16
Stem Cells in Neurological Disorders: Emerging Therapy with Stunning Hopes Ghanshyam Upadhyay & Sharmila Shankar & Rakesh K. Srivastava Received: 28 June 2014 /Accepted: 27 August 2014 # Springer Science+Business Media New York (outside the USA) 2014 Abstract Neurodegenerative diseases are still a challenge for researchers and clinicians due to its complexity. Traditional medicines usually do not provide sufficient protection against these diseases due to drug resistance and relapse. The discov- ery of the therapeutic potential of stem cells offers new op- portunities for the treatment of incurable neurological dis- eases. Based on their biological properties, stem cells can differentiate into specific tissue type and maintain the cellular tissue/organ homeostasis in physiological and pathological conditions. Recently, it has been demonstrated that somatic cells of patients can be reprogrammed to a pluripotent state from which neural lineage cells can be derived. Potential strategies such as cell replacement therapy and gene transfer to the diseased or injured brain have opened a new line of therapeutic approach for a broad spectrum of human neuro- logical diseases. Thus, stem cell replacement therapy for central and peripheral nervous system disorders aims at repopulating the affected neural tissue with new neurons. However, the limiting factors that have hampered the development of this promising therapeutic approach are the lack of suitable cell types for cell replacement therapy in patients suffering from neurological disorders. In this review, we have discussed the recent advances in stem cell replacement therapy with particular emphasis to neurological disorders. Keywords Neuron . Stem cells . Neurodegenerative diseases Abbreviations DA Dopaminergic ESCs Embryonic stem cells iPSCs Induced pluripotent stem cells iPS cells Induced pluripotent stem cells ADSCs Adipose-derived stromal cells BM-MSCs Bone marrow-derived mesenchymal stem cells NSCs Neural stem cells NGF Nerve growth factor BDNF Brain-derived growth factor Oct4 Octamer-binding transcription factor 4 Mbd3 Methyl-CpG-binding domain protein 3 NuRD Nucleosome remodeling and deacetylase Ascl1 Achaete-scute complex homolog 1 Brn2 Brain-2 Myt1l Myelin transcription factor 1-like NeuroD Neuronal differentiation OLIG2 Oligodendrocyte lineage transcription factor 2 Zic1 Zinc finger protein of the cerebellum 1 miR-9/9* Bifunctional microRNA strands 9 miR-124 MicroRNA 124 Lmx1a LIM homeobox transcription factor 1 alpha Nurr1 Nuclear receptor related 1 Pitx3 Paired-like homeodomain 3 Foxa2 Forkhead box A2 EN1 Engrailed homeobox 1 Lhx3 LIM homeobox 3 Hb9 Homeobox 9 G. Upadhyay Department of Biology, City College of New York, Marshak Building, 160 Convent Avenue, New York, NY 10031, USA e-mail: [email protected] S. Shankar (*) : R. K. Srivastava (*) Kansas City VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO 66128, USA e-mail: [email protected] e-mail: [email protected] S. Shankar Department of Pathology, University of Missouri-Kansas City, Kansas City, MO 64108, USA Mol Neurobiol DOI 10.1007/s12035-014-8883-6

Mol Neurobiol

Embed Size (px)

Citation preview

Page 1: Mol Neurobiol

Stem Cells in Neurological Disorders: Emerging Therapywith Stunning Hopes

Ghanshyam Upadhyay & Sharmila Shankar &

Rakesh K. Srivastava

Received: 28 June 2014 /Accepted: 27 August 2014# Springer Science+Business Media New York (outside the USA) 2014

Abstract Neurodegenerative diseases are still a challenge forresearchers and clinicians due to its complexity. Traditionalmedicines usually do not provide sufficient protection againstthese diseases due to drug resistance and relapse. The discov-ery of the therapeutic potential of stem cells offers new op-portunities for the treatment of incurable neurological dis-eases. Based on their biological properties, stem cells candifferentiate into specific tissue type and maintain the cellulartissue/organ homeostasis in physiological and pathologicalconditions. Recently, it has been demonstrated that somaticcells of patients can be reprogrammed to a pluripotent statefrom which neural lineage cells can be derived. Potentialstrategies such as cell replacement therapy and gene transferto the diseased or injured brain have opened a new line oftherapeutic approach for a broad spectrum of human neuro-logical diseases. Thus, stem cell replacement therapy forcentral and peripheral nervous system disorders aims atrepopulating the affected neural tissue with new neurons.However, the limiting factors that have hampered thedevelopment of this promising therapeutic approach arethe lack of suitable cell types for cell replacementtherapy in patients suffering from neurological disorders.

In this review, we have discussed the recent advances instem cell replacement therapy with particular emphasisto neurological disorders.

Keywords Neuron . Stem cells . Neurodegenerative diseases

AbbreviationsDA DopaminergicESCs Embryonic stem cellsiPSCs Induced pluripotent stem cellsiPS cells Induced pluripotent stem cellsADSCs Adipose-derived stromal cellsBM-MSCs Bonemarrow-derived mesenchymal stem cellsNSCs Neural stem cellsNGF Nerve growth factorBDNF Brain-derived growth factorOct4 Octamer-binding transcription factor 4Mbd3 Methyl-CpG-binding domain protein 3NuRD Nucleosome remodeling and deacetylaseAscl1 Achaete-scute complex homolog 1Brn2 Brain-2Myt1l Myelin transcription factor 1-likeNeuroD Neuronal differentiationOLIG2 Oligodendrocyte lineage transcription factor 2Zic1 Zinc finger protein of the cerebellum 1miR-9/9* Bifunctional microRNA strands 9miR-124 MicroRNA 124Lmx1a LIM homeobox transcription factor 1 alphaNurr1 Nuclear receptor related 1Pitx3 Paired-like homeodomain 3Foxa2 Forkhead box A2EN1 Engrailed homeobox 1Lhx3 LIM homeobox 3Hb9 Homeobox 9

G. UpadhyayDepartment of Biology, City College of New York, MarshakBuilding, 160 Convent Avenue, New York, NY 10031, USAe-mail: [email protected]

S. Shankar (*) : R. K. Srivastava (*)Kansas City VA Medical Center, 4801 Linwood Boulevard, KansasCity, MO 66128, USAe-mail: [email protected]: [email protected]

S. ShankarDepartment of Pathology, University of Missouri-Kansas City,Kansas City, MO 64108, USA

Mol NeurobiolDOI 10.1007/s12035-014-8883-6

Page 2: Mol Neurobiol

Isl1 Islet 1 (ISL LIM homeobox 1)Ngn2 Neurogenin 2Sox2 Sex determining region Y box 2Klf4 Krüppel-like factor 4c-Myc v-Myc avian myelocytomatosis viral

oncogene homologE47/Tcf3 Transcription factor 3Aβ Amyloid-betaAPP Amyloid precursor proteinASC Adult Stem CellsBM-MSC Bone marrow mesenchymal stem cellChAT Choline-acetyltransferaseEPI-NCSC Epidermal neural crest stem cellES Embryonic stem cellNGFR Nerve growth factor receptorhNSC Human neural stem cellmNSC Murine neural stem cellUCB-MSC Umbilical cord blood mesenchymal stem cellLRRK2 Leucine-rich repeat kinase 2PINK1 PTEN-induced putative kinase 1Fbxo7 F-Box only protein 7PSEN1 Presenilin protein 1PSEN2 Presenilin protein 2TREM2 Triggering receptor expressed on myeloid

cells 2APP Amyloid precursor proteinSOD1 Superoxide dismutase 1VAPB Vesicle-associated membrane protein-

associated protein B/CTDP43 TAR DNA-binding protein 43C9ORF72 Chromosome 9 open reading frame 72FUS Fused in sarcomaABCG2 ATP-binding cassette sub-family G member 2CXCR4 C-X-C chemokine receptor type 4FGF R4 Fibroblast growth factor receptor 4Frizzled-9 Frizzled class receptor 9Glut1 Glucose transporter 1SSEA-1 Stage-specific embryonic antigen 1BLBP/FABP7

Brain lipid binding protein (also fatty acidbinding protein 7)

GLAST/SLC1A3

Glutamate aspartate transporter or solutecarrier family 1 (glial high-affinity glutamatetransporter) member 3

GFAP Glial fibrillary acidic proteinS100B S100 calcium binding protein BPAX6 Paired box protein 6TBR2 T-box brain protein 2 or eomesoderminFGF Fibroblast growth factorIslet-1 and 2 ISL LIM homeobox 1 and 2Lhx3 LIM/homeobox protein 3Olig2 Oligodendrocyte transcription factorMOG Myelin oligodendrocyte glycoproteinGalC Galactosylceramidase

NeuN Feminizing locus on X-3 Fox-3 Rbfox3, orhexaribonucleotide binding protein-3

NF-L Light neurofilamentNF-M Medium neurofilamentTH Tyrosine hydroxylaseGAD Glutamic acid decarboxylasePSD-95 Postsynaptic density protein 95VAMP Vesicle-associated membrane proteinsEMT Mesenchymal to endothelial transition

Introduction

Neurological diseases are characterized by any disorder in thecentral nervous system (CNS) or the peripheral nervous sys-tem (PNS) due to structural, biochemical, and electrophysio-logical dysfunctions of neurons or glial cells. Neurodegener-ative diseases are common types of neurological disordersresulting from progressive degeneration or functional loss ofneurons [1–3]. Parkinson’s disease (PD), Alzheimer’s disease(AD), Huntington’s disease (HD), and amyotrophic lateralsclerosis (ALS) are included in the category of neurodegener-ative diseases (Fig. 1). Regardless of the type of neurologicaldisease, all symptoms result from the significant loss of neu-rons or glial cells in the nervous system. For example, PD iscaused by the death of dopaminergic (DA) neurons in thesubstantia nigra pars compacta of the midbrain; HD resultsfrom the death of medium spiny neurons in the basal ganglia,and ALS is caused by the death of motor neurons [1–3].

Although being a most prominent elderly problem, nopromising therapy is available for the treatment of neurode-generative diseases. Various scientific attempts have beenmade in past to address this alarming issue with limited orno success. Advancement of knowledge about stem cells hasdrawn attention towards stem cell therapy as a promisingtherapeutic option for the treatment of various neurologicdisorders. Stem cells hold great promise for regenerativetherapies by virtue of their ability to regenerate tissues andcontribute to their homeostasis [4]. The stem cells have bio-logical properties of self-renewal and have capability to giverise to differentiated cell progenies that maintain tissue ho-meostasis in physiological and pathological conditions. It isnow well known that stem cells can be specifically and effi-ciently directed toward distinct cell lineages in vitro apart frompluripotency. These properties of stem cells could be translat-ed in clinics for the treatment of disease that have limitedtherapeutic hopes [5–7]. The baseline and clinical studies,harboring these properties of stem cells, are underway, andthe hopes are stunning. Advancement in the approach of stemcell therapy is not limited to neurological disorders but in-cludes a wide spectrum of diseases such as cardiovascular,gastrointestinal, pulmonary diseases, etc.

Mol Neurobiol

Page 3: Mol Neurobiol

Currently, a number of stem cells such as embryonic stemcells (ESCs), induced pluripotent stem cells (iPSCs), adipose-derived stromal cells, bone marrow-derived mesenchymalstem cells (BM-MSCs), and neural stem cells (NSCs) arebeing investigated for their therapeutic potential using animalmodels. Studies have shown that neurons, oligodendrocytes,and astrocytes could be generated from ESCs and adult stemcells (ASCs) [8–11] (Fig. 2). Moreover, it has been also foundthat the NSCs in the sub-ventricular zone of the lateral ventri-cles and the sub-granular zone of the dentate gyrus in thehippocampus of the mammalian brain maintain the capabilityto generate new neural cells throughout the lifetime [9, 12, 13].

Although various animal studies to date have been con-ducted indicating the therapeutic potential of stem cells incombating against neurological diseases, a lot needs to bedone before clinical application on human subjects. In thisreview, we have discussed the recent advances in stem cellreplacement therapy, challenges, and future prospects withparticular emphasis to neurological disorders.

Strategies in stem cell replacement therapyfor neurological diseases

The process of stem cell replacement therapy is very stringent,particularly in case of neurological disorders. The success de-pends on precautions and fine-tuning of the development oftightly controlled isolation, expansion, and sorting processes.Maintaining the self-renewal and multi-potential properties ofstem cells is critical along with safety and efficacy of transplan-tation procedures [14, 15]. The basic strategies in stem cellreplacement therapy for neurological diseases include stem cellstransplantation within the brain or infusion by blood circulation;stem and progenitor cells mobilization by cytokines, or trophicand growth factors, into the brain in vivo; delivery of stem cells

through bone marrow transplantation; engineering of stem cellsto correct the genetic defect; reprogramming of neuronal cells;and coupling of stem cells with biomaterials (Fig. 3).

A study by Osanai et al. reported that the carotid arteryinjection of stem cells could, together with in vivo opticalimaging to track the injected cells after transplantation, poten-tially be part of a new approach for stem cell transplantation inhuman brain trauma injuries [16]. In this study, the stem cellsobtained from the rats’ bone marrow were labeled with quan-tum dots (a biocompatible, fluorescent semiconductor), con-stituted a non-invasive method of monitoring the stem cellsfor a period of 4 weeks following transplantation [16]. Theywere able to show that the injected cells entered the brainwithout entering the general circulation. They also observedthat the stem cells started migrating from the capillaries intothe injured part of the brain within 3 h.

For the proper migration and spreading of transplanted stemcells, it is important to select the best injection site, the correcttherapeutic cell number, and the monitoring of proper engraft-ment and differentiation of stem cells within the brain [8]. Somepositives came out while transplanting ESCs or ASCs into thebrain of animal models of AD, PD, HD, lysosomal storagedisorders (LSDs), demyelinating disorders, and traumatic lesionsof the brain and spinal cord [5, 17, 18]. Combination of stem celltransplantation with nerve growth factor (NGF) and brain-derived growth factor (BDNF), therapeutic molecules, or genetransfer technology was also investigated [10, 19, 20] withlimited success. Another alternative approach was the transplan-tation of more lineage restricted progenitor cells. In this context,novel technologies were used to facilitate the generation of largenumbers of committed cell populations for testing in cell replace-ment and regenerative paradigms [5, 21].

Concurrently, serious efforts of tissue engineering weremade that include the transplantation of stem cells in combi-nation with natural or synthetic biomaterials citing the studies

Fig. 1 Genetic mutationsassociated withneurodegenerative diseases:Mutations in the LRRK2, parkin,PINK1, SNCA, Fbxo7 result inPD; mutations in PSEN1, PSEN2,APP, TREM2, etc., can lead toAD; expansion CAG repeat inHuntingtin gene (HTT) is wellknown cause of HD; mutations inSOD1, VAPB, TDP43,C9ORF72, FUS led to ALS

Mol Neurobiol

Page 4: Mol Neurobiol

that have shown the enhancement of donor cell survival aftertransplantation of intact tissue [22, 23]. This success could beattributed to the presence of a three-dimensional architectureand a higher accessibility of extracellular adhesive proteinswith which donor cells can interact [23]. Remarkably, thetissue-engineering approaches have some success, and thetransplantation of encapsulated growth factor-secreting cellsand NSCs within porous poly-glycolic acid scaffolds haveshown positive results against hypoxia/ischemia [24]. Theseefforts also got momentum from the reports stating that chem-ical and biological modifications of biomaterials could direct-ly influence stem cell behavior such as alteration of substrateproperties, nano-pattern design, and scaffold degradation rate[23, 25]. Additionally, these stem cells were also used incombination with a three-dimensional biomaterial (scaffolds)for the treatment of traumatic brain injury or the regenerationof injured spinal cord [26].

However, limitation of transplantation therapy, such as hostimmune response to the cells or specific antigens and otherassociated risks, in neurogenesis and differentiation in vivo,

and ethical issues, particularly of scientific work on abortedfetuses, drove the focus on the development of more ethicalmethods (such as using ASCs) that could enable us to researchethically [15]. Reprogramming of neuronal cells is of majorfocus these days. Adult somatic cells, such as skin fibroblasts,are targeted to generate new class of pluripotent stem cells(induced pluripotent cells (iPS)) by introduction ofembryogenesis-related genes like Oct4, Sox2, Klf4, and c-Myc [11, 27–32]. The process requires initial cell proliferationfollowed by a conversion of a fraction of the cell progeny intoan embryonic stem (ES)-like state with different time latencies[27]. Since then, a number of successful attempts ofreprogramming with different compositions of these geneshave been tried (Table 1). Epigenetic regulation by a varietyof chromatin modifiers also play critical role inreprogramming. Rais et al. recently showed that depletion ofMbd3, a core member of the Mbd3/NuRD repressor complexfor nucleosome remodelling and deacetylation, together withOct4, Sox2, Klf4, and c-Myc co-transduction andreprogramming in naive pluripotency promoting conditions,

Astrocytes: CD44

FGF R3, GFAP,

Glast, S100B

Early progenitor

cells

Differentiated

cells

Neural progenitors can broadly

classified in radial glial cells

(BLBP/FABP7,

GLAST/SLC1A3, GFAP,

Nestin, S100B, Vimentin) and

basal progenitors (PAX6–

SOX2– TBR2)

Differentiated Post-Mitotic

Neuronal Cells: NeuN

Neurofilaments (e.g. NF-L,

NF-M), GAD, TH, PSD-95,

Synaptophysin, VAMP

Oligodendrocytes:

GalC, MOG, Myelination

Antigen O1, O4 and Sox10

Neural stem

cells

Neural stem cells:

ABCG2, CD133, CXCR4,

FGF R4, Frizzled-9, Glut1,

SSEA-1, Notch-1 & -2

Glial-restricted

progenitors: A2B5, FGF

Receptors, Nestin

Motor neuron progenitors:

Islet-1 & -2, Lhx3,

Neurogenin-2, Olig2

Neuron-restricted

progenitors:

Doublecortin, NCAM,

MAP2, β-III Tubulin

Motor Neurons: Islet 1 & -2,

Lhx3, Neurogenin 2, Olig2

Late progenitor

cells

Fig. 2 Differentiation of stem cells in neuronal lineages: Neural stemcells have capability of self renew itself and give rise early neuralprogenitors. These early progenitors further differentiate into committedprogenitors like glial-restricted progenitors, motor neuron progenitors,

and neural-restricted progenitors that ultimately generate mature neuralpopulation. Each neural population can be identified and purified on thebasis of certain markers/proteins they express during the course ofdifferentiation

Mol Neurobiol

Page 5: Mol Neurobiol

result in deterministic and synchronized iPS cellreprogramming [54]. These findings have mobilized the ap-proach as iPS cells could offer the opportunity to generatepatient-specific stem cells from somatic differentiated cells ofpatients affected by several neurological disorders as com-pared with the traditional ESCs [11, 55, 56].

Applications of Stem Cells in Neurological Diseases

The stem cell research, in particular, translational stem cell-based research and stem cell-based clinical applications, hasincreased during recent years [8, 17, 57]. Stem celltransplanting approaches for neurological diseases compriseof ESCs and several types of ASCs. These include bonemarrow-derived cells (both hematopoietic stem cells and bonemarrow-derived stem cells), NSCs, human umbilical cordblood stem cells, and mesenchymal stem cells [8, 17]. More-over, patient-specific iPS cells also have influence on thedevelopment of new therapies for neurological disorders [6,27].

Generally, clinical trials based on stem cell replacementtherapy are confined to patients with PD, HD, ALS, and someLSDs [5]. Although stem cell research for these diseases to

date has limited success and variable results, the positive is theability of stem cells to integrate into host brain tissues in mostof the cases with some positive effects for the recipients. In afew cases, stem cells were able to restore the degeneratedtissue [58–63].

Neurological diseases include a variety of disorders of thebrain, the spinal cord, and the PNS and have some commonfeatures such as neural dysfunctions, progressive deteriora-tion, and extensive loss of neural cells. The pathogenic mech-anism is diverging and complex. Therefore, the effective stemcell replacement therapy demands that these cells should betailored to the specific dysfunctions.

Parkinson’s Disease

PD is a progressive neurological disorder characterized byshaking (tremors) and difficulty with walking, movement,and coordination. It is one of the most common nervoussystem disorders of the elderly and is characterized by the lossof DA neurons and glial cells [1, 64]. To date, no specifictreatment strategy has been developed for the treatment of PD;however, recent focus is stem cell therapy (Table 2). Strategiesin PD stem cell therapy basically involve the replacement of

Gene Therapy

Stem cells

Implantation

Stem cells

Implantation by

blood infusion

Stem cells and

biomaterials

(Neural-

engineering)

Foetal brain

Pre-implantation embryo

Embryonic

stem cells

Fibroblast cells

Mesenchymal

stem cells

iPS cells

Neural progenitor or

neuronal cells

Reprogramming

EMT Sox2

c-Myc

Oct4

Klf4

Stem cells

Implantation by

BMT

Fig. 3 Strategies in stem cell replacement therapy for neurologicaldiseases: The major problem with stem cell therapy is to obtain startingcell population. Embryonic stem cells that have capability to differentiatein neural progenitors can be obtained from pre-implantation embryoswhich is a major ethical consideration. Ethical consideration is associated

with neural stem cells and committed neural progenitors obtained fromfetal brain also. Other methods to obtain starting cell population could betrans-differentiation of mesenchymal cells to neural progenitors orreprogramming of adult cells (like fibroblast) to iPSCs, which can bedifferentiated into neural progenitors for the therapy

Mol Neurobiol

Page 6: Mol Neurobiol

degenerated neurons with other dopamine-producing cells[77]. The first clinical stem cell therapy of PD took place in1987 using aborted human fetal ventral midbrain tissue, andsince then, more than 400 PD patients have undergone stemcell therapy [78–80]. Initial attempts in this direction were

consisted of the transplantation of cells isolated from pieces offetal midbrain tissue containing neuronal progenitor celltypes, as donor cells [81]. In PD, where the DA neurons areprogressively lost over time, the goals of stem cell therapy wasthe replacement of those neurons and for the restoration of

Table 1 Summary of primarycells, infection method, and com-position of factors for directreprogramming to neuronal cells

Primary cells Infectionmethod

Factors References

Fibroblasts Lentiviral Ascl1, Brn2, Myt1l [33, 34]

Fibroblasts Lentiviral Ascl1, Brn2, Myt1l, NeuroD [35]

Fibroblasts Lentiviral Ascl1, Myt1l, NeuroD2, miR-9/9*,and miR-124

[36]

Fibroblasts Lentiviral Ooc4, Sox2, Nanog, and Lin28 [37]

Fibroblasts Lentiviral Oct4, Sox2, Klf4, and Myc [38]

Fibroblasts Lentiviral Ascl1, Brn2, Myt1, Lmx1a, Fox2 [39]

Fibroblasts Retroviral Ascl1, Brn2, Myt1l, NeuroD1,Lhx3, Hb9, Isl1, Ngn2

[40]

Fibroblasts: Coriell, GM11272,GM16548, GM17880, GM11270

Retroviral Sox2, Oct4, c-Myc, and Klf4 [41]

Fibroblasts: Patient biopsy andCoriell, GM17880, GM11270

Retroviral Oct4, Sox2, Klf4, and Myc [42]

Fibroblasts Lentiviral Sox2, Brn2, FoxG1 [43]

Fibroblasts Retroviral Sox2, Brn4/Pou3f4, Klf4, c-Myc, E47/Tcf3 [44]

Fibroblasts Retroviral Oct3/4, Sox2, Klf4, and c-Myc [45]

Fibroblasts Retroviral Oct4, Sox2, Klf4, and c-Myc [46]

Fibroblasts Lentiviral Brn2, Myt1l, miR-124 [47]

Fibroblasts Lentiviral Ascl1, Lmx1a, Nurr1 [48]

Fibroblasts Retroviral Sox2 [49]

hiPSCs derived from fibroblasts Lentiviral Rex1, Nanog, Crupto, Oct4, and Sox2 [50]

Fibroblasts Retroviral c-Myc, Oct3/4, Klf4, and Sox2 [51]

Fibroblasts: Coriell, GM02038,GM01792, GM01835, GM02497

Lentiviral Oct4, Sox2, Klf4, Myc, and Lin28 [52]

Fibroblasts Lentiviral Ascl1, Lmx1a, Nurr1, Pitx3, FoxA2, EN1 [53]

Table 2 Summary of stem cell research conducted in animal models of PD

Animal model Cell source Outcome Reference

6-OHDA unilateral lesion rat iPS clone O9 Ipsilateral rotation (amphetamine)—decrease [65]

iPS cells IMR90 clone 4 Decline in rotation score [66]

iPS cells excisable virus Ipsilateralrotation (amphetamine) —decrease [67]

Mesencephalic DA neurons Ipsilateralrotation (amphetamine) —complete reversal [68]

mES cells-derived dopamine neurons Ipsilateralrotation (amphetamine) —complete reversal [69]

hES cells-derived dopamine neurons Ipsilateralrotation (amphetamine) —complete reversal [70]

hES cells-derived dopamine neurons Ipsilateralrotation (amphetamine) and contralateralrotation (apomorphine)—decrease

[71]

hES cells Ipsilateralrotation (amphetamine) and contralateralrotation (apomorphine)—decrease

[72]

6-OHDA bilateral lesion rat mES cells-derived dopamine neurons Ipsilateralrotation (amphetamine) —complete reversal [73]

6-OHDA unilateral lesion mouse mES cells-derived dopamine neurons Ipsilateralrotation (amphetamine) —decrease [74]

ntES cell-derived dopamine neurons Ipsilateralrotation (amphetamine) —decrease [75]

ntES cell-derived from parkinsonian mice Ipsilateralrotation (amphetamine) —complete reversal [76]

Mol Neurobiol

Page 7: Mol Neurobiol

normal neuronal sprouting, connectivity, and functionality[82]. Many groups harbor the molecular mechanisms bywhich ESCs generate various kinds of neurons includingDA neurons. Friling et al. showed the generation of DAneurons from mouse and human ESCs due to over-expression of Lmx1a, a transcription factor acting as a deter-minant of mesencephalic dopamine neurons during embryo-genesis [83].

Several studies reported symptomatic improvement in PDpatients after transplantation of these cells [84, 85]; however,several studies showed negative results also [86, 87]. Failureof these studies could be attributed to the difficulties in surgi-cal transplantation of fetal mesencephalic cells, or from non-specific cells within the fetal tissue that could have interferedwith cell growth and differentiation [88–90]. During the lastdecade, the advancement of the knowledge aboutreprogramming of adult cells to iPS cells has given a lot ofhope. Byers et al. established iPS cells with capacity to dif-ferentiate into functional neurons from a patient with a dom-inant autosomal form of PD [91]. Later on,Wernig et al., usinga mouse model, showed that iPS cells can be efficientlydifferentiated into neural precursor cells with the potential togive rise to glial and neuronal cells including glutamatergic,GABAergic, and catecholaminergic subtypes upon transplan-tation into the fetal mouse brain. They further illustrated thatthe iPS cells induced to differentiate into dopamine neuronswere able to improve behavior in a rat model of PD upontransplantation into the adult brain [65]. Recently, Sagal et al.showed that ectopic expression of Atoh1, in combination withcell extrinsic factors, drives the hPSCs into functional DAneurons with >80 % purity. These neurons recapitulate keybiochemical and electrophysiological features of midbrain DAneurons and provide a reliable disease model for studying PDpathogenesis [92].

Alzheimer’s Disease

AD is an irreversible, progressive neurological disorder thatslowly destroys memory and thinking skills and eventuallyeven the ability to carry out the simplest tasks [93]. It is a late-onset disease, and symptoms appear mostly after the age of60 years. AD is the most common cause of dementia, charac-terized by the loss of cognitive functioning-thinking, remem-bering, and reasoning, to such an extent that it interferes with aperson’s daily life and activities [93, 94]. An early symptom ismemory lapses (amnesia). As disease progresses, memoryproblems worsen, increasing confusion and disorientation,obsessive, repetitive, or impulsive behavior, recognition prob-lem (agnosia), and delusions. In the later stages ofAlzheimer’s disease, the symptoms become increasingly se-vere with problems with speech or language (aphasia), dis-turbed sleep, changes in mood, difficulty performing spatial

tasks (apraxia), problems with eyesight, etc. Estimates vary,but experts suggest that as many as 5.2 million Americansmay have AD [95]. The cause of AD is not entirely known,but it is thought to include both genetic and environmentalfactors. A diagnosis of AD is made when certain symptomsare present, and by making sure other causes of dementia arenot present.

Stem cell-based therapy for AD is confined to theneurogenesis or repopulation of the degenerated brain. Atpresent, stem cell treatment is limited to preclinical studieson AD animal models, and preclinical findings have to beextrapolated in clinics. However, preclinical studies havesome points, and hopes are rising for the development ofnew therapeutic approach (Table 3). ESCs, as well as ofseveral ASC types, has already been shown to be effective;the recent approach combining stem cells with molecules witha neurological function is more promising. In fact, the intro-duction of additional factors is a necessary support for thetherapy of the disease [109–111]. Recently, Sugaya et al.generated neural cells in the mouse brain by using both neuraland mesenchymal stem cell transplantation and a small mo-lecular compound (phenserine, a cholinesterase inhibitordrug), leading to an improvement of cognitive function inanimal models [110]. These results are interesting, and animprovement in the mouse’s cognitive functions and otherAD pathogenesis hallmarks was observed.

Evidence from in vitro mouse ESC culture studies hasindicated that neural progenitor cells (NPCs) can be generatedfrom ESCs, expanded, and differentiated efficiently into neu-rons and glial cells by serum-free culture [112, 113]. Wanget al. showed that ESC-derived neurospheres transplanted intothe frontal cortex of animals in an nbM lesion mouse modelsurvive and produce many ChAT-positive neurons and a fewserotonin-positive neurons in and around the engrafted cellsand improve working memory [106]. Moghadam et al.transplanted primed and unprimed mESC-derived NPCs intothe unilateral nbM in a rat model of AD. The results showedsignificant learning and memory improvements and also themajority of the engrafted NPCs retain a neuronal phenotypeand approximately 40 % of these cells show a cholinergic cellphenotype [114]. Tang et al. also showed therapeutic effects ofNPCs derived from ESCs following transplantation into a ratAD model with dorsal hippocampal injury induced by Aβ.The transplantation resulted in amelioration of memory im-pairment after 16 weeks [115]. Furthermore, some studieshave also shown hESCs having the potential to treat manyneurodegenerative diseases and brain injuries including AD[116–118].

In contrast to ESC-based therapy, which is complicated byimmune rejection because of immunological incompatibilitybetween patients and donor cells, iPSCs can be generatedfrom the patient’s own somatic cells, thus avoiding the ethi-cally controversial use of human embryos and immunogenic

Mol Neurobiol

Page 8: Mol Neurobiol

rejection [119]. Patient-derived iPSCs retain the patient’s ge-notype, therefore enabling the applicable recapitulation of ADphenotype in a dish. The high-content screening assays andthe barriers that exist to evaluate the full potential in predictiveefficacy, toxicology, and disease modeling in these cells couldenable rapid analysis of thousands of drugs for AD simulta-neously [120]. Yagi et al. generated iPSCs from fibroblasts ofearly onset familial AD (FAD) patients with mutations in PS1(A246E) and PS2 (N141I), and showed that secretion ofAβ42 from FAD-iPSC-derived neurons is increased signifi-cantly and markedly reduced by γ-secretase inhibitors andmodulators [121, 122]. Using this human iPSC-based modelof AD, Yahata et al. screened for the effectiveness of anti-Aβdrugs, such as β-secretase inhibitors, γ-secretase inhibitors(GSI), and a nonsteroidal anti-inflammatory drug (NSAID),between early and late differentiation stages. As a result, theyfound that GSI treatment results in a drastic decline of Aβproduction [123].

Huntington’s Disease

HD is a neurodegenerative genetic disorder that affects musclecoordination and leads to cognitive decline and dementia. It is

middle-age-onset disease and is the most common geneticcause of abnormal involuntary writhing movements calledchorea [124, 125]. Physical symptoms of HD may appear onearly age, but frequency is more in between 35 and 44 years ofage [126]. In some cases (∼6 %), the symptoms may appearbefore the age of 21 years with an akinetic-rigid syndrome.The prevalence of this disease ismore in the people ofWesternEuropean origin than in those from Asia or Africa. It is aninheritable disease caused by an autosomal dominantmutationon either of an individual’s two copies of a gene calledHuntingtin with a 50 % risk of inheriting the disease to thechild by the affected person [124, 125].

HD is an ideal model for the development of stem cell-based therapeutic strategies due to its unique pathogenesischaracteristics. The approach is basically centered on theprevention of neuronal dysfunction/cell death and replace-ment of lost neurons in the striatum. In fact, a phase-I clinicaltrial consisted of implantation of fetal striatal primordium intothe striatum of five HD patients [59]. Such kind of efforts islimited due to ethical, social, and logistical issues associatedwith the use of human fetal tissues for brain transplantation.Adult/autologous stem cells, therefore, could be an alternativesource, and wide preclinical research transplanting differenttypes of stem cells into HD animal models is needed in this

Table 3 Summary of stem cell research conducted in animal models of AD

Animal model Stem cells Outcomes Ref

Aged rats (30 months) Murine BM-MSC Aged rats—learn more rapidly [96]

Ibotenic acid-inducedNBM lesion rats

Murine BM-MSC Ibo-induced memory impairment group—significant reduction in latency tofind platform in Morris water maze

[96]

Acute Aβ-inducedmodel mice

Murine BM-MSC BM-MSCs promoted microglial activation; reduced Aβ deposits of acutelyinduced AD mice

[97]

AF64A cholinotoxininjection in rats

hNSC Rats receiving NSCs overexpressing ChAT showed full recovery in learning and memoryfunctions, whereas those receiving NSCs only remained memory impaired

[98]

Aged rats (22 months) hNSC SGZ increased in cell number [99]

Matured rats (6 months) hNSC Cognitive function significantly improved in matured and aged memory-impaired groups [100]

Aged rats (24 months) hNSC Morphologically functional hNSC-derived cells were found in the hippocampus and cortex [100]

DS model mice (Ts65Dn) mNSC Decreased tau-positive clusters in trisomic (28.6 %) and disomic (58.6 %) mice [101]

Triple transgenic ADmodel mice

mNSC NSC transplant rescues learning and memory deficits; no change in Aβ, tau pathologybut increased synaptic density in mice’s hippocampus

[102]

Double transgenic neuronalinjury model mice

mNSC Improved hippocampal-dependent memory and increased synaptic density and neuronalnumber

[103]

Rat fimbria–fornix transaction mNSC Improved memory and learning in Y-maze testing; increased in the number ofp75NGFR-positive neurons

[104]

Transgenic AD-model mice Human adipose-derived stem cell

Both intravenous and intracerebral adipose-derived stem cell transplantation rescued memoryimpairment and improved spatial learning; reduced amyloid plaque formation, upregulatedinterleukin-10 and neurotrophic factors in the brain of Tg2576 mice

[105]

Ibotenic acid-induced NBMlesion mice

Murine ES andES-derived NSC

NPC restored memory, ES significantly decrease working memory; ES induced massiveteratoma formation

[106]

Rat hippocampal Aβ injection EPI-NCSC Significant improvement in cognitive tasks (Y-maze and passive avoidance tests), increasedneuron number and differentiation into other cell type

[107]

APP and presenilin (PS1)double-transgenic mice

Human UCB-MSC Improved spatial learning and memory in Morris water maze tests; reduced Aβ load andtauhyperphosphorylation, inhibited proinflammatory cytokine release from microglia

[108]

Mol Neurobiol

Page 9: Mol Neurobiol

context [7, 127]. Using quinolinic acid (QA) lesion rat modelof HD, Vazey et al. showed that adult neural progenitor cellssurvive transplantation, undergo neuronal differentiation witha proportion of newly generated cells expressing markerscharacteristic of striatal neurons, and reduce functional im-pairment in the QA lesion model of HD [128]. These resultsgot further support by studies of Amin et al. who used bonemarrow mesenchymal stem cells [129]. Some other studiesexplored the benefits of combining stem cells with therapeuticdrugs [7, 127]. Juopperi et al. showed that iPSCs derived froma father with adult-onset HD and 50 CAG repeats (F-HD-iPSC) and his daughter with juvenile HD and 109 CAGrepeats (D-HD-iPSC) were capable of producing phenotypi-cally normal, functional neurons in vitro and were able tosurvive and differentiate into neurons in the adult mouse brainin vivo after transplantation. They further stated that theastrocytes derived from patient-specific iPSCs exhibit a vac-uolation phenotype, a phenomenon previously documented inprimary lymphocytes from HD patients [130]. Recently, Finket al., using 3-nitropropionic acid rat model of HD, showedthat transplantation of adenovirus-generated iPSCs preservedmotor function [131]. Transplanted iPSCs were found tosurvive and differentiate into region-specific neurons in thestriatum of 3-NP rats, at all transplantation time points, andcould provide a potential avenue for therapeutic treatment ofHD [131].

Amyotrophic Lateral Sclerosis

ALS, also known as Lou Gehrig’s disease, is a disease of thenerve cells in the brain and spinal cord that control voluntarymuscle movement [132, 133]. Pathogenesis is complex andpoorly understood, but it is observed that in about 10 % ofcases; ALS is caused by a genetic defect. It is characterized byneuronal loss and the inability to send messages to muscleseventually leading to muscle weakening, twitching, and aninability to move the arms, legs, and the body. Its prevalence ishigh, and ALS affects approximately 5 out of every 100,000people worldwide. The only known risk factor is familyhistory. This is late-onset disease, and symptoms usually donot develop until after age 50 years. Persons with ALS have aloss of muscle strength and coordination that eventually getsworse and makes it impossible to do routine tasks [132–134].Initially, breathing or swallowing muscles are affected; subse-quently, as the disease progresses, more muscle groups devel-op problems. ALS does not affect the senses (sight, smell,taste, hearing, touch). It only rarely affects bladder or bowelfunction, or a person’s ability to think or reason [132–134].

Stem cell treatment is a reality for ALS, and some clinicaltrials have been conducted in the past [135]. Ongoing preclin-ical studies have shown the efficacy of stem cell transplanta-tion in restoring neural loss as well as in treating the adverse

effects of ALS clinical implications. The hopes for ALSpatients are that stem cell transplantation will be able toreplace motor neurons that may further lead to the recoveryof neuromuscular functions. Results from a follow-up studywith 13 patients 1 year after stem cell transplantation showed asignificant improvement in nine patients as confirmed byelectro-neuromyography [60]. Some positives also came outfrom other stem cell clinical procedures, such as allogeneichematopoietic stem cell transplantation, confirming the abilityof stem cells to restore neural dysfunctions [61]. UsinghSOD1G93A mouse model, Sun et al. showed that intrave-nous administration of human amniotic mesenchymal stemcells (hAMSCs), derived from human amniotic membrane(HAM), significantly retarded disease progression, extendedsurvival, improved motor function, prevented motor neuronloss, and decreased neuro-inflammation [136]. Boido et al.showed that hMSCs, when intracisternally administered, canexert their paracrine potential, influencing the inflammatoryresponse of the host [137]. Recently, Chestkov et al. used thereprogramming technology to generate iPSCs with patientswith familial ALS. The iPS cells were obtained by bothintegration and transgene-free delivery methods ofreprogramming transcription factors and had the propertiesof pluripotent cells and were capable of direct differentiationinto motor neurons [138]. Nizzardo et al. observed that aspecific NSC population characterized by high aldehyde de-hydrogenase activity, low side scatter, and integrin VLA4positivity that could migrate and get engrafted into the CNSand could improve neuromuscular function and motor unitpathology in ALS mouse model. They linked these positiveeffects to multiple mechanisms, including production of neu-rotrophic factors and reduction of micro- and macrogliosis[139].

Miscellaneous

Neurological symptoms are the hallmarks of many otherdiseases, such as LSDs [18, 140–143]. Major causes of LSDsare mutations of genes that encode the lysosomal enzymeproteins and related cofactors. LSDs are rare and usuallymanifests in the accumulation of un-degraded products inlysosomes leading to enlargement of cells, cellular dysfunc-tion, and cell death. Most LSDs are autosomal recessive.Manifestations of neurological disease begin in infancy orchildhood with initial delay and subsequent arrest of psycho-motor development, neurological regression, blindness, andseizures. Inexorable progression leads to a vegetative state.

Current therapies for LSDs are ineffective, and results arequite poor. The stem cell replacement approach for this dis-ease generally is focused on the production of a functionalenzyme and the recovery of the damaged brain. The combi-nation of gene/cell therapy strategy could be a better approach

Mol Neurobiol

Page 10: Mol Neurobiol

in comparison to the conventional allogeneic hematopoieticstem cell transplantation [144].

Ongoing studies are exploring the efficacy of stem cell-based treatment in other neurological diseases also [55, 145].The treatment for spinal cord injury now ranges from stem celltransplantation procedures to their combination with bioactivemolecules [145]. Studies with mouse model of multiple scle-rosis have also shown promising results. The basic strategy forthese studies was to increase the number of hematopoietic,mesenchymal, and NSCs in order to provide practical vehiclesfor in situ immunomodulation, neuroprotection, and regener-ation of the damaged tissue. Besides these approaches, inves-tigations are also directed in line of gene/cell therapy strategiesto restore antigen-specific tolerance or to deliver anti-inflammatory molecules [146]. Finally, the efforts made forthe treatment of epilepsy exploring therapeutic potential ofstem cells indicated a restoration of the normal balance be-tween excitation and inhibition in epileptic patients (e.g., NSCand ESC-derived neural progenitors) [20].

A promise and demands towards the generation of newtherapies for neurological disorders

Prospective studies have made a promise that stem cells willbe a powerful part of the therapeutic armamentarium of neu-rologists in upcoming years and decades. Therapeutic ap-proaches with stem cells are appealing and have captured theattention of not only the scientists and clinicians but also ofcommon people. The expectations have some emotional touchas individuals who have suffered greatly owing to injury ordisease thinks that their suffering and disability could bereduced or eliminated by these exciting pleas. Many feel thatstem cells have the potential to treat, cure, and restore functionto patients suffering from almost any disease. The reality willbe much more sobering on several fronts. Some diseases willbe intractable to recovery or even slowing of disease by virtueof the mechanism or extent of neural injury. Assumptionscould only be translated if the demands of stem cell therapywill be fulfilled. Neurogenesis will need to be combined withprolonged rehabilitation therapies to ‘retrain’ the nervoussystem, much as the prolonged rehabilitation after an organor joint replacement or even more. But even with thosecautionary notes, there is the likelihood that we will be ableto alter the course of devastating neurologic diseases usingstem cells in the future, and we will begin to see this potentialin human patients in the next 5–10 years.

Future Perspective

Although our understanding of neural stem cell (NSCs) biol-ogy has increased, there are still enormous hurdles before

universal acceptance of its transplantation for neurologicaldisorders. Certain key issues related to cell selection forstarting point, time of rehabilitation following transplantation,programming of cells in vitro to generate predictable cell typesin vivo, and ethical concerns need to be addressed. ES cell-derived NSCs for stem cell therapy have ethical consider-ations and require neural specification and purification apartfrom its tendency to generate tumors. NSCs, on the otherhand, represent less of a risk for tumor formation followingtransplantation (as they are more lineage-restricted than EScells); however, ethical considerations, long-term mainte-nance, restricted potential, and low availability have limitedtheir uses in stem cell therapy.

In last few years, the capacity of non-NSCs, including bonemarrow, skin, and umbilical cord stem cells, to generatevarious types of neurons and glia has been widely investigated[147–151]. The recent advancement is the patient-derivediPSCs that allow scientists and clinicians to model, in vitro,the progression of neurodegenerative diseases for each indi-vidual patient and provide a real opportunity for stratifiedmedicine applications. Additionally, iPSC technology couldbe coupled with high-throughput screening that can provide amore efficacious and cost-effective platform to assess a num-ber of former and new drug candidates for treatment of neu-rodegenerative disorders simultaneously. Although promis-ing, implementation of iPSC-based therapy is far from becom-ing a reality. The complex nature of various neurologicaldisorders has made the process even more unpredictable.Methods described in mouse and human ESCs translate fairlywell into iPS cells in the specific cases of the spinal motorneurons affected in ALS and midbrain DA neurons in PD, butstill they are far from perfect. It may further be necessary toidentify culture conditions to produce specific subclasses ofthe desired cell type. Where we go after that depends on manythings. First and foremost will be some clinical trials, and ifthese trials will demonstrate hints of the effectiveness of stemcells in treating neurologic disease, we could decide a path.One cannot draw any final conclusion on the basis of theclinical trials performed to date, as they involve small num-bers of patients and often have an open-label design. Howev-er, these trials have some hopes, and the results may beconsidered to be ‘suggestive,’ therefore justifying larger, morecomplex clinical trials.

Conclusion

Stem cell research has revolutionized the approaches andefforts towards the generation of new and effective therapyfor these disorders. The promise of stem cell therapy in theCNS is to regenerate and reconstruct the original pathway topromote functional recovery. Although genetically engineer-ing cells has proven valuable to understand gene function and

Mol Neurobiol

Page 11: Mol Neurobiol

to deliver missing trophic factors or neurotransmitter synthe-sizing enzymes in the CNS, it will need the establishedmethods of translation from the laboratory to the clinic. Todate, it is just a hope and will progress with time. It is must tobe shown in the laboratory that the approach works and how itworks before it can even be considered for clinical use. Inrecent past, neurons and glia have been generated successfullyfrom stem cells in culture such as ESCs, mesenchymal stemcells, and NSCs. A new line of research is neurogenesis andprevention of neuronal/glial cell death using endogenous stemcells within the adult CNS. iPS cell-based models may alsoprovide a new opportunity to understand selective vulnerabil-ity of populations of neurons to discrete degenerative stimuli,a theme common to many neurological disorders.

Now, the major challenge to the researchers and cliniciansis to translate these exciting advances from the laboratory intoclinically useful therapies. Although successful in some cases,stem cell replacement therapy is still in its early days, andbefore envisaging any therapeutic application of such cells inhumans with neurological disorders, we need to confront withseveral key issues related to cell selection, targeting, andtiming.

Acknowledgments We thank our lab members for critical reading ofthe manuscript.

Conflicts of Interest The authors declare no conflicts of interest.

Authors’Contributions GUwrote themanuscript; SS and RKS editedthe manuscript. All authors read and approved the manuscript.

References

1. Patel S, Singh V, Kumar A, Gupta YK, Singh MP (2006) Status ofantioxidant defense system and expression of toxicant responsivegenes in striatum of maneb- and paraquat-induced Parkinson’sdisease phenotype in mouse: mechanism of neurodegeneration.Brain Res 1081(1):9–18. doi:10.1016/j.brainres.2006.01.060

2. Ferrante RJ, Kowall NW, Beal MF, Richardson EP Jr, Bird ED,Martin JB (1985) Selective sparing of a class of striatal neurons inHuntington’s disease. Science 230(4725):561–563

3. Boillee S, Vande Velde C, Cleveland DW (2006) ALS: a disease ofmotor neurons and their nonneuronal neighbors. Neuron 52(1):39–59. doi:10.1016/j.neuron.2006.09.018

4. Rando TA, Wyss-Coray T Stem cells as vehicles for youthfulregeneration of aged tissues. J Gerontol A Biol Sci Med Sci 69Suppl 1:S39-42. doi:10.1093/gerona/glu043

5. Orlacchio A, Bernardi G, Martino S (2010) Stem cells: an overviewof the current status of therapies for central and peripheral nervoussystem diseases. Curr Med Chem 17(7):595–608

6. SendtnerM (2009) Stem cells: tailor-made diseased neurons. Nature457(7227):269–270. doi:10.1038/457269a

7. Yu D, Silva GA (2008) Stem cell sources and therapeutic ap-proaches for central nervous system and neural retinal disorders.Neurosurg Focus 24(3–4):E11. doi:10.3171/FOC/2008/24/3–4/E10

8. Park DH, Eve DJ (2009) Regenerative medicine: advances in newmethods and technologies. Med Sci Monit 15(11):RA233–251

9. Galli R, Gritti A, Vescovi AL (2008) Adult neural stem cells.Methods Mol Biol 438:67–84. doi:10.1007/978–1–59745–133–8_7

10. Srivastava AS, Malhotra R, Sharp J, Berggren T (2008) Potentialsof ES cell therapy in neurodegenerative diseases. Curr Pharm Des14(36):3873–3879

11. Park IH, Lerou PH, ZhaoR, HuoH, Daley GQ (2008)Generation ofhuman-induced pluripotent stem cells. Nat Protoc 3(7):1180–1186.doi:10.1038/nprot.2008.92

12. Conti L, Cattaneo E, Papadimou E (2008) Novel neural stem cellsystems. Expert Opin Biol Ther 8(2):153–160. doi:10.1517/14712598.8.2.153

13. Ma DK, Bonaguidi MA, Ming GL, Song H (2009) Adult neuralstem cells in the mammalian central nervous system. Cell Res 19(6):672–682. doi:10.1038/cr.2009.56

14. Lindvall O, Kokaia Z,Martinez-Serrano A (2004) Stem cell therapyfor human neurodegenerative disorders—how to make it work. NatMed 10(Suppl):S42–50. doi:10.1038/nm1064

15. Rayment EA, Williams DJ (2010) Concise review: mind the gap:challenges in characterizing and quantifying cell- and tissue-basedtherapies for clinical translation. Stem Cells 28(5):996–1004. doi:10.1002/stem.416

16. Osanai T, Kuroda S, Sugiyama T, Kawabori M, Ito M, ShichinoheH, Kuge Y, Houkin K, Tamaki N, Iwasaki Y Therapeutic effects ofintra-arterial delivery of bone marrow stromal cells in traumaticbrain injury of rats—in vivo cell tracking study by near-infraredfluorescence imaging. Neurosurgery 70 (2):435–444; discussion444. doi:10.1227/NEU.0b013e318230a795

17. Baker PS, Brown GC (2009) Stem-cell therapy in retinal disease.Curr Opin Ophthalmol 20(3):175–181

18. Enns GM, Huhn SL (2008) Central nervous system therapy forlysosomal storage disorders. Neurosurg Focus 24(3–4):E12. doi:10.3171/FOC/2008/24/3–4/E11

19. Martino S, di Girolamo I, Orlacchio A, Datti A (2009) MicroRNAimplications across neurodevelopment and neuropathology. JBiomed Biotechnol 2009:654346. doi:10.1155/2009/654346

20. Naegele JR, Maisano X, Yang J, Royston S, Ribeiro E (2010)Recent advancements in stem cell and gene therapies for neurolog-ical disorders and intractable epilepsy. Neuropharmacology 58(6):855–864. doi:10.1016/j.neuropharm.2010.01.019

21. Lindvall O, Kokaia Z (2010) Stem cells in human neurodegenera-tive disorders—time for clinical translation? J Clin Invest 120(1):29–40. doi:10.1172/JCI40543

22. Dawson E, Mapili G, Erickson K, Taqvi S, Roy K (2008)Biomaterials for stem cell differentiation. Adv Drug Deliv Rev60(2):215–228. doi:10.1016/j.addr.2007.08.037

23. Atala A (2009) Engineering organs. Curr Opin Biotechnol 20(5):575–592. doi:10.1016/j.copbio.2009.10.003

24. Zhong Y, Bellamkonda RV (2008) Biomaterials for the centralnervous system. J R Soc Interface 5(26):957–975. doi:10.1098/rsif.2008.0071

25. Martino S, D’Angelo F, Armentano I, Tiribuzi R, Pennacchi M,Dottori M, Mattioli S, Caraffa A, Cerulli GG, Kenny JM, OrlacchioA (2009) Hydrogenated amorphous carbon nanopatterned film de-signs drive human bone marrow mesenchymal stem cell cytoskel-eton architecture. Tissue Eng Part A 15(10):3139–3149. doi:10.1089/ten.TEA.2008.0552

26. Orive G, Anitua E, Pedraz JL, Emerich DF (2009) Biomaterials forpromoting brain protection, repair and regeneration. Nat RevNeurosci 10(9):682–692. doi:10.1038/nrn2685

27. Takahashi K, Yamanaka S (2006) Induction of pluripotentstem cells from mouse embryonic and adult fibroblast cul-tures by defined factors. Cell 126(4):663–676. doi:10.1016/j.cell.2006.07.024

Mol Neurobiol

Page 12: Mol Neurobiol

28. Takahashi J (2007) Stem cell therapy for Parkinson’s disease. ExpertRev Neurother 7(6):667–675. doi:10.1586/14737175.7.6.667

29. Yu SC, Yi L, Zhou ZH, Yao XH, Ping YF, Bian XW (2007)Isolation and identification of tumor stem-like cells from humanglioma cell line U87 after treatment of vincristine. Ai Zheng 26(12):1388–1391

30. Yu Y, Gu S, HuangH,Wen T (2007) Combination of bFGF, heparinand laminin induce the generation of dopaminergic neurons from ratneural stem cells both in vitro and in vivo. J Neurol Sci 255(1–2):81–86. doi:10.1016/j.jns.2007.01.076

31. HovakimyanM, Haas SJ, Schmitt O, Gerber B,Wree A, AndressenC (2008)Mesencephalic human neural progenitor cells transplantedinto the adult hemiparkinsonian rat striatum lack dopaminergicdifferentiation but improve motor behavior. Cells Tissues Organs188(4):373–383. doi:10.1159/000140680

32. Park HJ, Lee PH, Bang OY, Lee G, Ahn YH (2008) Mesenchymalstem cells therapy exerts neuroprotection in a progressive animalmodel of Parkinson’s disease. J Neurochem 107(1):141–151. doi:10.1111/j.1471–4159.2008.05589.x

33. Vierbuchen T, Ostermeier A, Pang ZP, Kokubu Y, Sudhof TC,Wernig M (2010) Direct conversion of fibroblasts to functionalneurons by defined factors. Nature 463(7284):1035–1041. doi:10.1038/nature08797

34. Marro S, Pang ZP, Yang N, Tsai MC, QuK, Chang HY, Sudhof TC,Wernig M (2011) Direct lineage conversion of terminally differen-tiated hepatocytes to functional neurons. Cell Stem Cell 9(4):374–382. doi:10.1016/j.stem.2011.09.002

35. Pang ZP, Yang N, Vierbuchen T, Ostermeier A, Fuentes DR, YangTQ, Citri A, Sebastiano V, Marro S, Sudhof TC, Wernig M (2011)Induction of human neuronal cells by defined transcription factors.Nature 476(7359):220–223. doi:10.1038/nature10202

36. Yoo AS, Sun AX, Li L, Shcheglovitov A, Portmann T, Li Y, Lee-Messer C, Dolmetsch RE, Tsien RW, Crabtree GR (2011)MicroRNA-mediated conversion of human fibroblasts to neurons.Nature 476(7359):228–231. doi:10.1038/nature10323

37. Ebert AD, Yu J, Rose FF Jr, Mattis VB, Lorson CL, Thomson JA,Svendsen CN (2009) Induced pluripotent stem cells from a spinalmuscular atrophy patient. Nature 457(7227):277–280. doi:10.1038/nature07677

38. Lee G, Papapetrou EP, Kim H, Chambers SM, Tomishima MJ,Fasano CA, Ganat YM, Menon J, Shimizu F, Viale A, Tabar V,Sadelain M, Studer L (2009) Modelling pathogenesis and treatmentof familial dysautonomia using patient-specific iPSCs. Nature461(7262):402–406. doi:10.1038/nature08320

39. Pfisterer U, Kirkeby A, Torper O, Wood J, Nelander J, Dufour A,Bjorklund A, Lindvall O, Jakobsson J, Parmar M (2011) Directconversion of human fibroblasts to dopaminergic neurons. ProcNatl Acad Sci U S A 108(25):10343–10348. doi:10.1073/pnas.1105135108

40. Son EY, Ichida JK, Wainger BJ, Toma JS, Rafuse VF, Woolf CJ,Eggan K (2011) Conversion of mouse and human fibroblasts intofunctional spinal motor neurons. Cell Stem Cell 9(3):205–218. doi:10.1016/j.stem.2011.07.014

41. Marchetto MC, Carromeu C, Acab A, Yu D, Yeo GW, Mu Y, ChenG, Gage FH, Muotri AR (2010) A model for neural developmentand treatment of Rett syndrome using human induced pluripotentstem cells. Cell 143(4):527–539. doi:10.1016/j.cell.2010.10.016

42. Cheung AY, Horvath LM, Grafodatskaya D, Pasceri P, Weksberg R,Hotta A, Carrel L, Ellis J (2011) Isolation of MECP2-null Rettsyndrome patient hiPS cells and isogenic controls through X-chromosome inactivation. Hum Mol Genet 20(11):2103–2115.doi:10.1093/hmg/ddr093

43. Lujan E, Chanda S, Ahlenius H, Sudhof TC, Wernig M (2012)Direct conversion of mouse fibroblasts to self-renewing, tripotentneural precursor cells. Proc Natl Acad Sci U S A 109(7):2527–2532. doi:10.1073/pnas.1121003109

44. Han DW, Tapia N, Hermann A, Hemmer K, Hoing S, Arauzo-Bravo MJ, Zaehres H, Wu G, Frank S, Moritz S, Greber B, YangJH, Lee HT, Schwamborn JC, Storch A, Scholer HR (2012) Directreprogramming of fibroblasts into neural stem cells by definedfactors. Cell Stem Cell 10(4):465–472. doi:10.1016/j.stem.2012.02.021

45. Nguyen HN, Byers B, Cord B, Shcheglovitov A, Byrne J, Gujar P,Kee K, Schule B, Dolmetsch RE, LangstonW, Palmer TD, Pera RR(2011) LRRK2 mutant iPSC-derived DA neurons demonstrate in-creased susceptibility to oxidative stress. Cell Stem Cell 8(3):267–280. doi:10.1016/j.stem.2011.01.013

46. Seibler P, Graziotto J, Jeong H, Simunovic F, Klein C, Krainc D(2011) Mitochondrial Parkin recruitment is impaired in neuronsderived from mutant PINK1 induced pluripotent stem cells. JNeurosci 31(16):5970–5976. doi:10.1523/JNEUROSCI.4441–10.2011

47. Ambasudhan R, Talantova M, Coleman R, Yuan X, Zhu S, LiptonSA, Ding S (2011) Direct reprogramming of adult human fibro-blasts to functional neurons under defined conditions. Cell StemCell 9(2):113–118. doi:10.1016/j.stem.2011.07.002

48. Caiazzo M, Dell’Anno MT, Dvoretskova E, Lazarevic D, TavernaS, Leo D, Sotnikova TD, Menegon A, Roncaglia P, Colciago G,Russo G, Carninci P, Pezzoli G, Gainetdinov RR, Gustincich S,Dityatev A, Broccoli V (2011) Direct generation of functionaldopaminergic neurons from mouse and human fibroblasts. Nature476(7359):224–227. doi:10.1038/nature10284

49. Ring KL, Tong LM, BalestraME, Javier R, Andrews-Zwilling Y, LiG, Walker D, Zhang WR, Kreitzer AC, Huang Y (2012) Directreprogramming of mouse and human fibroblasts into multipotentneural stem cells with a single factor. Cell Stem Cell 11(1):100–109.doi:10.1016/j.stem.2012.05.018

50. Hashimoto-Torii K, Torii M, Fujimoto M, Nakai A, El Fatimy R,Mezger V, Ju MJ, Ishii S, Chao SH, Brennand KJ, Gage FH, RakicP (2014) Roles of heat shock factor 1 in neuronal response to fetalenvironmental risks and its relevance to brain disorders. Neuron82(3):560–572. doi:10.1016/j.neuron.2014.03.002

51. Mitne-Neto M, Machado-Costa M, Marchetto MC, Bengtson MH,Joazeiro CA, Tsuda H, Bellen HJ, Silva HC, Oliveira AS, Lazar M,Muotri AR, Zatz M (2011) Downregulation of VAPB expression inmotor neurons derived from induced pluripotent stem cells of ALS8patients. Hum Mol Genet 20(18):3642–3652. doi:10.1093/hmg/ddr284

52. Brennand KJ, Simone A, Jou J, Gelboin-Burkhart C, Tran N,Sangar S, Li Y, Mu Y, Chen G, Yu D, McCarthy S, Sebat J, GageFH (2011) Modelling schizophrenia using human induced pluripo-tent stem cells. Nature 473(7346):221–225. doi:10.1038/nature09915

53. Kim J, Su SC, Wang H, Cheng AW, Cassady JP, Lodato MA,Lengner CJ, Chung CY, Dawlaty MM, Tsai LH, Jaenisch R(2011) Functional integration of dopaminergic neurons directlyconverted from mouse fibroblasts. Cell Stem Cell 9(5):413–419.doi:10.1016/j.stem.2011.09.011

54. Rais Y, Zviran A, Geula S, Gafni O, Chomsky E, Viukov S,Mansour AA, Caspi I, Krupalnik V, Zerbib M, Maza I, Mor N,Baran D, Weinberger L, Jaitin DA, Lara-Astiaso D, Blecher-GonenR, Shipony Z, Mukamel Z, Hagai T, Gilad S, Amann-ZalcensteinD, Tanay A, Amit I, Novershtern N, Hanna JH Deterministic directreprogramming of somatic cells to pluripotency. Nature 502 (7469):65–70. doi:10.1038/nature1258

55. Salewski RP, Eftekharpour E, Fehlings MG (2010) Are inducedpluripotent stem cells the future of cell-based regenerative therapiesfor spinal cord injury? J Cell Physiol 222(3):515–521. doi:10.1002/jcp.21995

56. Kiskinis E, Eggan K (2010) Progress toward the clinical applicationof patient-specific pluripotent stem cells. J Clin Invest 120(1):51–59. doi:10.1172/JCI40553

Mol Neurobiol

Page 13: Mol Neurobiol

57. Zhang ZG, Chopp M (2009) Neurorestorative therapies for stroke:underlying mechanisms and translation to the clinic. Lancet Neurol8(5):491–500. doi:10.1016/S1474–4422(09)70061–4

58. Li JY, Christophersen NS, Hall V, Soulet D, Brundin P(2008) Critical issues of clinical human embryonic stem celltherapy for brain repair. Trends Neurosci 31(3):146–153. doi:10.1016/j.tins.2007.12.001

59. Bachoud-Levi AC, Gaura V, Brugieres P, Lefaucheur JP, BoisseMF, Maison P, Baudic S, Ribeiro MJ, Bourdet C, Remy P, Cesaro P,Hantraye P, PeschanskiM (2006) Effect of fetal neural transplants inpatients with Huntington’s disease 6 years after surgery: a long-termfollow-up study. Lancet Neurol 5(4):303–309. doi:10.1016/S1474–4422(06)70381–7

60. Deda H, Inci MC, Kurekci AE, Sav A, Kayihan K, Ozgun E,Ustunsoy GE, Kocabay S (2009) Treatment of amyotrophic lateralsclerosis patients by autologous bone marrow-derived hematopoi-etic stem cell transplantation: a 1 year follow-up. Cytotherapy 11(1):18–25. doi:10.1080/14653240802549470

61. Appel SH, Engelhardt JI, Henkel JS, Siklos L, Beers DR, Yen AA,Simpson EP, Luo Y, Carrum G, Heslop HE, Brenner MK, Popat U(2008) Hematopoietic stem cell transplantation in patients withsporadic amyotrophic lateral sclerosis. Neurology 71(17):1326–1334. doi:10.1212/01.wnl.0000327668.43541.22

62. Akesson E, Sandelin M, Kanaykina N, Aldskogius H, Kozlova EN(2008) Long-term survival, robust neuronal differentiation, andextensive migration of human forebrain stem/progenitor cellstransplanted to the adult rat dorsal root ganglion cavity. CellTransplant 17(10–11):1115–1123

63. Biffi A, Lucchini G, Rovelli A, Sessa M (2008) Metachromaticleukodystrophy: an overview of current and prospective treatments.BoneMarrow Transplant 42(Suppl 2):S2–6. doi:10.1038/bmt.2008.275

64. Lindvall O, Kokaia Z (2006) Stem cells for the treatment of neuro-logical disorders. Nature 441(7097):1094–1096. doi:10.1038/nature04960

65. Wernig M, Zhao JP, Pruszak J, Hedlund E, Fu D, Soldner F,Broccoli V, Constantine-Paton M, Isacson O, Jaenisch R (2008)Neurons derived from reprogrammed fibroblasts functionally inte-grate into the fetal brain and improve symptoms of rats withParkinson’s disease. Proc Natl Acad Sci U S A 105(15):5856–5861. doi:10.1073/pnas.0801677105

66. Cai J, Yang M, Poremsky E, Kidd S, Schneider JS, Iacovitti LDopaminergic neurons derived from human induced pluripotentstem cells survive and integrate into 6-OHDA-lesioned rats. StemCells Dev 19 (7):1017–1023. doi:10.1089/scd.2009.0319

67. Hargus G, Cooper O, Deleidi M, LevyA, Lee K,MarlowE, YowA,Soldner F, Hockemeyer D, Hallett PJ, Osborn T, Jaenisch R, IsacsonODifferentiated Parkinson patient-derived induced pluripotent stemcells grow in the adult rodent brain and reduce motor asymmetry inparkinsonian rats. Proc Natl Acad Sci U S A 107 (36):15921–15926. doi:10.1073/pnas.1010209107

68. Jonsson ME, Ono Y, Bjorklund A, Thompson LH (2009)Identification of transplantable dopamine neuron precursors at dif-ferent stages of midbrain neurogenesis. Exp Neurol 219(1):341–354. doi:10.1016/j.expneurol.2009.06.006

69. Rodriguez-Gomez JA, Lu JQ, Velasco I, Rivera S, Zoghbi SS, LiowJS, Musachio JL, Chin FT, Toyama H, Seidel J, Green MV, ThanosPK, Ichise M, Pike VW, Innis RB, McKay RD (2007) Persistentdopamine functions of neurons derived from embryonic stem cellsin a rodent model of Parkinson disease. Stem Cells 25(4):918–928.doi:10.1634/stemcells.2006–0386

70. Roy NS, Cleren C, Singh SK, Yang L, Beal MF, Goldman SA(2006) Functional engraftment of human ES cell-derived dopami-nergic neurons enriched by coculture with telomerase-immortalizedmidbrain astrocytes. Nat Med 12(11):1259–1268. doi:10.1038/nm1495

71. Cho MS, Lee YE, Kim JY, Chung S, Cho YH, Kim DS, Kang SM,Lee H, KimMH, Kim JH, Leem JW, Oh SK, Choi YM, HwangDY,Chang JW, Kim DW (2008) Highly efficient and large-scale gener-ation of functional dopamine neurons from human embryonic stemcells. Proc Natl Acad Sci U S A 105(9):3392–3397. doi:10.1073/pnas.0712359105

72. Ben-Hur T, Idelson M, Khaner H, Pera M, Reinhartz E, Itzik A,Reubinoff BE (2004) Transplantation of human embryonic stemcell-derived neural progenitors improves behavioral deficit in par-kinsonian rats. Stem Cells 22(7):1246–1255. doi:10.1634/stemcells.2004–0094

73. Kim JH, Auerbach JM, Rodriguez-Gomez JA, Velasco I, Gavin D,Lumelsky N, Lee SH, Nguyen J, Sanchez-Pernaute R, BankiewiczK, McKay R (2002) Dopamine neurons derived from embryonicstem cells function in an animal model of Parkinson’s disease.Nature 418(6893):50–56. doi:10.1038/nature00900

74. Nishimura F, Yoshikawa M, Kanda S, Nonaka M, Yokota H, ShiroiA, Nakase H, Hirabayashi H, Ouji Y, Birumachi J, Ishizaka S,Sakaki T (2003) Potential use of embryonic stem cells for thetreatment of mouse parkinsonian models: improved behavior bytransplantation of in vitro differentiated dopaminergic neurons fromembryonic stem cells. Stem Cells 21(2):171–180. doi:10.1634/stemcells.21–2–171

75. Barberi T, Klivenyi P, Calingasan NY, Lee H, Kawamata H,Loonam K, Perrier AL, Bruses J, Rubio ME, Topf N, Tabar V,Harrison NL, BealMF,MooreMA, Studer L (2003) Neural subtypespecification of fertilization and nuclear transfer embryonic stemcells and application in parkinsonian mice. Nat Biotechnol 21(10):1200–1207. doi:10.1038/nbt870

76. Tabar V, Tomishima M, Panagiotakos G, Wakayama S, Menon J,Chan B, Mizutani E, Al-Shamy G, Ohta H, Wakayama T, Studer L(2008) Therapeutic cloning in individual parkinsonian mice. NatMed 14(4):379–381. doi:10.1038/nm1732

77. Villaescusa JC, Arenas E (2010) Transplantable midbrain dopamineneurons: a moving target. Exp Neurol 222(2):173–178. doi:10.1016/j.expneurol.2009.12.028

78. Brundin P, Strecker RE, Lindvall O, Isacson O, Nilsson OG, BarbinG, Prochiantz A, Forni C, Nieoullon A, Widner H et al (1987)Intracerebral grafting of dopamine neurons. Experimental basis forclinical trials in patients with Parkinson’s disease. Ann N Y AcadSci 495:473–496

79. Nishimura K, Takahashi J Therapeutic application of stem celltechnology toward the treatment of Parkinson’s disease. BiolPharm Bull 36 (2):171–175

80. Tarazi FI, Sahli ZT, Wolny M, Mousa SA Emerging therapies forParkinson’s disease: from bench to bedside. Pharmacol Ther. doi:10.1016/j.pharmthera.2014.05.010

81. Mendez I, Sanchez-Pernaute R, Cooper O, Vinuela A, Ferrari D,Bjorklund L, Dagher A, Isacson O (2005) Cell type analysis offunctional fetal dopamine cell suspension transplants in the striatumand substantia nigra of patients with Parkinson’s disease. Brain128(Pt 7):1498–1510. doi:10.1093/brain/awh510

82. Geraerts M, Krylyshkina O, Debyser Z, Baekelandt V (2007)Concise review: therapeutic strategies for Parkinson disease basedon the modulation of adult neurogenesis. Stem Cells 25(2):263–270. doi:10.1634/stemcells.2006–0364

83. Friling S, Andersson E, Thompson LH, Jonsson ME,Hebsgaard JB, Nanou E, Alekseenko Z, Marklund U,Kjellander S, Volakakis N, Hovatta O, El Manira A,Bjorklund A, Perlmann T, Ericson J (2009) Efficient produc-tion of mesencephalic dopamine neurons by Lmx1a expres-sion in embryonic stem cells. Proc Natl Acad Sci U S A106(18):7613–7618. doi:10.1073/pnas.0902396106

84. Hallett M, Litvan I (1999) Evaluation of surgery for Parkinson’sdisease: a report of the Therapeutics and Technology AssessmentSubcommittee of the American Academy of Neurology. The Task

Mol Neurobiol

Page 14: Mol Neurobiol

Force on Surgery for Parkinson’s Disease. Neurology 53(9):1910–1921

85. Hallett M, Litvan I (2000) Scientific position paper of theMovement Disorder Society evaluation of surgery for Parkinson’sdisease. Task Force on Surgery for Parkinson’s Disease of theAmerican Academy of Neurology Therapeutic and TechnologyAssessment Committee. Mov Disord 15(3):436–438

86. Freed CR, Greene PE, Breeze RE, Tsai WY, DuMouchel W, Kao R,Dillon S,Winfield H, Culver S, Trojanowski JQ, Eidelberg D, FahnS (2001) Transplantation of embryonic dopamine neurons for se-vere Parkinson’s disease. N Engl J Med 344(10):710–719. doi:10.1056/NEJM200103083441002

87. Olanow CW, Goetz CG, Kordower JH, Stoessl AJ, Sossi V, BrinMF, Shannon KM, Nauert GM, Perl DP, Godbold J, Freeman TB(2003) A double-blind controlled trial of bilateral fetal nigral trans-plantation in Parkinson’s disease. Ann Neurol 54(3):403–414. doi:10.1002/ana.10720

88. Bjorklund A, Dunnett SB, Brundin P, Stoessl AJ, Freed CR, BreezeRE, Levivier M, Peschanski M, Studer L, Barker R (2003) Neuraltransplantation for the treatment of Parkinson’s disease. LancetNeurol 2(7):437–445

89. Freed CR, Leehey MA, Zawada M, Bjugstad K, Thompson L,Breeze RE (2003) Do patients with Parkinson’s disease benefit fromembryonic dopamine cell transplantation? J Neurol 250(Suppl 3):III44–46. doi:10.1007/s00415–003–1308–5

90. Freed CR, Breeze RE, Fahn S, Eidelberg D (2004) Preoperativeresponse to levodopa is the best predictor of transplant outcome.Ann Neurol 55 (6):896; author reply 896–897. doi:10.1002/ana.20085

91. Byers B, Cord B, Nguyen HN, Schule B, Fenno L, Lee PC,Deisseroth K, Langston JW, Pera RR, Palmer TD (2011)SNCA triplication Parkinson’s patient’s iPSC-derived DAneurons accumulate alpha-synuclein and are susceptible tooxidative stress. PLoS One 6(11):e26159. doi:10.1371/journal.pone.0026159

92. Sagal J, Zhan X, Xu J, Tilghman J, Karuppagounder SS, Chen L,Dawson VL, Dawson TM, Laterra J, Ying M Proneural transcrip-tion factor Atoh1 drives highly efficient differentiation of humanpluripotent stem cells into dopaminergic neurons. Stem Cells TranslMed. doi:10.5966/sctm.2013–0213

93. ThiesW, Bleiler L, Alzheimer’s A (2013) 2013 Alzheimer’s diseasefacts and figures. Alzheimers Dement 9(2):208–245. doi:10.1016/j.jalz.2013.02.003

94. Gurav AN (2014) Alzheimer’s disease and periodontitis—an elu-sive link. Rev Assoc Med Bras 60(2):173–180

95. Hansen N (2014) Brain stimulation for combating Alzheimer’sdisease. Front Neurol 5:80. doi:10.3389/fneur.2014.00080

96. Babaei P, Soltani Tehrani B, Alizadeh ATransplanted bone marrowmesenchymal stem cells improve memory in rat models ofAlzheimer’s disease. Stem Cells Int 2012:369417. doi:10.1155/2012/369417

97. Lee JK, Jin HK, Bae JS (2009) Bone marrow-derived mesenchymalstem cells reduce brain amyloid-beta deposition and accelerate theactivation of microglia in an acutely induced Alzheimer’s diseasemouse model. Neurosci Lett 450(2):136–141. doi:10.1016/j.neulet.2008.11.059

98. Park D, Lee HJ, Joo SS, Bae DK, Yang G, Yang YH, Lim I, MatsuoA, Tooyama I, Kim YB, Kim SU Human neural stem cells over-expressing choline acetyltransferase restore cognition in rat modelof cognitive dysfunction. Exp Neurol 234 (2):521–526. doi:10.1016/j.expneurol.2011.12.040

99. Park DH, Eve DJ, Sanberg PR, Musso J, 3rd, Bachstetter AD,Wolfson A, Schlunk A, Baradez MO, Sinden JD, Gemma CIncreased neuronal proliferation in the dentate gyrus of aged ratsfollowing neural stem cell implantation. Stem Cells Dev 19 (2):175–180. doi:10.1089/scd.2009.0172

100. Qu T, Brannen CL, Kim HM, Sugaya K (2001) Human neural stemcells improve cognitive function of aged brain. Neuroreport 12(6):1127–1132

101. Kern DS, Maclean KN, Jiang H, Synder EY, Sladek JR, Jr.,Bjugstad KB Neural stem cells reduce hippocampal tau andreelin accumulation in aged Ts65Dn Down syndrome mice.C e l l T r a n s p l a n t 2 0 ( 3 ) : 3 7 1 – 3 79 . d o i : 1 0 . 3 7 2 7 /096368910X528085

102. Blurton-Jones M, Kitazawa M, Martinez-Coria H, Castello NA,Muller FJ, Loring JF, Yamasaki TR, Poon WW, Green KN,LaFerla FM (2009) Neural stem cells improve cognition viaBDNF in a transgenic model of Alzheimer disease. Proc NatlAcad Sci U S A 106(32):13594–13599. doi:10.1073/pnas.0901402106

103. Yamasaki TR, Blurton-Jones M, Morrissette DA, Kitazawa M,Oddo S, LaFerla FM (2007) Neural stem cells improve memoryin an inducible mouse model of neuronal loss. J Neurosci 27(44):11925–11933. doi:10.1523/JNEUROSCI.1627–07.2007

104. Xuan AG, Luo M, Ji WD, Long DH (2009) Effects of engraftedneural stem cells in Alzheimer’s disease rats. Neurosci Lett 450(2):167–171. doi:10.1016/j.neulet.2008.12.001

105. Kim S, Chang KA, Kim J, Park HG, Ra JC, Kim HS, Suh YH Thepreventive and therapeutic effects of intravenous human adipose-derived stem cells in Alzheimer’s disease mice. PLoS One 7 (9):e45757. doi:10.1371/journal.pone.0045757

106. Wang Q, Matsumoto Y, Shindo T, Miyake K, Shindo A, KawanishiM, Kawai N, Tamiya T, Nagao S (2006) Neural stem cells trans-plantation in cortex in a mouse model of Alzheimer’s disease. JMedInvest 53(1–2):61–69

107. Esmaeilzade B, Nobakht M, Joghataei MT, Rahbar Roshandel N,Rasouli H, Samadi Kuchaksaraei A, Hosseini SM, Najafzade N,Asalgoo S, Hejazian LB, Moghani Ghoroghi F Delivery of epider-mal neural crest stem cells (EPI-NCSC) to hippocamp inAlzheimer’s disease rat model. Iran Biomed J 16 (1):1–9

108. Lee GJ, Lu PH, Medina LD, Rodriguez-Agudelo Y, Melchor S,Coppola G, Braskie MN, Hua X, Apostolova LG, Leow AD,Thompson PM, Ringman JM Regional brain volume differencesin symptomatic and presymptomatic carriers of familial Alzheimer’sdisease mutations. J Neurol Neurosurg Psychiatry 84 (2):154–162.doi:10.1136/jnnp-2011–302087

109. Wu L, Sluiter AA, Guo HF, Balesar RA, Swaab DF, Zhou JN,Verwer RW (2008) Neural stem cells improve neuronal survival incultured postmortem brain tissue from aged and Alzheimer patients.J Cell Mol Med 12(5A):1611–1621. doi:10.1111/j.1582–4934.2007.00203.x

110. Sugaya K,Merchant S (2008) How to approachAlzheimer’s diseasetherapy using stem cell technologies. J Alzheimers Dis 15(2):241–254

111. Waldau B, Shetty AK (2008) Behavior of neural stem cells in theAlzheimer brain. Cell Mol Life Sci 65(15):2372–2384. doi:10.1007/s00018–008–8053-y

112. Ying QL, Stavridis M, Griffiths D, Li M, Smith A (2003)Conversion of embryonic stem cells into neuroectodermal precur-sors in adherent monoculture. Nat Biotechnol 21(2):183–186. doi:10.1038/nbt780

113. Xu Y, Tamamaki N, Noda T, Kimura K, Itokazu Y, Matsumoto N,Dezawa M, Ide C (2005) Neurogenesis in the ependymal layer ofthe adult rat 3rd ventricle. Exp Neurol 192(2):251–264. doi:10.1016/j.expneurol.2004.12.021

114. Moghadam FH, Alaie H, Karbalaie K, Tanhaei S, Nasr EsfahaniMH, Baharvand H (2009) Transplantation of primed or unprimedmouse embryonic stem cell-derived neural precursor cells improvescognitive function in Alzheimerian rats. Differentiation 78(2–3):59–68. doi:10.1016/j.diff.2009.06.005

115. Tang J, Xu H, Fan X, Li D, Rancourt D, Zhou G, Li Z, Yang L(2008) Embryonic stem cell-derived neural precursor cells improve

Mol Neurobiol

Page 15: Mol Neurobiol

memory dysfunction in Abeta (1–40) injured rats. Neurosci Res62(2):86–96. doi:10.1016/j.neures.2008.06.005

116. Schwartz CM, Tavakoli T, Jamias C, Park SS, Maudsley S,Martin B, Phillips TM, Yao PJ, Itoh K, Ma W, Rao MS,Arenas E, Mattson MP Stromal factors SDF1alpha, sFRP1,and VEGFD induce dopaminergic neuron differentiation ofhuman pluripotent stem cells. J Neurosci Res 90 (7):1367–1381. doi:10.1002/jnr.23064

117. Cho EG, Zaremba JD, McKercher SR, Talantova M, Tu S, MasliahE, Chan SF, Nakanishi N, Terskikh A, Lipton SAMEF2C enhancesdopaminergic neuron differentiation of human embryonic stem cellsin a parkinsonian rat model. PLoS One 6 (8):e24027. doi:10.1371/journal.pone.0024027PONE-D-11–07261

118. Liu Y, Weick JP, Liu H, Krencik R, Zhang X, Ma L, Zhou GM,Ayala M, Zhang SC Medial ganglionic eminence-like cells derivedfrom human embryonic stem cells correct learning and memorydeficits. Nat Biotechnol 31 (5):440–447. doi:10.1038/nbt.2565

119. Fan X, Sun D, TangX, Cai Y, Yin ZQ, XuH Stem-cell challenges inthe treatment of Alzheimer’s disease: a long way from bench tobedside. Med Res Rev. doi:10.1002/med.21309

120. Mohamet L, Miazga NJ, Ward CM Familial Alzheimer’s diseasemodelling using induced pluripotent stem cell technology. World JStem Cells 6 (2):239–247. doi:10.4252/wjsc.v6.i2.239

121. Yagi T, Ito D, Okada Y, Akamatsu W, Nihei Y, Okano H, Suzuki N[Modeling familial Alzheimer’s disease with induced pluripotentstem cells]. Rinsho Shinkeigaku 52 (11):1134–1136

122. Yagi T, Ito D, Okada Y, Akamatsu W, Nihei Y, Yoshizaki T,Yamanaka S, Okano H, Suzuki N Modeling familial Alzheimer’sdisease with induced pluripotent stem cells. Hum Mol Genet 20(23):4530–4539. doi:10.1093/hmg/ddr394

123. Yahata N, Asai M, Kitaoka S, Takahashi K, Asaka I, Hioki H,Kaneko T, Maruyama K, Saido TC, Nakahata T, Asada T,Yamanaka S, Iwata N, Inoue H Anti-Abeta drug screening platformusing human iPS cell-derived neurons for the treatment ofAlzheimer’s disease. PLoS One 6 (9):e25788. doi:10.1371/journal.pone.0025788PONE-D-11–09488

124. Kremer B, Goldberg P, Andrew SE, Theilmann J, Telenius H,Zeisler J, Squitieri F, Lin B, Bassett A, Almqvist E et al (1994) Aworldwide study of the Huntington’s disease mutation. The sensi-tivity and specificity of measuring CAG repeats. N Engl J Med330(20):1401–1406. doi:10.1056/NEJM199405193302001

125. Walker FO (2007) Huntington’s disease. Lancet 369(9557):218–228. doi:10.1016/S0140–6736(07)60111–1

126. Im W, Kim M (2014) Cell Therapy strategies vs. paracrine effect inHuntington’s disease. J Mov Disord 7 (1):1–6. doi:10.14802/jmd.14001

127. Visnyei K, Tatsukawa KJ, Erickson RI, Simonian S, Oknaian N,Carmichael ST, Kornblum HI (2006) Neural progenitor implanta-tion restores metabolic deficits in the brain following striatalquinolinic acid lesion. Exp Neurol 197(2):465–474. doi:10.1016/j.expneurol.2005.10.023

128. Vazey EM, Chen K, Hughes SM, Connor B (2006) Transplantedadult neural progenitor cells survive, differentiate and reduce motorfunction impairment in a rodent model of Huntington’s disease. ExpNeurol 199(2):384–396. doi:10.1016/j.expneurol.2006.01.034

129. Amin EM, Reza BA, Morteza BR, Maryam MM, Ali M, Zeinab N(2008) Microanatomical evidences for potential of mesenchymalstem cells in amelioration of striatal degeneration. Neurol Res30(10):1086–1090. doi:10.1179/174313208X327955

130. Juopperi TA, Kim WR, Chiang CH, Yu H, Margolis RL, Ross CA,Ming GL, Song H Astrocytes generated from patient induced plu-ripotent stem cells recapitulate features of Huntington’s diseasepatient cells. Mol Brain 5:17. doi:10.1186/1756–6606–5–17

131. FinkKD, Crane AT, Leveque X, Dues DJ, Huffman LD,MooreAC,Story DT, Dejonge RE, Antcliff A, Starski PA, LuM, Lescaudron L,Rossignol J, Dunbar GL Intrastriatal transplantation of adenovirus-

generated induced pluripotent stem cells for treating neuropatholog-ical and functional deficits in a rodent model of Huntington’sdisease. Stem Cells Transl Med 3 (5):620–631. doi:10.5966/sctm.2013–0151

132. Traxinger K, Kelly C, Johnson BA, Lyles RH, Glass JD (2013)Prognosis and epidemiology of amyotrophic lateral sclerosis: anal-ysis of a clinic population, 1997–2011. Neurol Clin Pract 3(4):313–320. doi:10.1212/CPJ.0b013e3182a1b8ab

133. Vucic S, Rothstein JD, Kiernan MC (2014) Advances intreating amyotrophic lateral sclerosis: insights from patho-physiological studies. Trends Neurosci. doi:10.1016/j.tins.2014.05.006

134. Burrell JR, Vucic S, Kiernan MC (2011) Isolated bulbar phenotypeof amyotrophic lateral sclerosis. Amyotroph Lateral Scler 12(4):283–289. doi:10.3109/17482968.2011.551940

135. Wolfson C, Kilborn S, Oskoui M, Genge A (2009) Incidence andprevalence of amyotrophic lateral sclerosis in Canada: a systematicreview of the literature. Neuroepidemiology 33(2):79–88. doi:10.1159/000222089

136. Sun H, Hou Z, YangH,MengM, Li P, Zou Q, Yang L, Chen Y, ChaiH, Zhong H, Yang ZZ, Zhao J, Lai L, Jiang X, Xiao Z Multiplesystemic transplantations of human amniotic mesenchymal stemcells exert therapeutic effects in an ALS mouse model. Cell TissueRes. doi:10.1007/s00441–014–1903-z

137. Boido M, Piras A, Valsecchi V, Spigolon G, Mareschi K, Ferrero I,Vizzini A, Temi S, Mazzini L, Fagioli F, Vercelli A. Human mes-e nchyma l s t r oma l c e l l t r a n sp l a n t a t i on modu l a t e sneuroinflammatory milieu in a mouse model of amyotrophic lateralsclerosis. Cytotherapy. doi:10.1016/j.jcyt.2014.02.003

138. Chestkov IV, Vasilieva EA, Illarioshkin SN, Lagarkova MA,Kiselev SL Patient-specific induced pluripotent stem cells forSOD1-associated amyotrophic lateral sclerosis pathogenesis stud-ies. Acta Naturae 6 (1):54–60

139. Nizzardo M, Simone C, Rizzo F, Ruggieri M, Salani S, Riboldi G,Faravelli I, Zanetta C, Bresolin N, Comi GP, Corti S Minimallyinvasive transplantation of iPSC-derived ALDHhiSSCloVLA4+neural stem cells effectively improves the phenotype of an amyo-trophic lateral sclerosis model. Hum Mol Genet 23 (2):342–354.doi:10.1093/hmg/ddt425

140. Pan T, Kondo S, Le W, Jankovic J (2008) The role of autophagy–lysosome pathway in neurodegeneration associated withParkinson’s disease. Brain 131(Pt 8):1969–1978. doi:10.1093/brain/awm318

141. Lattanzi A, Neri M, Maderna C, di Girolamo I, Martino S,Orlacchio A, Amendola M, Naldini L, Gritti A (2010)Widespread enzymatic correction of CNS tissues by a singleintracerebral injection of therapeutic lentiviral vector in leu-kodystrophy mouse models. Hum Mol Genet 19(11):2208–2227. doi:10.1093/hmg/ddq099

142. Martino S, di Girolamo I, Cavazzin C, Tiribuzi R, Galli R,Rivaroli A, Valsecchi M, Sandhoff K, Sonnino S, Vescovi A,Gritti A, Orlacchio A (2009) Neural precursor cell culturesfrom GM2 gangliosidosis animal models recapitulate thebiochemical and molecular hallmarks of the brain pathology.J Neurochem 109(1):135–147. doi:10.1111/j.1471–4159.2009.05919.x

143. Martino S, Marconi P, Tancini B, Dolcetta D, De Angelis MG,Montanucci P, Bregola G, Sandhoff K, Bordignon C, Emiliani C,Manservigi R, Orlacchio A (2005) A direct gene transfer strategyvia brain internal capsule reverses the biochemical defect in Tay-Sachs disease. Hum Mol Genet 14(15):2113–2123. doi:10.1093/hmg/ddi216

144. Biffi A, Cesani M, Fumagalli F, Del Carro U, Baldoli C, Canale S,Gerevini S, Amadio S, FalautanoM, Rovelli A, Comi G, RoncaroloMG, Sessa M (2008) Metachromatic leukodystrophy—mutationanalysis provides further evidence of genotype-phenotype

Mol Neurobiol

Page 16: Mol Neurobiol

correlation. Clin Genet 74(4):349–357. doi:10.1111/j.1399–0004.2008.01058.x

145. Hu YF, Gourab K, Wells C, Clewes O, Schmit BD, Sieber-Blum M (2010) Epidermal neural crest stem cell (EPI-NCSC)-mediated recovery of sensory function in a mousemodel of spinal cord injury. Stem Cell Rev 6(2):186–198.doi:10.1007/s12015–010–9152–3

146. Siatskas C, Bernard CC (2009) Stem cell and gene therapeuticstrategies for the treatment of multiple sclerosis. Curr Mol Med9(8):992–1016

147. Bithell A, Williams BP (2005) Neural stem cells and cell replace-ment therapy: making the right cells. Clin Sci (Lond) 108(1):13–22.doi:10.1042/CS20040276->

148. Hermann A, Gastl R, Liebau S, Popa MO, Fiedler J, Boehm BO,Maisel M, Lerche H, Schwarz J, Brenner R, Storch A (2004)Efficient generation of neural stem cell-like cells from adult human

bone marrow stromal cells. J Cell Sci 117(Pt 19):4411–4422. doi:10.1242/jcs.01307

149. Jang YK, Park JJ, Lee MC, Yoon BH, Yang YS, Yang SE,Kim SU (2004) Retinoic acid-mediated induction of neuronsand glial cells from human umbilical cord-derived hemato-poietic stem cells. J Neurosci Res 75(4):573–584. doi:10.1002/jnr.10789

150. Joannides A, Gaughwin P, Schwiening C, Majed H, Sterling J,Compston A, Chandran S (2004) Efficient generation of neuralprecursors from adult human skin: astrocytes promote neurogenesisfrom skin-derived stem cells. Lancet 364(9429):172–178. doi:10.1016/S0140–6736(04)16630–0

151. McGuckin CP, Forraz N, Allouard Q, Pettengell R (2004) Umbilicalcord blood stem cells can expand hematopoietic and neuroglialprogenitors in vitro. Exp Cell Res 295(2):350–359. doi:10.1016/j.yexcr.2003.12.028

Mol Neurobiol