90
Views
11
CrossRef citations to date
0
Altmetric
Review

Mesenchymal stem cells in the treatment of ischemic stroke: progress and possibilities

&
Pages 157-163 | Published online: 12 Nov 2010

Abstract

Stroke is a major cause of death and long-term disability in industrialized countries, and the only causal therapy for stroke comprises recombinant tissue plasminogen activator(rt-PA)-mediated recanalization of the occluded vessel. New experimental strategies focus on neuroregenerative approaches, among which the application of mesenchymal stem cells (MSCs) has gained increasing attention. MSCs, like other stem cells, have the capacity of unlimited self-renewal giving rise to differentiated cells from various cell lineages. Bone marrow (BM)-derived MSCs are the most frequently used MSC type in experimental stroke studies. Application of BM-derived MSCs and, in some studies, transplantation of MSCs from other tissue sources resulted in an improved functional recovery in experimental animals, although stroke volumes were not always affected by MSC transplantation. The underlying precise mechanisms of this phenomenon remain elusive, although MSC transplantation is considered to affect many diverse events, eg, by modulating the inflammatory milieu, stimulating endogenous neurogenesis and angiogenesis, and reducing glial scar formation. On the contrary, neuronal differentiation and integration of transplanted MSCs do not seem to affect stroke outcome significantly. On the basis of these preclinical studies, first clinical trials confirmed improved functional recovery in patients who had received BM-derived MSCs systemically, although the number of patients enrolled in these studies was low and there were no adequate control groups. In this review, we describe some fundamental biological characteristics of MSCs and further review some preclinical experimental studies, with special emphasis on BM-derived MSCs. We also review clinical trials in which MSCs have been used and conclude with a short outlook on the application of MSCs in stroke research.

Introduction

Ischemic stroke is a major cause of death and the leading cause of long-term disability in industrialized countries. The only causal therapy for this devastating disease comprises local or systemic thrombolysis of the occluded vessel by using recombinant tissue plasminogen activator.Citation1Citation3 Thrombolytic therapy, however, is limited by a narrow time window. Only a few stroke patients receive this therapy.Citation4 Despite the complex pathophysiological mechanisms underlying ischemic stroke,Citation5,Citation6 many experimental studies focused on manipulating various cell injury-inducing cascades. Although these studies reported neuroprotective effects of many drugs in animal models of ischemic stroke,Citation7Citation11 a successful translation from bench to bedside is still lacking.Citation12 Hence, the recent experimental focus has shifted from studies on acute neuroprotection toward neuroregenerative approaches with special emphasis on cell-based therapies. Precursor cells and stem cells of different origins have been studied thoroughly in experimental stroke models.Citation13Citation16

Stem cells have unique characteristics as they are capable of unlimited self-renewal giving rise to mature cells from various lineages.Citation16 Based on their origin, stem cells are further classified as embryonic, fetal, and adult stem cells. Although embryonic stem cells and fetal stem cells are considered an attractive source for tissue engineering processes yielding beneficial effects after experimental stroke, their application is limited due to restricted availability, formation of teratomas, and ethical concerns.Citation13,Citation17 Hence, adult stem cells or precursor cells have been thoroughly studied, and their neuroprotective potential after cerebral ischemia has been repeatedly shown.Citation15,Citation16,Citation18Citation21 Application of mesenchymal stem cells (MSCs) has become an interesting tool in stroke research as they offer broad therapeutic strategies, including clinically relevant autologous transplantation of bone marrow (BM)-derived MSCs.Citation22 In this review, we first describe some fundamental characteristics of MSCs with special emphasis on BM-derived MSCs. Thereafter, studies on MSCs in animal models of experimental ischemia will be reviewed, which will be followed by a critical review of MSC-based stroke therapy in patients. We conclude this review with a short outlook on the application of MSCs in ischemic stroke therapy.

Definition and biological characteristics of MSCs

Adult mammalian bone marrows contain a distinct but rare population of stem cells that are critically involved in hematopoiesis giving rise to differentiated cells from various cell lineages such as mesenchymal, neuronal, hepatic, or cardiac cells.Citation22Citation27 These cells are referred to as MSCs, marrow stromal cells, or mesenchymal stromal cells and have been studied intensively for more than 2 decades. However, a lack of common definition and an imprecise terminology have hampered the development of this field until recently.Citation28 For this review, we refer to these cells as mesenchymal stem cells or simply as MSCs. The defining characteristics of MSCs used by investigators are still inconsistent and occasionally induce confusion. Taking into account that isolation methods, expansion methods, and tissue sources differ between various studies, substantial differences between these cultivated MSCs cannot be excluded.Citation29 The Mesenchymal and Tissue Stem Cell Committee of the International Society for Cellular Therapy has suggested some fundamental standards to better define characteristics of MSCs.Citation29 This characterization of MSCs includes 1) cell adherence on plastic surfaces; 2) expression of CD105, CD73, and CD90 while lacking other surface markers such as CD45, CD34, or CD14; and 3) differentiation into osteoblasts, adipocytes, and chondroblasts in vitro.

Several studies suggest that MSCs have unique immunemodulating properties. MSCs have been recognized to evade immune recognition and decrease immune responses, properties that are essential in successful allogeneic transplantation medicine. It has been reported that MSCs do not induce a proliferative lymphocytic response,Citation30Citation32 a finding that might be related to the expression levels of major histocompatibility complex class II molecules.Citation28 Because the postulated immunoprivileged status of MSCs is essentially derived from in vitro experiments, the exact in vivo situation remains unclear. The extent of MSC-mediated immunosuppression in vitro seems to depend on the concentration levels of MSCs in culture; ie, when applied at low concentrations, MSCs induce immune responses rather than depressing them.Citation33 However, there is also some evidence suggesting that MSCs might not be suitable as “universal donor cells” for allogeneic transplantation. Allogeneic MSCs injected in mice have been reported to significantly increase the invasion of various immune-competent cells.Citation34

MSCs are not only harbored within the BM of adult organisms but also found in various other tissues and compartments of both fetal and adult organisms, including blood, placenta, adipose tissue, skin, liver, and lung.Citation35Citation39 Although these cells share many similarities, some differences with regard to their differentiation profile and gene expression patterns still exist.Citation40 BM-derived MSCs are regarded to have the highest multilineage potential, which is a reason for their preferred use in experimental or therapeutic applications. We focus on reviewing recent studies on BM-derived MSCs in experimental stroke research in the next section.

MSCs in experimental stroke research

Neuroprotection after MSC treatment in experimental stroke research has been reported by many authors. However, questions as to the most appropriate application routes, transplantation timing, and mechanisms underlying MSC-mediated neuroprotection remain. Because the majority of MSC experiments in stroke research have been performed on BM-derived MSCs, we first focus on some fundamental studies using this cell type. We first review reports on systemic application of BM-derived MSCs and then the studies on local application of MSCs and conclude this chapter with some examples of non-BM-derived MSCs.

intravenous application of BM-derived MSCs

Although many studies have shown neuroprotection after the intravenous injection of BM-derived MSCs,Citation41Citation48 the underlying mechanisms are still elusive. This is in part due to differences in study designs. MSCs have the capacity to migrate into the ischemic lesion zone,Citation41 possibly via interaction between stromal cell-derived factor (SDF)-1 and its chemokine receptor-4,Citation46 where they act by two distinct mechanisms: 1) secretion of growth factors and 2) stimulation of angiogenesis within the peri-infarct zone. A study by Song et alCitation49 has suggested that MSCs also express and secrete brain natriuretic peptides (BNPs) among other growth factors. BNP, as a close homolog of the atrial natriuretic peptide (ANP), might therefore significantly help reduce the formation of postischemic edema as has been described for ANP before.Citation50 The biological relevance of BNP in cell-based stroke therapy, however, remains elusive because the study by Song et al was performed in vitro only. Growth factors that are secreted by the ischemic host tissue itself, such as vascular endothelial growth factor (VEGF) or epidermal growth factor (EGF), are also thought to be critically involved in MSC-mediated neuroprotection. For instance, Wakabayashi et alCitation47 observed that MSCs secrete insulin-like growth factor-1 followed by an enhanced expression of VEGF, EGF, and basic fibroblast growth factor within endogenous neural cells, which results in reduced infarct injury.Citation47 The systemic injection of MSCs was associated with direct antiapoptotic effects and modulation of inflammatory responses within the ischemic tissue resulting in reduced neural damage in the peri-infarct zone, where glial scar formation has been described to be reduced after MSC transplantation.Citation48,Citation51 As MSC therapy results in enhanced levels of endogenous growth factors such as VEGF, MSCs have been reported to stimulate angiogenesis along the ischemic boundary zone via mechanisms involving enhanced expression of both endogenous VEGF and VEGF receptors.Citation52 Some studies have reported other mechanisms, such as the differentiation of transplanted cells into mature neural cells or the induction of endogenous neurogenesis by enhanced proliferation and differentiation rates of subventricular zone (SVZ)-derived neural precursor cells (NPCs).Citation43,Citation51,Citation53,Citation54 Neuronal differentiation rates observed in these studies were, however, low and are therefore unlikely to substantially improve postischemic brain injury.Citation43,Citation55

Although there is no debate on the beneficial effect of an intravenous BM-derived MSC therapy in experimental stroke research, the most appropriate transplantation timing and the number of cells required for successful transplantation rates are still elusive. Hence, Omori et alCitation56 analyzed MSC-mediated effects on infarct injury and its dependence on both transplantation time and cell dosage. Although postischemic amelioration of brain injury was also observed in animals that received late transplantation of fewer MSCs, ie, a single dose of 1 × 106 cells, MSC therapy was most effective when given within 6 hours after stroke in dosages of 3 × 106 cells. Because no difference with regard to vessel density was observed between the treatment groups, the authors inferred that the early transplantation of more MSCs was beneficial due to immediate neuroprotection rather than the stimulation of postischemic angiogenesis. On the contrary, other studies described significantly improved poststroke recovery in animals that received MSC therapy 4 weeks after stroke,Citation46,Citation57 albeit reduced infarct volumes were only observed in animals that had received MSC treatment within 7 days after stroke.Citation57 Komatsu et alCitation57 observed increased angiogenesis within the peri-infarct area of virtually all animals treated with MSCs, which – in their opinion – might explain the improved functional outcome of MSC-treated experimental groups. Different interpretations made on appropriate systemic MSC transplantation time points depend on the selection of the outcome. Although functional recovery is observed after late and early transplantation, reduction of infarct volume requires early transplantation. However, the assessment of functional recovery in the aforementioned studies was frequently restricted to simple neurological scoring missing a bunch of subtle and reliable motor coordination tests such as the cylinder test, rotarod test, or corner turn test.Citation58

Among the aforementioned studies, a recent work by Zacharek et alCitation59 on the intravenous administration of BM-derived MSCs after transient focal cerebral ischemia showed that BM-derived MSCs proved to ameliorate poststroke functional outcome. The authors observed that MSCs derived from donor animals that had undergone stroke before MSC preparation were superior to MSCs derived from nonischemic animals. In other words, MSCs derived from ischemic rats enhanced angiogenesis, arterial density, and axonal regeneration and modulated growth factor expression patterns within the ischemic tissue of the recipient much more effectively than those derived from nonischemic animals. The mechanism by which ischemic lesions actually influence stem cell properties within the BM compartment was, unfortunately, not analyzed in this study. Hence, further studies on stroke-mediated modulation of BM-derived MSCs and MSCs from other compartments such as the blood are urgently needed.

Other application routes of BM-derived MSCs

Among the application routes, intravenous injection of MSCs is the most studied application protocol in experimental stroke research. However, systemic application of MSCs results in poor neuronal differentiation rates of relatively low intracerebral cell numbers (see above), and studies focusing on neuroregeneration, ie, cell differentiation and integration of transplanted cells, might require higher local cell amounts. Alternatively, some studies analyzed effects of an intra-arterial injection of MSCs after stroke.Citation60Citation62 In line with intravenous transplantation studies, intra-arterial injection of BM-derived MSCs resulted in sustained and improved functional recovery of rodents. MSC-induced enhanced poststroke recovery was associated with increased axonal sprouting, remyelination, and synaptophysin expression, whereas glial scar formation and Nogo-A expression were reduced. Long-term beneficial effects were observed for as long as 1 year, and the MSC-mediated beneficial outcomes were found to be – at least in part – due to the abovementioned structural and molecular changes within the ischemic milieu.Citation60 MSC-mediated beneficial effects on stroke outcome in the latter study were already observed at 2 weeks after transplantation, when MSC-mediated neuroprotection might rather be a consequence of changes within the inflammatory milieu including paracrine secretion of local factors as has been described earlier for intravenous application routes. As such, the structural changes after intra-arterial MSC transplantation as observed at later time points might therefore only be one factor contributing to sustained neuroprotection, or this might be an epiphenomenon only.

Although intracerebral transplantation of BM-derived MSCs is of minor clinical relevance as compared with the intravenous application, local injections of MSCs might provide a higher number of cells, which is a prerequisite for further analysis of how MSCs induce postischemic neuroprotection. As has been described for other application routes, intracerebral transplantation of MSCs also resulted in improved functional recovery of recipient animals, with controversial results of MSC-mediated effects on infarct sizes.Citation63Citation65 Although neuronal differentiation of transplanted cells and stimulation of both proliferation and differentiation of SVZ-derived NPCs were observed, neuronal differentiation rates were low. Consequently, MSC-induced enhancement of endogenous neurogenesis is unlikely to significantly contribute to an enhanced poststroke recovery of recipient animals. On the other hand, MSC-mediated beneficial effects after intracerebral transplantation might rather be due to changes within the inflammatory ischemic tissue such as modulation of IL-10 expression.Citation64

Transplantation of non-BM-derived MSCs in experimental stroke models

Although BM-derived MSCs have been thoroughly characterized in vitro and used for experimental stroke research, data on the application of MSCs derived from sources other than BM are still scarce. Nevertheless, a significant number of studies analyzed the therapeutic potential of non-BM-derived MSCs originating from different sources, such as embryonic stem cells, blood, placenta, or adipose tissue.Citation66Citation69 As has been described for BM-derived cells, systemic application of these cells yielded significantly improved functional outcome after stroke in each experimental condition. The majority of studies, however, lack a systematic in vivo comparison between the cell type analyzed and BM-derived MSCs that is a “therapeutic gold standard”. Furthermore, the quality of the study is often limited to descriptive data implying two parameters: the extent of tissue injury and the extent of functional impairment. Therefore, the therapeutic potential of these MSCs as compared with BM-derived MSCs in experimental stroke research cannot be sufficiently assessed. As a consequence, further studies with emphasis on systematic comparison between different cell types in vivo and their modulating characteristics within the ischemic milieu are required.

Transplantation of MSCs in stroke patients

While considering the abovementioned beneficial effects of BM-derived MSCs on postischemic recovery in rodent models, only limited data are available on translational approaches into the clinic.Citation70,Citation71 In 2005, Bang et alCitation70 described a successful autologous transplantation of BM-derived MSCs in patients suffering from severe stroke. In that study, eligibility for enrolment in the study was defined by the National Institutes of Health Stroke Scale as at least 7.Citation70 The authors observed improved modified Rankin scores and higher Barthel indexes in patients who had systemically received MSCs during an observation period of 1 year. Although this is an interesting observation, some critical aspects have to be addressed. Out of 30 patients, only five patients received MSC therapy, leaving the treatment group small in comparison with the control group. Furthermore, study blindness was restricted to an initial observation period, ie, during the first week after stroke, which was followed by an allocation of patients to the experimental groups on day 7 after stroke. Treatment procedures themselves were, however, not blinded. MSC transplantation was late with a first injection between weeks 4 and 5 and an additional injection between weeks 7 and 9 after the onset of symptoms. Although autologous MSC transplantation requires time-consuming ex vivo cell culture expansion before the injection of cells, transplantation timing seems to be late, taking into account the reduced infarct sizes after early transplantation time points in rodent stroke models as described earlier. MSC-induced beneficial modulation in stroke treatment seems to decline with increasing cell passage numbers in rodents.Citation72 The aforementioned clinical trial was succeeded by a 5-year follow-up studyCitation71 due to several reasons, including safety concerns as to the use of fetal calf serum and fetal bovine serum in cell culture.Citation73 The authors observed sustained improved functional outcome in patients treated with MSCs for as long as 5 years with no significant side effects and no changes in mortality rates as compared with control patients. Beneficial outcome in patients treated with MSCs was associated with enhanced serum levels of SDF-1, which might be regarded as an epiphenomenon. Because MSCs induce endogenous neurogenesis in animal stroke models, the authors further studied an ischemic involvement of the lateral border of the SVZ and correlated the extent of injury of this region with the functional outcome of patients. Thus, the authors described a correlation between reduced ischemic injury of the lateral border of the SVZ and improved functional outcome of patients treated with MSCs. Whether the reduced involvement of the SVZ within the treatment group was due to genuine stimulation of endogenous neurogenesis by transplanted cells or it is due to MSC-mediated secretion of trophic factors followed by subsequent neuroprotection of residing cells remained unclear. Nevertheless, the promising results of systemic MSC transplantation in stroke patients warrants further preclinical studies aiming at increasing our understanding of how MSCs induce poststroke protection so that further clinical trials might benefit from these findings, resulting in improved study designs in the future.

Conclusion

There is no doubt that MSCs from different tissue sources enhance functional recovery in experimental stroke research models, even when applied at later time points regardless of application routes. There is also some evidence that early transplantation is required for the reduction of infarct size. The mechanisms underlying these observations are, however, not yet fully understood. Therefore, more sophisticated preclinical studies involving systematic analysis of the properties of MSCs from different tissues are needed. Special emphasis should also be put on ex vivo cell culture expansion and on how these procedures alter the biological properties of the cells to be transplanted. Depending on the outcome of these studies, clinical trials should not only be restricted to BM-derived MSCs but also be performed using MSCs from other sources such as blood, skin, or adipose tissue.

Disclosure

The authors declare that they have no conflict of interest.

References

  • HackeWDonnanGFieschiCAssociation of outcome with early stroke treatment: pooled analysis of ATLANTIS, ECASS, and NINDS rt-PA stroke trialsLancet200436376877415016487
  • BroderickJPHackeWTreatment of acute ischemic stroke: Part I: recanalization strategiesCirculation20021061563156912234965
  • BroderickJPHackeWTreatment of acute ischemic stroke. Part II: neuroprotection and medical managementCirculation20021061736174012270871
  • EvensonKRForakerREMorrisDLRosamondWDA comprehensive review of prehospital and in-hospital delay times in acute stroke careInt J Stroke2009418719919659821
  • DirnaglUIadecolaCMoskowitzMAPathobiology of ischaemic stroke: an integrated viewTrends Neurosci19992239139710441299
  • BroughtonBRReutensDCSobeyCGApoptotic mechanisms after cerebral ischemiaStroke200940e331e33919182083
  • Alonso de LecinanaMDiez-TejedorECarcellerFRodaJMCerebral ischemia: from animal studies to clinical practice. Should the methods be reviewed?Cerebrovasc Dis200111Suppl 1S20S30
  • Alonso de LecinanaMDiez-TejedorEGutierrezMGuerreroSCarcellerFRodaJMNew goals in ischemic stroke therapy: the experimental approach – harmonizing science with practiceCerebrovasc Dis200520Suppl 2S159S168
  • GladstoneDJBlackSEHakimAMHeart and Stroke Foundation of Ontario Centre of Excellence in Stroke RecoveryToward wisdom from failure: lessons from neuroprotective stroke trials and new therapeutic directionsStroke2002332123213612154275
  • SaeedSAShadKFSaleemTJavedFKhanMUSome new prospects in the understanding of the molecular basis of the pathogenesis of strokeExp Brain Res200718211017665180
  • MehtaSLManhasNRaghubirRMolecular targets in cerebral ischemia for developing novel therapeuticsBrain Res Rev200754346617222914
  • DirnaglUBench to bedside: the quest for quality in experimental stroke researchJ Cereb Blood Flow Metab2006261465147816525413
  • DelcroixGJSchillerPCBenoitJPMontero-MeneiCNAdult cell therapy for brain neuronal damages and the role of tissue engineeringBiomaterials2010312105212020005569
  • BurnsTCVerfaillieCMLowWCStem cells for ischemic brain injury: a critical reviewJ Comp Neurol200951512514419399885
  • HaasSWeidnerNWinklerJAdult stem cell therapy in strokeCurr Opin Neurol200518596415655404
  • HessDCBorlonganCVStem cells and neurological diseasesCell Prolif200841Suppl 1S94S114
  • BlumBBenvenistyNThe tumorigenicity of human embryonic stem cellsAdv Cancer Res200810013315818620095
  • FanYShenFFrenzelTEndothelial progenitor cell transplantation improves long-term stroke outcome in miceAnn Neurol20106748849720437584
  • SchwartingSLitwakSHaoWBahrMWeiseJNeumannHHematopoietic stem cells reduce postischemic inflammation and ameliorate ischemic brain injuryStroke2008392867287518658037
  • BacigaluppiMPluchinoSMartinoGKilicEHermannDMNeural stem/precursor cells for the treatment of ischemic strokeJ Neurol Sci2008265737717610905
  • BacigaluppiMPluchinoSPeruzzotti JamettiLDelayed post-ischaemic neuroprotection following systemic neural stem cell transplantation involves multiple mechanismsBrain20091322239225119617198
  • CaplanAIAdult mesenchymal stem cells for tissue engineering versus regenerative medicineJ Cell Physiol200721334134717620285
  • DexterTMSpooncerESchofieldRLordBISimmonsPHaemopoietic stem cells and the problem of self-renewalBlood Cells1984103153396543654
  • PittengerMFMackayAMBeckSCMultilineage potential of adult human mesenchymal stem cellsScience199928414314710102814
  • Sanchez-RamosJSongSCardozo-PelaezFAdult bone marrow stromal cells differentiate into neural cells in vitroExp Neurol200016424725610915564
  • TomaCPittengerMFCahillKSByrneBJKesslerPDHuman mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heartCirculation2002105939811772882
  • WengYSLinHYHsiangYJHsiehCTLiWTThe effects of different growth factors on human bone marrow stromal cells differentiating into hepatocyte-like cellsAdv Exp Med Biol200353411912812903715
  • KeatingAMesenchymal stromal cellsCurr Opin Hematol20061341942517053453
  • DominiciMLe BlancKMuellerIMinimal criteria for defining multipotent mesenchymal stromal cells. The international society for cellular therapy position statementCytotherapy2006831531716923606
  • BartholomewASturgeonCSiatskasMMesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivoExp Hematol200230424811823036
  • Le BlancKTammikCRosendahlKZetterbergERingdenOHLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cellsExp Hematol20033189089614550804
  • MaitraBSzekelyEGjiniKHuman mesenchymal stem cells support unrelated donor hematopoietic stem cells and suppress t-cell activationBone Marrow Transplant20043359760414716336
  • Le BlancKTammikLSundbergBHaynesworthSERingdénOMesenchymal stem cells inhibit and stimulate mixed lymphocyte cultures and mitogenic responses independently of the major histocompatibility complexScand J Immunol200357112012542793
  • EliopoulosNStaggJLejeuneLPommeySGalipeauJAllogeneic marrow stromal cells are immune rejected by MHC class I- and class II-mismatched recipient miceBlood20051064057406516118325
  • ChunmengSTianminCEffects of plastic-adherent dermal multipotent cells on peripheral blood leukocytes and CFU-GM in ratsTransplant Proc2004361578158115251389
  • in ’t AnkerPSNoortWAScherjonSAMesenchymal stem cells in human second-trimester bone marrow, liver, lung, and spleen exhibit a similar immunophenotype but a heterogeneous multilineage differentiation potentialHaematologica20038884585212935972
  • JacksonLJonesDRScottingPSottileVAdult mesenchymal stem cells: differentiation potential and therapeutic applicationsJ Postgrad Med20075312112717495381
  • ZukPAZhuMMizunoHMultilineage cells from human adipose tissue: implications for cell-based therapiesTissue Eng2001721122811304456
  • IguraKZhangXTakahashiKMitsuruAYamaguchiSTakashiTAIsolation and characterization of mesenchymal progenitor cells from chorionic villi of human placentaCytotherapy2004654355315770794
  • WagnerWWeinFSeckingerAComparative characteristics of mesenchymal stem cells from human bone marrow, adipose tissue, and umbilical cord bloodExp Hematol2005331402141616263424
  • ChenJLiYWangLTherapeutic benefit of intravenous administration of bone marrow stromal cells after cerebral ischemia in ratsStroke2001321005101111283404
  • HonmaTHonmouOIihoshiSIntravenous infusion of immortalized human mesenchymal stem cells protects against injury in a cerebral ischemia model in adult ratExp Neurol2006199566615967439
  • LiYChenJChenXGHuman marrow stromal cell therapy for stroke in rat: neurotrophins and functional recoveryNeurology20025951452312196642
  • OndaTHonmouOHaradaKHoukinKHamadaHKocsisJDTherapeutic benefits by human mesenchymal stem cells (hMSCs) and Ang-1 gene-modified hMSCs after cerebral ischemiaJ Cereb Blood Flow Metab20082832934017637706
  • PavlichenkoNSokolovaIVijdeSMesenchymal stem cells transplantation could be beneficial for treatment of experimental ischemic stroke in ratsBrain Res2008123320321318675258
  • ShenLHLiYChenJTherapeutic benefit of bone marrow stromal cells administered 1 month after strokeJ Cereb Blood Flow Metab20072761316596121
  • WakabayashiKNagaiASheikhAMTransplantation of human mesenchymal stem cells promotes functional improvement and increased expression of neurotrophic factors in a rat focal cerebral ischemia modelJ Neurosci Res2010881017102519885863
  • ZhengWHonmouOMiyataKTherapeutic benefits of human mesenchymal stem cells derived from bone marrow after global cerebral ischemiaBrain Res2010131081619913518
  • SongSKamathSMosqueraDExpression of brain natriuretic peptide by human bone marrow stromal cellsExp Neurol200418519119714697330
  • NakaoNItakuraTYokoteHNakaiKKomaiNEffect of atrial natriuretic peptide on ischemic brain edema: changes in brain water and electrolytesNeurosurgery19902739432143001
  • LiYChenJZhangCLGliosis and brain remodeling after treatment of stroke in rats with marrow stromal cellsGlia20054940741715540231
  • ChenJZhangZGLiYIntravenous administration of human bone marrow stromal cells induces angiogenesis in the ischemic boundary zone after stroke in ratsCirc Res20039269269912609969
  • ChenJLiYKatakowskiMIntravenous bone marrow stromal cell therapy reduces apoptosis and promotes endogenous cell proliferation after stroke in female ratJ Neurosci Res20037377878612949903
  • EsneaultEPacaryEEddiDCombined therapeutic strategy using erythropoietin and mesenchymal stem cells potentiates neurogenesis after transient focal cerebral ischemia in ratsJ Cereb Blood Flow Metab2008281552156318478023
  • EnglandTMartinPBathPMStem cells for enhancing recovery after stroke: a reviewInt J Stroke2009410111019383051
  • OmoriYHonmouOHaradaKSuzukiJHoukinKKocsisJDOptimization of a therapeutic protocol for intravenous injection of human mesenchymal stem cells after cerebral ischemia in adult ratsBrain Res20081236303818722359
  • KomatsuKHonmouOSuzukiJHoukinKHamadaHKocsisJDTherapeutic time window of mesenchymal stem cells derived from bone marrow after cerebral ischemiaBrain Res20101334849220382136
  • SchaarKLBrennemanMMSavitzSIFunctional assessments in the rodent stroke modelExp Transl Stroke Med201021320642841
  • ZacharekAShehadahAChenJComparison of bone marrow stromal cells derived from stroke and normal rats for stroke treatmentStroke20104152453020056925
  • ShenLHLiYChenJOne-year follow-up after bone marrow stromal cell treatment in middle-aged female rats with strokeStroke2007382150215617525391
  • ShenLHLiYChenJIntracarotid transplantation of bone marrow stromal cells increases axon-myelin remodeling after strokeNeuroscience200613739339916298076
  • LiYChenJWangLLuMChoppMTreatment of stroke in rat with intracarotid administration of marrow stromal cellsNeurology2001561666167211425931
  • LiYChoppMChenJIntrastriatal transplantation of bone marrow nonhematopoietic cells improves functional recovery after stroke in adult miceJ Cereb Blood Flow Metab2000201311131910994853
  • LiJZhuHLiuYHuman mesenchymal stem cell transplantation protects against cerebral ischemic injury and upregulates interleukin-10 expression in MacacafascicularisBrain Res20101334657220353760
  • YooSWKimSSLeeSYMesenchymal stem cells promote proliferation of endogenous neural stem cells and survival of newborn cells in a rat stroke modelExp Mol Med20084038739718779651
  • LeuSLinYCYuenCMAdipose-derived mesenchymal stem cells markedly attenuate brain infarct size and improve neurological function in ratsJ Transl Med201086320584315
  • UkaiRHonmouOHaradaKHoukinKHamadaHKocsisJDMesenchymal stem cells derived from peripheral blood protects against ischemiaJ Neurotrauma20072450852017402856
  • LiuYPSeckinHIzciYDuZWYanYPBaskayaMKNeuroprotective effects of mesenchymal stem cells derived from human embryonic stem cells in transient focal cerebral ischemia in ratsJ Cereb Blood Flow Metab20092978079119209181
  • KranzAWagnerDCKampradMTransplantation of placenta-derived mesenchymal stromal cells upon experimental stroke in ratsBrain Res2010131512813620004649
  • BangOYLeeJSLeePHLeeGAutologous mesenchymal stem cell transplantation in stroke patientsAnn Neurol20055787488215929052
  • LeeJSHongJMMoonGJLeePHAhnYHBangOYA long-term follow-up study of intravenous autologous mesenchymal stem cell transplantation in patients with ischemic strokeStem Cells2010281099110620506226
  • LiWYChoiYJLeePHMesenchymal stem cells for ischemic stroke: changes in effects after ex vivo culturingCell Transplant2008171045105919177841
  • SpeesJLGregoryCASinghHInternalized antigens must be removed to prepare hypoimmunogenic mesenchymal stem cells for cell and gene therapyMol Ther2004974775615120336