain injury in rats: Secretion of neurotrophic factors and inhibition of apoptosis. J Neurotrauma 2010; 27: 131-8
  • 60 Anbari F, Khalili MA, Bahrami AR, Khoradmehr A, Sadeghian F, Fesahat F. et al. Intravenous transplantation of bone marrow mesenchymal stem cells promotes neural regeneration after traumatic brain injury. Neural Regen Res 2014; 9: 919-23
  • 61 Bakhtiary M, Marzban M, Mehdizadeh M, Joghataei MT, Khoei S, Pirhajati Mahabadi V. et al. Comparison of transplantation of bone marrow stromal cells (BMSC) and stem cell mobilization by granulocyte colony stimulating factor after traumatic brain injury in rat. Iran Biomed J 2010; 14: 142-9
  • 62 Vawda R, Woodbury J, Covey M, Levison SW, Mehmet H. Stem cell therapies for perinatal brain injuries. Semin Fetal Neonatal Med 2007; 12: 259-72
  • 63 Titomanlio L, Kavelaars A, Dalous J, Mani S, El Ghouzzi V, Heijnen C. et al. Stem cell therapy for neonatal brain injury: Perspectives and challenges. Ann Neurol 2011; 70: 698-712
  • 64 Hargus G, Cui YF, Dihné M, Bernreuther C, Schachner M. In vitro generation of three-dimensional substrate-adherent embryonic stem cell-derived neural aggregates for application in animal models of neurological disorders. Curr Protoc Stem Cell Biol. 2012 Chapter 2:Unit 2D.11.
  • 65 Alvarim LT, Nucci LP, Mamani JB, Marti LC, Aguiar MF, Silva HR. et al. Therapeutics with SPION-labeled stem cells for the main diseases related to brain aging: A systematic review. Int J Nanomedicine 2014; 9: 3749-70
  • 66 Harrower TP, Barker RA. The emerging technologies of neural xenografting and stem cell transplantation for treating neurodegenerative disorders. Drugs Today (Barc) 2004; 40: 171-89
  • 67 Lo B, Parham L. Resolving ethical issues in stem cell clinical trials: The example of Parkinson disease. J Law Med Ethics 2010; 38: 257-66
  • 68 Barker RA, de Beaufort I. Scientific and ethical issues related to stem cell research and interventions inneurodegenerative disorders of the brain. Prog Neurobiol 2013; 110: 63-73
  • 69 Ostenfeld T, Svendsen CN. Recent advances in stem cell neurobiology. Adv Tech Stand Neurosurg 2003; 28: 3-89
  • 70 Dunnett SB, Rosser AE. Cell transplantation for Huntington's disease Should we continue?. Brain Res Bull 2007; 72: 132-47
  • 71 Brundin P, Barker RA, Parmar M. Neural grafting in Parkinson's disease Problems and possibilities. Prog Brain Res 2010; 184: 265-94
  • 72 Liu SP, Fu RH, Huang SJ, Huang YC, Chen SY, Chang CH. et al. Stem cell applications in regenerative medicine for neurological disorders. Cell Transplant 2013; 22: 631-7
  • 73 Perrier A, Peschanski M. How can human pluripotent stem cells help decipher and cure Huntington's disease?. Cell Stem Cell 2012; 11: 153-61
  • 74 Wijeyekoon R, Barker RA. Cell replacement therapy for Parkinson's disease. Biochim Biophys Acta 2009; 1792: 688-702
  • 75 Luo Y, Fan Y, Chen X, Yu B, Yue L, Wang D. et al. Generation of induced pluripotent stem cells from Asian patients with chronic neurodegenerative diseases. J Reprod Dev 2012; 58: 515-21
  • 76 Chen LW, Kuang F, Wei LC, Ding YX, Yung KK, Chan YS. Potential application of induced pluripotent stem cells in cell replacement therapy for Parkinson's disease. CNS Neurol Disord Drug Targets 2011; 10: 449-58
  • 77 Feng Z, Gao F. Stem cell challenges in the treatment of neurodegenerative disease. CNS Neurosci Ther 2012; 18: 142-8
  • 78 Balami JS, Fricker RA, Chen R. Stem cell therapy for ischaemic stroke: Translation from preclinical studies to clinical treatment. CNS Neurol Disord Drug Targets 2013; 12: 209-19
  • 79 Mochizuki H, Choong CJ, Yasuda T. The promises of stem cells: Stem cell therapy for movement disorders. Parkinsonism Relat Disord 2014; 20 Suppl (Suppl. 01) S128-31
  • 80 Meyer M, Jensen P, Rasmussen JZ. Stem cell therapy for neurodegenerative disorders. Ugeskr Laeger 2010; 172: 2604-7
  • 81 Trueman RC, Klein A, Lindgren HS, Lelos MJ, Dunnett SB. Repair of the CNS using endogenous and transplanted neural stem cells. Curr Top Behav Neurosci 2013; 15: 357-98

Address for correspondence:

Dr. Shilpa Sharma
Department of Pediatric Surgery
All India Institute of Medical Sciences, Ansari Nagar, Room no 4001, PC Block, New Delhi - 110 029
India   

  • REFERENCES

  • 1 Gore A, Li Z, Fung HL, Young JE, Agarwal S, Antosiewicz-Bourget J. et al. Somatic coding mutations in human induced pluripotent stem cells. Nature 2011; 471: 63-7
  • 2 Yoo J, Kim HS, Hwang DY. Stem cells as promising therapeutic options for neurological disorders. J Cell Biochem 2013; 114: 743-53
  • 3 Kim SU, de Vellis J. Stem cell-based cell therapy in neurological diseases: A review. J Neurosci Res 2009; 87: 2183-200
  • 4 Hou L, Hong T. Stem cells and neurodegenerative diseases. Sci China C Life Sci 2008; 51: 287-94
  • 5 Holden C. Neuroscience. Versatile cells against intractable diseases. Science 2002; 297: 500-2
  • 6 Lescaudron L, Naveilhan P, Neveu I. The use of stem cells in regenerative medicine for Parkinson's and Huntington's diseases. Curr Med Chem 2012; 19: 6018-35
  • 7 Kim SU, Lee HJ, Kim YB. Neural stem cell-based treatment for neurodegenerative diseases. Neuropathology 2013; 33: 491-504
  • 8 Baetge EE. Neural stem cells for CNS transplantation. Ann N Y Acad Sci 1993; 695: 285-91
  • 9 Conti L, Reitano E, Cattaneo E. Neural stem cell systems: Diversities and properties after transplantation in animal models of diseases. Brain Pathol 2006; 16: 143-54
  • 10 Yan J, Xu L, Welsh AM, Hatfield G, Hazel T, Johe K. et al. Extensive neuronal differentiation of human neural stem cell grafts in adult rat spinal cord. PLoS Med 2007; 4: e39
  • 11 Wichterle H, Lieberam I, Porter JA, Jessell TM. Directed differentiation of embryonic stem cells into motor neurons. Cell 2002; 110: 385-97
  • 12 Gaspard N, Vanderhaeghen P. Mechanisms of neural specification from embryonic stem cells. Curr Opin Neurobiol 2010; 20: 37-43
  • 13 Satija NK, Singh VK, Verma YK, Gupta P, Sharma S, Afrin F. et al. Mesenchymal stem cell-based therapy: A new paradigm in regenerative medicine. J Cell Mol Med 2009; 13: 4385-402
  • 14 Yamanaka S. Induction of pluripotent stem cells from mouse fibroblasts by four transcription factors. Cell Prolif 2008; 41 Suppl (Suppl. 01) 51-6
  • 15 Yakubov E, Rechavi G, Rozenblatt S, Givol D. Reprogramming of human fibroblasts to pluripotent stem cells using mRNA of four transcription factors. Biochem Biophys Res Commun 2010; 394: 189-93
  • 16 Sharma S, Gupta DK, Venugopal P, Kumar L, Dattagupta S, Arora MK. Therapeutic use of stem cells in congenital anomalies: A pilot study. J Indian Assoc Pediatr Surg 2006; 11: 211-7
  • 17 Costa C, Comabella M, Montalban X. Stem cell-based treatment of neurologic diseases. Med Clin (Barc) 2012; 139: 208-14
  • 18 Lindvall O, Kokaia Z, Martinez-Serrano A. Stem cell therapy for human neurodegenerative disorders-how to make it work. Nat Med 2004; 10: S42-50
  • 19 Lunn JS, Sakowski SA, Feldman EL. Stem cell technology for neurodegenerative diseases. Ann Neurol 2011; 70: 353-61
  • 20 Hess DC, Borlongan CV. Stem cells and neurological diseases. Cell Prolif 2008; 41 Suppl (Suppl. 01) 94-114
  • 21 Suzuki M, Svendsen CN. Combining growth factor and stem cell therapy for amyotrophic lateral sclerosis. Trends Neurosci 2008; 31: 192-8
  • 22 Lunn JS, Hefferan MP, Marsala M, Feldman EL. Stem cells: Comprehensive treatments for amyotrophic lateral sclerosis in conjunction with growth factor delivery. Growth Factors 2009; 27: 133-40
  • 23 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: 753-62
  • 24 Ebert AD, Barber AE, Heins BM, Svendsen CN. Ex vivo delivery of GDNF maintains motor function and prevents neuronal loss in a transgenic mouse model of Huntington's disease. Exp Neurol 2010; 224: 155-62
  • 25 Tuszynski MH, Thal L, Pay M, Salmon DP, UHS. Bakay R. et al. A phase 1 clinical trial of nerve growth factor gene therapy for Alzheimer disease. Nat Med 2005; 11: 551-5
  • 26 Drapeau E, Nora Abrous D. Stem cell review series: Role of neurogenesis in age-related memory disorders. Aging Cell 2008; 7: 569-89
  • 27 Li G, Peskind ER, Millard SP, Chi P, Sokal I, Yu CE. et al. Cerebrospinal fluid concentration of brain-derived neurotrophic factor and cognitive function in non-demented subjects. PLoS One 2009; 4: e5424
  • 28 Tuszynski MH. Nerve growth factor gene therapy in Alzheimer disease. Alzheimer Dis Assoc Disord 2007; 21: 179-89
  • 29 Shruster A, Melamed E, Offen D. Neurogenesis in the aged and neurodegenerative brain. Apoptosis 2010; 15: 1415-21
  • 30 Morizane A, Darsalia V, Guloglu MO, Hjalt T, Carta M, Li JY. et al. A simple method for large-scale generation of dopamine neurons from human embryonic stem cells. J Neurosci Res 2010; 88: 3467-78
  • 31 Cui YF, Hargus G, Xu JC, Schmid JS, Shen YQ, Glatzel M. et al. Embryonic stem cell-derived L1 overexpressing neural aggregates enhance recovery in Parkinsonian mice. Brain 2010; 133: 189-204
  • 32 Cooper O, Hargus G, Deleidi M, Blak A, Osborn T, Marlow E. et al. Differentiation of human ES and Parkinson's disease iPS cells into ventral midbrain dopaminergic neurons requires a high activity form of SHH, FGF8a and specific regionalization by retinoic acid. Mol Cell Neurosci 2010; 45: 258-66
  • 33 Xiong N, Zhang Z, Huang J, Chen C, Zhang Z, Jia M. et al. VEGF-expressing human umbilical cord mesenchymal stem cells, an improved therapy strategy for Parkinson's disease. Gene Ther 2011; 18: 394-402
  • 34 Garbayo E, Montero-Menei CN, Ansorena E, Lanciego JL, Aymerich MS, Blanco-Prieto MJ. Effective GDNF brain delivery using microspheres--a promising strategy for Parkinson's disease. J Control Release 2009; 135: 119-26
  • 35 Ebert AD, Beres AJ, Barber AE, Svendsen CN. Human neural progenitor cells over-expressing IGF-1 protect dopamine neurons and restore function in a rat model of Parkinson's disease. Exp Neurol 2008; 209: 213-23
  • 36 Clelland CD, Barker RA, Watts C. Cell therapy in Huntington disease. Neurosurg Focus 2008; 24: E9
  • 37 Kelly CM, Dunnett SB, Rosser AE. Medium spiny neurons for transplantation in Huntington's disease. Biochem Soc Trans 2009; 37: 323-8
  • 38 Nakao N, Ogura M, Nakai K, Itakura T. Embryonic striatal grafts restore neuronal activity of the globus pallidus in a rodent model of Huntington's disease. Neuroscience 1999; 88: 469-77
  • 39 Olson SD, Pollock K, Kambal A, Cary W, Mitchell GM, Tempkin J. et al. Genetically engineered mesenchymal stem cells as a proposed therapeutic for Huntington's disease. Mol Neurobiol 2012; 45: 87-98
  • 40 McBride JL, Behrstock SP, Chen EY, Jakel RJ, Siegel I, Svendsen CN. et al. Human neural stem cell transplants improve motor function in a rat model of Huntington's disease. J Comp Neurol 2004; 475: 211-9
  • 41 Maucksch C, Vazey EM, Gordon RJ, Connor B. Stem cell-based therapy for Huntington's disease. J Cell Biochem 2013; 114: 754-63
  • 42 Rosser A, Svendsen CN. Stem cells for cell replacement therapy: A therapeutic strategy for HD?. Mov Disord 2014; 29: 1446-54
  • 43 Nicoleau C, Viegas P, Peschanski M, Perrier AL. Human pluripotent stem cell therapy for Huntington's disease: Technical, immunological, and safety challenges human pluripotent stem cell therapy for Huntington's disease: Technical, immunological, and safety challenges. Neurotherapeutics 2011; 8: 562-76
  • 44 Garbuzova-Davis S, Willing AE, Zigova T, Saporta S, Justen EB, Lane JC. et al. Intravenous administration of human umbilical cord blood cells in a mouse model of amyotrophic lateral sclerosis: Distribution, migration, and differentiation. J Hematother Stem Cell Res 2003; 12: 255-70
  • 45 Suzuki M, McHugh J, Tork C, Shelley B, Hayes A, Bellantuono I. et al. Direct muscle delivery of GDNF with human mesenchymal stem cells improves motor neuron survival and function in a rat model of familial ALS. Mol Ther 2008; 16: 2002-10
  • 46 Lunn JS, Sakowski SA, Federici T, Glass JD, Boulis NM, Feldman EL. Stem cell technology for the study and treatment of motor neuron diseases. Regen Med 2011; 6: 201-13
  • 47 Corti S, Nizzardo M, Nardini M, Donadoni C, Salani S, Ronchi D. et al. Neural stem cell transplantation can ameliorate the phenotype of a mouse model of spinal muscular atrophy. J Clin Invest 2008; 118: 3316-30
  • 48 Corti S, Nizzardo M, Nardini M, Donadoni C, Salani S, Ronchi D. et al. Embryonic stem cell-derived neural stem cells improve spinal muscular atrophy phenotype in mice. Brain 2010; 133: 465-81
  • 49 Shinozuka K, Dailey T, Tajiri N, Ishikawa H, Kaneko Y, Borlongan CV. Stem cell transplantation for neuroprotection in stroke. Brain Sci 2013; 3: 239-61
  • 50 Dailey T, Metcalf C, Mosley YI, Sullivan R, Shinozuka K, Tajiri N. et al. An update on translating stem cell therapy for stroke from bench to bedside. J Clin Med 2013; 2: 220-41
  • 51 Tang Y, Yasuhara T, Hara K, Matsukawa N, Maki M, Yu G. et al. Transplantation of bone marrow-derived stem cells: A promising therapy for stroke. Cell Transplant 2007; 16: 159-69
  • 52 Sinden JD, Vishnubhatla I, Muir KW. Prospects for stem cell-derived therapy in stroke. Prog Brain Res 2012; 201: 119-67
  • 53 Mendez-Otero R, de Freitas GR, André C, de Mendonça ML, Friedrich M, Oliveira-Filho J. Potential roles of bone marrow stem cells in stroke therapy. Regen Med 2007; 2: 417-23
  • 54 Bang OY. Current status of cell therapies in stroke. Int J Stem Cells 2009; 2: 35-44
  • 55 Rosado-de-Castro PH, Pimentel-Coelho PM, da Fonseca LM, de Freitas GR, Mendez-Otero R. The rise of cell therapy trials for stroke: Review of published and registeredstudies. Stem Cells Dev 2013; 22: 2095-111
  • 56 Nandoe Tewarie RS, Hurtado A, Bartels RH, Grotenhuis A, Oudega M. Stem cell–based therapies for spinal cord injury. J Spinal Cord Med 2009; 32: 105-14
  • 57 Ronaghi M, Erceg S, Moreno-Manzano V, Stojkovic M. Challenges of stem cell therapy for spinal cord injury: Human embryonic stem cells, endogenous neural stem cells, or induced pluripotent stem cells?. Stem Cells 2010; 28: 93-9
  • 58 Richardson RM, Singh A, Sun D, Fillmore HL, Dietrich 3rd DW, Bullock MR. Stem cell biology in traumatic brain injury: Effects of injury and strategies for repair. Neurosurg 2010; 112: 1125-38
  • 59 Kim HJ, Lee JH, Kim SH. Therapeutic effects of human mesenchymal stem cells on traumatic brain injury in rats: Secretion of neurotrophic factors and inhibition of apoptosis. J Neurotrauma 2010; 27: 131-8
  • 60 Anbari F, Khalili MA, Bahrami AR, Khoradmehr A, Sadeghian F, Fesahat F. et al. Intravenous transplantation of bone marrow mesenchymal stem cells promotes neural regeneration after traumatic brain injury. Neural Regen Res 2014; 9: 919-23
  • 61 Bakhtiary M, Marzban M, Mehdizadeh M, Joghataei MT, Khoei S, Pirhajati Mahabadi V. et al. Comparison of transplantation of bone marrow stromal cells (BMSC) and stem cell mobilization by granulocyte colony stimulating factor after traumatic brain injury in rat. Iran Biomed J 2010; 14: 142-9
  • 62 Vawda R, Woodbury J, Covey M, Levison SW, Mehmet H. Stem cell therapies for perinatal brain injuries. Semin Fetal Neonatal Med 2007; 12: 259-72
  • 63 Titomanlio L, Kavelaars A, Dalous J, Mani S, El Ghouzzi V, Heijnen C. et al. Stem cell therapy for neonatal brain injury: Perspectives and challenges. Ann Neurol 2011; 70: 698-712
  • 64 Hargus G, Cui YF, Dihné M, Bernreuther C, Schachner M. In vitro generation of three-dimensional substrate-adherent embryonic stem cell-derived neural aggregates for application in animal models of neurological disorders. Curr Protoc Stem Cell Biol. 2012 Chapter 2:Unit 2D.11.
  • 65 Alvarim LT, Nucci LP, Mamani JB, Marti LC, Aguiar MF, Silva HR. et al. Therapeutics with SPION-labeled stem cells for the main diseases related to brain aging: A systematic review. Int J Nanomedicine 2014; 9: 3749-70
  • 66 Harrower TP, Barker RA. The emerging technologies of neural xenografting and stem cell transplantation for treating neurodegenerative disorders. Drugs Today (Barc) 2004; 40: 171-89
  • 67 Lo B, Parham L. Resolving ethical issues in stem cell clinical trials: The example of Parkinson disease. J Law Med Ethics 2010; 38: 257-66
  • 68 Barker RA, de Beaufort I. Scientific and ethical issues related to stem cell research and interventions inneurodegenerative disorders of the brain. Prog Neurobiol 2013; 110: 63-73
  • 69 Ostenfeld T, Svendsen CN. Recent advances in stem cell neurobiology. Adv Tech Stand Neurosurg 2003; 28: 3-89
  • 70 Dunnett SB, Rosser AE. Cell transplantation for Huntington's disease Should we continue?. Brain Res Bull 2007; 72: 132-47
  • 71 Brundin P, Barker RA, Parmar M. Neural grafting in Parkinson's disease Problems and possibilities. Prog Brain Res 2010; 184: 265-94
  • 72 Liu SP, Fu RH, Huang SJ, Huang YC, Chen SY, Chang CH. et al. Stem cell applications in regenerative medicine for neurological disorders. Cell Transplant 2013; 22: 631-7
  • 73 Perrier A, Peschanski M. How can human pluripotent stem cells help decipher and cure Huntington's disease?. Cell Stem Cell 2012; 11: 153-61
  • 74 Wijeyekoon R, Barker RA. Cell replacement therapy for Parkinson's disease. Biochim Biophys Acta 2009; 1792: 688-702
  • 75 Luo Y, Fan Y, Chen X, Yu B, Yue L, Wang D. et al. Generation of induced pluripotent stem cells from Asian patients with chronic neurodegenerative diseases. J Reprod Dev 2012; 58: 515-21
  • 76 Chen LW, Kuang F, Wei LC, Ding YX, Yung KK, Chan YS. Potential application of induced pluripotent stem cells in cell replacement therapy for Parkinson's disease. CNS Neurol Disord Drug Targets 2011; 10: 449-58
  • 77 Feng Z, Gao F. Stem cell challenges in the treatment of neurodegenerative disease. CNS Neurosci Ther 2012; 18: 142-8
  • 78 Balami JS, Fricker RA, Chen R. Stem cell therapy for ischaemic stroke: Translation from preclinical studies to clinical treatment. CNS Neurol Disord Drug Targets 2013; 12: 209-19
  • 79 Mochizuki H, Choong CJ, Yasuda T. The promises of stem cells: Stem cell therapy for movement disorders. Parkinsonism Relat Disord 2014; 20 Suppl (Suppl. 01) S128-31
  • 80 Meyer M, Jensen P, Rasmussen JZ. Stem cell therapy for neurodegenerative disorders. Ugeskr Laeger 2010; 172: 2604-7
  • 81 Trueman RC, Klein A, Lindgren HS, Lelos MJ, Dunnett SB. Repair of the CNS using endogenous and transplanted neural stem cells. Curr Top Behav Neurosci 2013; 15: 357-98