Skip to main content
Cell Transplantation logoLink to Cell Transplantation
. 2018 Sep 25;27(12):1825–1834. doi: 10.1177/0963689718795424

Transplanting Mesenchymal Stem Cells for Treatment of Ischemic Stroke

Fan Wang 1,2, Hailiang Tang 1, Jianhong Zhu 1,, John H Zhang 3,
PMCID: PMC6300770  PMID: 30251564

Abstract

Stroke is a major disease that leads to high mortality and morbidity. Given the ageing population and the potential risk factors, the prevalence of stroke and socioeconomic burden associated with stroke are expected to increase. During the past decade, both prophylactic and therapeutic strategies for stroke have made significant progress. However, current therapies still cannot adequately improve the outcomes of stroke and may not apply to all patients. One of the significant advances in modern medicine is cell-derived neurovascular regeneration and neuronal repair. Progress in stem cell biology has greatly contributed to ameliorating stroke-related brain injuries in preclinical studies and demonstrated clinical potential in stroke treatment. Mesenchymal stem cells (MSCs) have the differentiating potential of chondrocytes, adipocytes, and osteoblasts, and they have the ability to transdifferentiate into endothelial cells, glial cells, and neurons. Due to their great plasticity, MSCs have drawn much attention from the scientific community. This review will focus on MSCs, stem cells widely utilized in current medical research, and evaluate their effect and potential of improving outcomes in ischemic stroke.

Keywords: Ischemic stroke, stem cell replacement, MSCs

Introduction

Stroke is a major disease with high mortality and morbidity. Given the currently ageing population and the potential risk factors, the prevalence of and socioeconomic burden associated with stroke are expected to increase1. During the past decade, both prophylactic and therapeutic strategies of stroke have made significant progress. However, the current therapies still cannot adequately improve the outcomes of the disease and may not apply to all patients2. For instance, ischemic stroke accounts for about 80% of all stroke events. Intravenous thrombolysis with recombinant tissue plasminogen activator (rtPA) added within 4.5 hours is the only FDA-approved remedy for treating acute ischemic stroke3. However, with the narrow time window, this treatment can only be applied to 5% or less of patients with ischemic stroke. Even with an efficient thrombolytic therapy, only 55 cases out of 1000 can survive with good prognosis4. Furthermore, 6% of tPA-treated ischemic patients will go under symptomatic intracerebral hemorrhage. Therefore, new therapeutic strategies with a wider time window and less hemorrhagic risk are highly needed. Cell-based remedies are emerging as ideal candidates for functional recovery in stroke patients5. Mesenchymal stem cells (MSCs) are the most commonly utilized stem cell in biological medical research and therefore will be the focus of this review.

MSC Characteristics and Sources

In the late 1960s, Friedenstein et al. first discovered MSCs in the bone marrow stromal cells (BMSCs)6. Later MSCs were found to be capable of differentiating into mesenchymal cells, including adipocytes, cartilage producing chondrocytes as well as osteogenic osteoblasts7. Besides bone marrow, scientists have separated MSCs from many different types of tissues, such as Wharton’s jelly (WJ) in the umbilical cord stromal cells (UMSCs), umbilical-cord blood, adipose-derived stromal cells (ADSCs) as well as dental tissues811. Further studies on MSCs differentiation have shown that these cells can differentiate into hepatocytes12, cardiomyocytes13, and neuron-like cells14. MSCs have become a promising type of cell for stem cell-based therapies as they exist in all kinds of readily available donor tissues, such as the tissue of pulp and adipose. However, a major issue in the broad study of MSCs is that comparison between different study groups is difficult. The research team usually has its own method of separating, extending and describing cells, resulting in different standards in defining the MSCs810. To begin addressing this issue, the Mesenchymal and Tissue Stem Cell Committee of the International Society for Cellular Therapy proposes set the minimum standards for defining human MSCs. First, MSCs must be plastic-adhered under standard culture conditions. Second, MSCs must express CD105, CD73, and CD90, lacking the expression of CD45, CD34, CD14 or CD11b, CD79a or CD19 and HLA-DR surface molecules. Third, MSCs must differentiate into osteoblasts, adipocytes, and chondroblasts in vitro15. With the update of knowledge, the majority of MSCs do not express CD14 or CD11b, CD19 or CD79a, CD34, CD45, HLA-DR, while they express markers CD10, CD13, CD29, CD44, CD73, CD90, CD105, CD117, CD146, CD271, Stro-1 as well as stage-specific embryonic antigen-4 (SSEA-4)1619.

MSCs demonstrate a few properties that attract much research interest20. For example, they have the capability of differentiating into neurons, are easy to isolate and amplify from bone marrow, and have relatively low risk of immune rejection in allogeneic transplantation. There is much evidence from animal studies to show that MSC transplantation can reduce infarct volume, improve neurological function, and promote endogenous neurogenesis2123. In this review, we will mainly focus on the underlying mechanisms by which MSCs exert neuroprotective effects after ischemic stroke in preclinical animal models and summarize the current clinical trials using MSCs in ischemic stroke.

Mechanisms of Action of MSCs in Ischemic Stroke

Mechanisms of action of MSCs are divided into two levels: a peripheral level that involves reducing the inflammation and immunomodulation, as well as a central level, which is affected by angiogenesis, astrocytes, neurogenesis, axons and oligodendrocytes.

Immunomodulation and Post-Stroke Inflammation

Although it is well accepted that immune response is important in the pathogenesis of ischemic stroke, the current knowledge on immune response in focal cerebral ischemia is far from sufficient24. The immune system becomes active in response to neuronal damage in the event of focal cerebral ischemia or transient ischemia. After stroke, immediate activation of innate immunity triggers inflammation25. Inflammatory mediators recruit more immune cells both in the central nervous system and from the periphery. The production of more inflammatory mediators will further activate the adaptive immunity26. Inflammation, regression of inflammation, and repair of nerve damage are key successions after stroke. Although the inflammatory response is initially beneficial for limiting and resolving ischemic stress, an unrestricted inflammatory response by the continuous infiltration of immune cells such as neutrophils, macrophages, natural killer (NK) cells and T cells, can cause significant damage to the penumbra after cerebral ischemic injury27,28.

In vivo and in vitro examination showed that after studying hypoxic stroke neuronal cells and animal models of ischemic stroke, researchers found that MSCs reduced the expression of tumor necrosis factor-α and NF-kB through the vascular endothelial growth factor (VEGF) signaling as well as the interleukin-6 (IL-6) signaling29. The inhibition of NF-kB is associated with the anti-inflammatory and anti-apoptotic effects of MSCs30. Human umbilical cord blood MSCs remarkably attenuated the expression of IL-23 and IL-17 in infarcts and serum31, reduced the infarct size, and alleviated neurological deficits in the middle cerebral artery occlusion (MCAO) model. MSCs secrete transforming growth factor-β (TGF-β) by attenuating the upregulation of monocyte chemoattractant protein-1 (MCP-1) as well as the penetration of CD68+ immune cells via the compromised blood–brain barrier (BBB) to prevent the peripheral immune cells from exacerbating the inflammatory response in the ischemic rat brain32. In the ischemic stroke rat model, human MSCs reduced microglia activation, as indicated by lower expression of ED1 and Iba-1. They also attenuated astrogliosis, as indicated by lower GFAP level. These beneficial effects involved the noncanonical JAK-STAT signaling with unphosphorylated STAT3 in the immune cells33.

Impact on Astrocyte, Microglia, Oligodendrocyte, and Axon

In the event of cerebral ischemia, astrocytes release neurotrophic factors and growth factors upon the simulation of MSCs administration. These factors include insulin-like growth factor 1 (IGF-1), VEGF, epidermal growth factor (EGF), and basic fibroblast growth factor (bFGF)34. Astrocytic apoptosis is attenuated and ischemic-induced aquaporin-4 upregulation is normalized by MSCs, which contributes to maintaining BBB integrity after cerebral infarction35. MSCs reduce the TGFβ1 expressing in microglia/macrophages at the border of the ischemic area and promote down-regulation of the levels of plasminogen activator inhibitor 1 (PAI-1) in astroglia cells. After stroke, ingrown tPA and depressor PAI-1 are connected to neurite reconstruction36.

Oligodendrocytes play an important role in the restoration after ischemia37. The oligodendrocyte precursor cells (OPCs) appear in the corpus callosum, corpus striatum and subventricular zone (SVZ) of adult mouse brain, differentiating into mature oligodendroglia cells (OLs). OLs are very sensitive to the ischemic stress since the white matter blood flow is lower than that in gray matter; the blood supply for deep white matter is even less38. Myelin sheaths in the form of mature oligodendrocytes are used to sprout axons in ischemic tissues. The transplantation of bone marrow (BM)-MSCs increased the number of oligodendrocyte progenitor cells in the ischemic hemisphere as well as the number of mature oligodendrocytes surrounding the lesion39.

During the experimental stroke, BM-MSCs increased the axonal density in the surrounding area, which persisted for at least 1 year after stroke40. MSCs decrease the protein expression of reticulin (Rtn4 or Nogo) and induced neurocan (Ncan), an inhibitor of axonal growth40. According to Alder et al.41, umbilical cord tissue-derived cells (hUTC) and MSCs of human secret the brain-derived neurotrophic factor (BDNF), leading to increased amount and size of main dendrites.

Increased Neurogenesis

Ischemic stroke injury causes a dramatic increase in the proliferation of NSCs, triggering gliogenesis and neurogenesis in SVZ and circumventricular organs (CVOs). Down the third and fourth ventricles, some niches of new stem cell are detected in the context of stroke. It is important that all niches share a common feature – rich in vasculature with high permeability42. The incremental post-stroke neurogenesis was observed in elderly patients43. Hypoxic preconditioning of transplanted BMSCs could promote their regenerative capability for the treatment of ischemic stroke44. BDNF-modified hBM-MSCs (MSCs-BDNF) promoted endogenous neurogenesis and functional recovery in MCAO rat models45. Systemic administration of exosomes released from mesenchymal stromal cells promoted endogenous neurogenesis after stroke in rats46. It was found that the ultrasound promoted neurogenesis when the mouse stroke models were exposed to 0.04 MHz ultrasound after hBM-MSCs injection47.

Although post-stroke neurogenesis has been largely described, its role on restoration is still unknown48. The cellular therapy triggers the phosphatidylinositol-3-kinase (PI3 K)/Akt pathway in neural precursor cells, promoting cell survival, proliferation, and differentiation, as well as migration49. Upon the stimulation by BM-MSCs, brain parenchymal cells release neurotrophic factors, such as fibroblast growth factor and BDNF, to activate Akt/PI3 k pathway50. A study showed that transplanting hBMSCs into the ipsilateral brain parenchyma of MCAO rats could increase the expression of BDNF, neurotrophin-3 (NT-3) and VEGF in ischemic brain tissue, reduce infarct volume, and improve neurological function. Possibly, mechanisms for these beneficial effects were increased proliferation of neuronal progenitor cells in SVZ and in the subgranular zone (SGZ), accelerated migration of newborn neuroblasts to the ischemic border region (IBZ), diminished apoptosis, and increased differentiation of these cells into mature neurons51. Wharton’s jelly (WJ-MSC) induced better neurogenesis via a paracrine mechanism, WJ-MSC expressed more genes involved in angiogenesis and neurogenesis, especially secretory factors52.

Angiogenesis

Intravenous administration of BM-MSCs leads to releasing angiogenic growth factors as well as neurotrophic factors in time order, which includes angiogenin, hepatocyte growth factor (HGF), BDNF, fibroblast growth factor-2 (FGF-2), IGF-1, neutrophil activating protein 2 (NAP-2), and VEGF, to stimulate post-stroke neuronal growth and vascular formation53. These growth factors and neurotrophins all function in a paracrine or autocrine manner54, which can regulate the cell differentiation, proliferation, and survival. In the peri-infarct area, some researchers found that the expression of these factors has been elevated by BM-MSCs, including stromal cell-derived factor-1 (SDF-1), BDNF, platelet-derived growth factor-AA (PDGF-AA), basic fibroblast growth factor, angiopoietin-2, CXCL-16, neutrophil-activating protein-2, and vascular endothelial growth factor receptor-355. Furthermore, the expression of the axonal growth linked protein-43 (GAP-43) was also increased significantly in the brain tissues treated with BM-MSCs, while the axonal growth inhibitory protein ROCK II and NG2 were inhibited.

In the cerebral infarction region, the transplantation of BM-MSCs enhances the directed immigration and survival of neuroblasts as well56. The level of VEGF, phosphorylated ERK1/2 and RAF1 increases notably due to BM-MNC treatment, which also decreases the damage by white matter, stimulates angiogenesis, and facilitates a cognitive recovery in rats having bilateral common carotid arteries occlusion56.

Other studies have compared various sources of MSCs based on biologically active molecular secretion, the effects of angiogenesis and functional recovery after experimental stroke, as well as different routes of administration. In the rat stroke model, BM-MSCs through intravenous and intra-arterial delivery improved brain perfusion and metastasis by the assessment of SPECT and PET, particularly in rats treated with arterial perfusion delivery. No micro-strokes have been found after intra-arterial injection57. In another research, it was found that MSCs from adipose tissue (AD-MSCs) had the same effect as BM-MSCs in facilitating functional recovery, reducing necrosis and increasing neurogenesis, cell proliferation, and the markers of angiogenesis (e.g. VEGF) expression at 14 days after infarction in the model of rat stroke58. As to cellular MSCs-created exosomes post-stroke and traumatic brain injury, intravenous administration can improve functional recovery and enhance neurite reconstruction, angiogenesis as well as neurogenesis46,59,60.

To improve growth factors delivery, some groups used transgenic stem cells of mesenchyme to overexpress growth factors that are known to trigger the survival as well as the differentiation of neurons. According to Van Velthoven et al.61, the over-expressed BDNF on MSCs have an advantage over regular MSCs in the rat MCAO model in improving dyskinesia. Via intravenous or intracranial administration, Kurozumi et al62 used MSCs transgene with fiber-mutant adenovirus vector having BDNF or GDNF (glial cell derived neurotrophic factor). Both decreased the infarct volume by 6% to 40% with equal efficacy. With herpes simplex virus type 1 (HSV-1) vector transgenic MSCs carrying VEGF, Miki et al.63 found that it reduced infarct volume by 10% and improved functional deficits. Onda et al.64 used an adenoviral vector-modified MSCs having angiopoietin 1, which reduced the infarct size by 30% and alleviated motor deficits.

Novel Mechanisms of MSCs

Several novel mechanisms of MSCs have been studied, for example, mitochondrial or exosomal transfer from transplanted MSCs or the use of gene therapy of MSCs. Currently, there are few research studies on mitochondrial or exosomal transfer from transplanted MSCs in ischemic stroke.

It has been shown that mitochondrial transfer can rescue stressed cells65 and restore the loss of mitochondrial function in recipient cells66. Han et al.67 found BM-MSCs rescued injured H9c2 cells via transferring mitochondria through tunneling nanotubes in an in vitro simulated ischemia/reperfusion model. A similar study showed that MSCs rescued injured endothelial cells in an in vitro ischemia-reperfusion model via tunneling nanotube like structure-mediated mitochondrial transfer68. Exosomes are microvesicles released by cells ranging from 40 to 100 nanometers69. They are produced by the endocytosis of cell membranes and the subsequent formation of intracellular vesicles, which are released into the extracellular space by exocytosis69. They exist in any biological fluid such as urine, cerebrospinal fluid, or blood. They are surrounded by a lipid bilayer. They contain nucleic acids such as messenger RNA (mRNA) and microRNA (miRNA) and other different molecules70. Thus, they represent a new kind of intercellular communication mechanism. There is growing evidence that exosomes play an important role in cell-cell communication71. MSCs also release exosomes, and data shows that exosomes released by MSCs mediate communication between the MSCs and other cells72. Xin et al.60 suggested that exosomes from MSCs mediated the transfer of miR-133b to neurons and astrocytes that regulated gene expression, subsequently contributing to neurite remodeling and functional recovery following ischemic stroke. A review outlines the role of exosomes from MSCs in the recovery of ischemic stroke73.

MSC-based gene therapy represents a novel potential therapeutic strategy for ischemic stroke in future. The current strategy based on cell therapy emphasizes the introduction of beneficial genes, which will improve the therapeutic ability of MSCs and have better homing efficiency. It has a wide range of implications in stem cell biology. The methods of MSC gene delivery include physical methods, chemical methods, and the use of viral vectors. Several physical methods such as nuclear transfection74, electroporation75, nanoparticle76, and ultrasound transfection77 were used to deliver the beneficial genes into MSCs. In addition to physical methods of gene delivery, several chemotherapeutics mediated by cationic lipids78, calcium phosphates79, cationic polymers80, cationic polysaccharides81, and cationic peptides82 have been used for gene delivery. The main advantage of these non-virus-mediated (physical and chemical) gene delivery techniques is that they could be easily performed. However, the use of physical and chemical methods is limited due to low efficiency, not being suitable for transfection of large numbers of cells, and the use of chemical drugs possibly leading to higher concentrations of toxicity83. In addition, safety concerns have been considered, due to the non-degradable nature of certain polymers: for example, polyethylenimine (PEI) is a cationic non-degradable synthetic polymer, which is the most commonly used polymer for the development of nanocarriers for siRNA delivery, but at the expense of cytotoxicity, due to limited degradability84. Due to the limitations and disadvantages of the non-viral methods of gene delivery described above, some studies have used viral vectors to improve gene delivery. The viruses currently used as vectors are lentiviruses, adenoviruses, adeno-associated viruses, retroviruses, and baculoviruses85. Huang et al.86 found that lentiviral vector-mediated BDNF gene-modified MSCs could enhance its therapeutic effect in ischemic stroke. Zhao et al.87 introduced a novel strategy for combining transfer of MSCs and ex vivo HGF gene with multiple mutant herpes simplex virus type 1 vectors in a rat model of transient MCAO; the study showed that combination therapy was more effective than treatment with MSCs alone and might extend the treatment window from hyperacute to acute. At present, viral vector-based cytogenetic modification is widely used. However, low transduction efficiency and transgene potential limit its application in clinical trials.

Safety in Preclinical Studies

Although using MSCs in animal stroke models was generally safe and had a significant effect on behavioral outcomes88, some studies still showed side effects such as embolism89, infection, and tumor formation90. Amyloid-β accumulation and calcium in the thalamus also appear91. Research on rat stroke models suggested that intra-arterial (IA) MSCs delivery is capable of reducing the flow of middle cerebral artery (MCA); however, this side effect appears to be dose-dependent. A dosage of 1 x 105 MSCs was shown to be the maximal tolerable dose of IA infusion, making no concessions to the blood flow of MCA. One study also showed that delivering MSCs at 24 hours after stroke significantly improved neurological function and reduced the infarct size at 1 month compared with control but delivering 1 hour after stroke did not confer such protective effects92. Wang et al. found that there was no standard dose for stem cell therapy currently associated with the route of administration and disease types. For intracerebral parenchymal transplantation, an excessively large transplant dose affected the nutrition of transplanted cells and could cause microemboli and vascular occlusion when administered intravascularly93. Although there is no uniform dose standard, dose control is very important in preventing embolism. Intravenous infusion is thought to be associated with embolization, and embolization can be reduced by intraperitoneal or other routes of transplantation94.

Many safety problems have emerged with the intracerebral transplantation and interventional neuroradiography in acute stroke settings, such as maintain biological stability of the therapeutic product, larger MSCs doses can potentially affect organ perfusion, and the safety of allogeneic MSCs88. Another study showed that amyloid-β and calcium accumulation in the thalamus following intravenous injection of human bone marrow MSCs in MCAO model in rats, quantification of the area of the deposits showed a highly significant increase in amyloid-β and calcium deposition in the thalamus after infusion of MSCs at 48 hours after MCAO, there was a clear correlation between impaired forelimb performance on postoperative day 42 and amyloid-β and calcium accumulation in the thalamus91. MSC transplantation animal experiments found no obvious immune rejection. However, studies had shown that in vitro licensed WJ-MSCs did not improve experimental autoimmune encephalomyelitis in rats, due to increased immunogenicity resulting in rapid rejection95.

Clinical Trials of MSC Transplantation

Cells derived from bone marrow displayed great prospects for safety and initial efficacy96,97. Some clinical tests in Phase I and Phase II have already begun, using cell populations originated from mesenchymal stem cells (Table 1). Early results revealed that intravenous injection of MSCs does not give raise to significant adverse effects but can improve functional measurements such as the Barthel Index (BI)98, the National Institutes of Health Stroke Score (NIHSS) and the modified Rankin Scale (MRS)96. A long-term follow-up study of intravenous autologous mesenchymal stem cells transplantation in patients with ischemic stroke showed that no significant side effects were observed, and the follow-up MRS score was decreased compared with the control group99. A meta-analysis from Lalu et al.100 suggested that MSC therapy appeared safe, but there was a significant association between MSC and transient fever based on the current clinical trials, so further larger scale controlled clinical trials with rigorous reporting of adverse events were required to further define the safety profile of MSCs. Contradictory data shows that MSC injection may not improve the results of the function101. There was no significant difference in the BI score, MRS shift analysis, NIHSS score, or change in infarct volume at day 180 compared with the control group101. These studies used autologous MSCs which were expanded in culture before MSC transplantation96101. Although no side effects of the products were reported, the cells were amplified in autologous serum, leading to faster cell expansion and reducing concern of heterogeneous contamination.

Table 1.

Mesenchymal Stem Cells Transplantation in Ischemic Stroke Clinical Trials.

NCT Country Phase Cell Source/
Autologous or allogenicr
Doses/Single(S)
or multiple(M)
Route Time from stroke onset Sample cases Current status
00875654 France II MSCs/Autologous ND/ND IV <6 weeks 30 Completed
01091701 Malaysia I/II MSCs/Allogenicr 2 × 106/S IV <10 days ND Withdrawn
01297413 USA I/II BMSCs/Allogenicr 0.5–1.5 × 106/S IV >24 weeks 38 Not yet recruiting
01389453 China II UMSCs/Allogenicr ND/M IV/LP 1–2 weeks 100 Withdrawn
01461720 Malaysia II BMSCs/Autologous ND/ND IV 2–8 weeks 50 Unknown
01468064 China I/II BMSCs/Autologous 2.5 × 106/M IV <1 week 20 Recruiting
01678534 Spain II ADSCs/Allogenicr 1 × 106/S IV <2 weeks 20 Recruiting
01714167 China I BMSCs/Autologous 2–4 × 106/ND IC 3–60 months 30 Unknown
01716481 Korea III MSCs/Autologous ND/ND IV <90 days 60 Recruiting
01849887 USA I BMSCs/Allogenicr ND/ND IV ND ND Withdrawn
01922908 USA I/II BMSCs/Allogenicr ND/S IV 3–10 days 48 Not yet recruiting
02378974 Korea I/II UMSCs/Allogenicr 2 × 108/M IV <1 week 18 Not yet recruiting
02425670 India II BMSCs/Autologous 3–50 × 107/ND IV 7–30 days 120 completed
02564328 China I BMSCs/Autologous ND/ND IV 6–60 months 40 Recruiting
02580019 China II UMSCs/Allogenicr 2 × 107/S IV <12 weeks 2 Not yet recruiting
02849613 Europe II/III ADSCs/Allogenicr ND/ND IV 1–4 days 400 Recruiting
03176498 China I/II UMSCs/Allogenicr ND/M IV ND 40 Not yet recruiting
03186456 China I UMSCs/Allogenicr 0.5–1 × 106/M IV <2 weeks 40 Not yet recruiting
03356821 Netherlands I/II BMSCs/Allogenicr 5 × 107/S Nasal <7 days 10 Not yet recruiting

From information available at ClinicalTrials.gov, searched by ‘MSCs’ and ‘ischemic stroke’. Table updated June 11, 2018.

IC: Intracerebral injection; IV: intravenous; LP: lumbar puncture; NCT: ClinicalTrials.gov identifier; ND: no data.

Conclusions and Future Prospects

There are many advantages of MSCs: they are easy to harvest, amplify and store for a long time; they can be quickly isolated with relative immune privileges; they can be managed in various manners; and their usage does not result in many ethical issues. However, so far, only the clinical trials of phase I and II have been reported, covering a small number of participants and a comparatively short duration of follow-up, while it stills lacks the larger-scale phase III clinical trials. Therefore, further research is needed to address the long-range safety and effect of therapy with MSCs102.

Acknowledgments

The study was supported by grant (2018YFA0107901) from the National Key Research and Development Plan, and grants (81000488, 81271003) from the National Natural Science Foundation of China.

Authors' Note: Wang and Tang contribute equally to this paper.

Declaration of Conflicting Interests: The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The authors received no financial support for the research, authorship, and/or publication of this article.

ORCID iD: Hailiang Tang Inline graphic http://orcid.org/0000-0003-1852-3128

References

  • 1. Feigin VL, Forouzanfar MH, Krishnamurthi R, Mensah GA, Connor M, Bennett DA, Moran AE, Sacco RL, Anderson L, Truelsen T, O’Donnell M, Venketasubramanian N, Barker-Collo S, Lawes CM, Wang W, Shinohara Y, Witt E, Ezzati M, Naghavi M, Murray C; Global Burden of Diseases, Injuries, and Risk Factors Study 2010 (GBD 2010) and the GBD Stroke Experts Group. Global and regional burden of stroke during 1990–2010: findings from the global burden of disease study 2010. Lancet. 2014;383(9913):245–254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Krishnamurthi RV, Feigin VL, Forouzanfar MH, Mensah GA, Connor M, Bennett DA, Moran AE, Sacco RL, Anderson LM, Truelsen T, O’Donnell M, Venketasubramanian N, Barker-Collo S, Lawes CM, Wang W, Shinohara Y, Witt E, Ezzati M, Naghavi M, Murray C; Global Burden of Diseases, Injuries, Risk Factors Study 2010 (GBD 2010); GBD Stroke Experts Group. Global and regional burden of first-ever ischaemic and haemorrhagic stroke during 1990–2010: findings from the Global Burden of Disease Study 2010. Lancet Glob Health. 2013;1(5):e259–e281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Lapchak PA, Zhang JH. The high cost of stroke and stroke cytoprotection research. Transl Stroke Res. 2017;8(4):307–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Wardlaw JM, Murray V, Berge E, del Zoppo G, Sandercock P, Lindley RL, Cohen G. Recombinant tissue plasminogen activator for acute ischaemic stroke: An updated systematic review and meta-analysis. Lancet. 2012;379(9834):2364–2372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Boltze J, Ayata C. Challenges and controversies in translational stroke research – an introduction. Transl Stroke Res. 2016;7(5):355–357. [DOI] [PubMed] [Google Scholar]
  • 6. Friedenstein AJ, Chailakhjan RK, Lalykina KS. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinet. 1970;3(4):393–403. [DOI] [PubMed] [Google Scholar]
  • 7. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284(5411):143–147. [DOI] [PubMed] [Google Scholar]
  • 8. Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P, Hedrick MH. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13(12):4279–4295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Gronthos S, Mankani M, Brahim J, Robey PG, Shi S. Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc Natl Acad Sci USA. 2000;97(25):13625–13630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Weiss ML, Medicetty S, Bledsoe AR, Rachakatla RS, Choi M, Merchav S, Luo Y, Rao MS, Velagaleti G, Troyer D. Human umbilical cord matrix stem cells: Preliminary characterization and effect of transplantation in a rodent model of Parkinson’s disease. Stem Cells. 2006;24(3):781–792. [DOI] [PubMed] [Google Scholar]
  • 11. Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol. 2000;109(1):235–242. [DOI] [PubMed] [Google Scholar]
  • 12. Lee KD, Kuo TK, Whang PJ, Chung YF, Lin CT, Chou SH, Chen JR, Chen YP, Lee OK. In vitro hepatic differentiation of human mesenchymal stem cells. Hepatology. 2004;40(6):1275–1284. [DOI] [PubMed] [Google Scholar]
  • 13. Toma C, Pittenger MF, Cahill KS, Byrne BJ, Kessler PD. Human mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart. Circulation. 2002;105(1):93–98. [DOI] [PubMed] [Google Scholar]
  • 14. Wislet-Gendebien S, Hans G, Leprince P, Rigo JM, Moonen G, Rogister B. Plasticity of cultured mesenchymal stem cells: Switch from nestin-positive to excitable neuron-like phenotype. Stem Cells. 2005;23(3):392–402. [DOI] [PubMed] [Google Scholar]
  • 15. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop DJ, Horwitz E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8(4):315–317. [DOI] [PubMed] [Google Scholar]
  • 16. Kaiser S, Hackanson B, Follo M, Mehlhorn A, Geiger K, Ihorst G, Kapp U. BM cells giving rise to MSC in culture have a heterogeneous CD34 and CD45 phenotype. Cytotherapy. 2007;9(5):439–450. [DOI] [PubMed] [Google Scholar]
  • 17. Kaveh B, Seyed MH, Samaneh T, Asadi Rad A, Assadzadeh-Aghdaei H, Sharifian A, Zali MR. Isolation, differentiation, and characterization of mesenchymal stem cells from human bone marrow. Gastroenterol Hepatol Bed Bench. 2017;10(3):208–213. [PMC free article] [PubMed] [Google Scholar]
  • 18. Lv FJ, Tuan RS, Cheung KM, Leung VY. Concise review: The surface markers and identity of human mesenchymal stem cells. Stem cells. 2014;32(6):1408–1419. [DOI] [PubMed] [Google Scholar]
  • 19. Ludovic Z, Donnenberg VS, Rubin JP, Donnenberg AD. Mesenchymal markers on human adipose stem/progenitor cells. Cytometry A. 2013;83(1):134–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Hsuan YC, Lin CH, Chang CP, Lin MT. Mesenchymal stem cell-based treatments for stroke, neural trauma, and heat stroke. Brain Behav. 2016;6(10):e00526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Li Y, Chen J, Chen XG, Wang L, Gautam SC, Xu YX, Katakowski M, Zhang LJ, Lu M, Janakiraman N, Chopp M. Human marrow stromal cell therapy for stroke in rat: neurotrophins and functional recovery. Neurology. 2002;59(4):514–523. [DOI] [PubMed] [Google Scholar]
  • 22. Liu H, Honmou O, Harada K, Nakamura K, Houkin K, Hamada H, Kocsis JD. Neuroprotection by PIGF gene-modified human mesenchymal stem cells after cerebral ischaemia. Brain. 2006;129(Pt10):2734–2745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Hao L, Zou Z, Tian H, Zhang Y, Zhou H, Liu L. Stem cell-based therapies for ischemic stroke. Biomed Res Int. 2014;2014:468748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Garcia-Bonilla L, Benakis C, Moore J, Iadecola C, Anrather J. Immune mechanisms in cerebral ischemic tolerance. Front Neurosci. 2014;8:44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Shi Y, Leak RK, Keep RF, Chen J. Translational stroke research on blood-brain barrier damage: Challenges, perspectives, and goals. Transl Stroke Res. 2016;7(2):89–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Xia Y, Cai W, Thomson AW, Hu X. Regulatory T cell therapy for ischemic stroke: How far from clinical translation? Transl Stroke Res. 2016;7(5):415–419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Petrovic-Djergovic D, Goonewardena SN, Pinsky DJ. Inflammatory disequilibrium in stroke. Circ Res. 2016;119(1):142–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Takashi S, Minako I, Akihiko Y. Post-ischemic inflammation regulates neural damage and protection. Front Cell Neurosci. 2014;8:319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Huang P, Gebhart N, Richelson E, Brott TG, Meschia JF, Zubair AC. Mechanism of mesenchymal stem cell-induced neuron recovery and antiinflammation. Cytotherapy. 2014;16(10):1336–1344. [DOI] [PubMed] [Google Scholar]
  • 30. Gu N, Rao C, Tian Y, Di Z, Liu Z, Chang M, Lei H. Anti-inflammatory and antiapoptotic effects of mesenchymal stem cells transplantation in rat brain with cerebral ischemia. J Stroke Cerebrovasc Dis. 2014;23(10):2598–2606. [DOI] [PubMed] [Google Scholar]
  • 31. Ma S, Zhong D, Chen H, Zheng Y, Sun Y, Luo J, Li H, Li G, Yin Y. The immunomodulatory effect of bone marrow stromal cells (BMSCs) on interleukin (IL)-23/IL-17-mediated ischemic stroke in mice. J Neuroimmunol. 2013;257(1–2):28–35. [DOI] [PubMed] [Google Scholar]
  • 32. Yoo SW, Chang DY, Lee HS, Kim GH, Park JS, Ryu BY, Joe EH, Lee YD, Kim SS, Suh-Kim H. Immune following suppression mesenchymal stem cell transplantation in the ischemic brain is mediated by TGF- β. Neurobiol Dis. 2013;58:249–257. [DOI] [PubMed] [Google Scholar]
  • 33. McGuckin CP, Jurga M, Miller AM, Sarnowska A, Wiedner M, Boyle NT, Lynch MA, Jablonska A, Drela K, Lukomska B, Domanska-Janik K, Kenner L, Moriggl R, Degoul O, Perruisseau-Carrier C, Forraz N. Ischemic brain injury: A consortium analysis of key factors involved in mesenchymal stem cell-mediated inflammatory reduction. Arch Biochem Biophys. 2013;534(1–2):88–97. [DOI] [PubMed] [Google Scholar]
  • 34. Zhu J, Liu Q, Jiang Y, Wu L, Xu G, Liu X. Enhanced angiogenesis promoted by human umbilical mesenchymal stem cell transplantation in stroked mouse is Notch1 signaling associated. Neuroscience. 2015;290:288–299. [DOI] [PubMed] [Google Scholar]
  • 35. Tang G, Liu Y, Zhang Z, Lu Y, Wang Y, Huang J, Li Y, Chen X, Gu X, Wang Y, Yang GY. Mesenchymal stem cells maintain blood-brain barrier integrity by inhibiting aquaporin-4 upregulation after cerebral ischemia. Stem Cells. 2014;32(12):3150–3162. [DOI] [PubMed] [Google Scholar]
  • 36. Xin H, Chopp M, Shen LH, Zhang RL, Zhang L, Zhang ZG, Li Y. Multipotent mesenchymal stromal cells decrease transforming growth factor β1 expression in microglia/macrophages and down-regulate plasminogen activator inhibitor 1 expression in astrocytes after stroke. Neurosci Lett. 2013;542:81–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Chen J, Zuo S, Wang J, Huang J, Zhang X, Liu Y, Zhang Y, Zhao J, Han J, Xiong L, Shi M, Liu Z. Aspirin promotes oligodendrocyte precursor cell proliferation and differentiation after white matter lesion. Front Aging Neurosci. 2014;6:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Pantoni L, Garcia JH, Gutierrez JA. Cerebral white matter is highly vulnerable to ischemia. Stroke. 1996;27(9):1641–1647. [DOI] [PubMed] [Google Scholar]
  • 39. Li Y, Chen J, Zhang CL, Wang L, Lu D, Katakowski M, Gao Q, Shen LH, Zhang J, Lu M, Chopp M. Gliosis and brain remodeling after treatment of stroke in rats with marrow stromal cells. Glia. 2005;49(3):407–417. [DOI] [PubMed] [Google Scholar]
  • 40. Shen LH, Li Y, Gao Q, Savant-Bhonsale S, Chopp M. Down-regulation of neurocan expression in reactive astrocytes promotes axonal regeneration and facilitates the neurorestorative effects of bone marrow stromal cells in the ischemic rat brain. Glia. 2008;56(16):1747–1754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Alder J, Kramer BC, Hoskin C, Thakker-Varia S. Brain-derived neurotrophic factor produced by human umbilical tissue-derived cells is required for its effect on hippocampal dendritic differentiation. Dev Neurobiol. 2012;72(6):755–765. [DOI] [PubMed] [Google Scholar]
  • 42. Lin R, Cai J, Nathan C, Wei X, Schleidt S, Rosenwasser R, Iacovitti L. Neurogenesis is enhanced by stroke in multiple new stem cell niches along the ventricular system at sites of high BBB permeability. Neurobiol Dis. 2015;74:229–239. [DOI] [PubMed] [Google Scholar]
  • 43. Thored P, Arvidsson A, Cacci E, Ahlenius H, Kallur T, Darsalia V, Ekdahl CT, Kokaia Z, Lindvall O. Persistent production of neurons from adult brain stem cells during recovery after stroke. Stem Cells. 2006;24(3):739–747. [DOI] [PubMed] [Google Scholar]
  • 44. Ling W, Jamie LF, Lu ZY, Hu X, Yu SP. Transplantation of hypoxia preconditioned bone marrow mesenchymal stem cells enhances angiogenesis and neurogenesis after cerebral ischemia in rats. Neurobiol Dis. 2012;46(3):635–645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Jeong CH, Kim SM, Lim JY, Ryu CH, Jun JA, Jeun SS. Mesenchymal stem cells expressing brain-derived neurotrophic factor enhance endogenous neurogenesis in an ischemic stroke model. Biomed Res Int. 2014;2014:129145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Xin H, Li Y, Cui Y, Yang JJ, Zhang ZG, Chopp M. Systemic administration of exosomes released from mesenchymal stromal cells promote functional recovery and neurovascular plasticity after stroke in rats. J Cereb Blood Flow Metab. 2013;33(11):1711–1715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Cho SE, Kim YM, Jeong JS, Seo YK. The effect of ultrasound for increasing neural differentiation in hBM-MSCs and inducing neurogenesis in ischemic stroke model. Life Sci. 2016;165:35–42. [DOI] [PubMed] [Google Scholar]
  • 48. Butti E, Bacigaluppi M, Rossi S, Cambiaghi M, Bari M, Cebrian Silla A, Brambilla E, Musella A, De Ceglia R, Teneud L, De Chiara V, D’Adamo P, Garcia-Verdugo JM, Comi G, Muzio L, Quattrini A, Leocani L, Maccarrone M, Centonze D, Martino G. Subventricular zone neural progenitors protect striatal neurons from glutamatergic excitotoxicity. Brain. 2012;135(Pt 11):3320–3335. [DOI] [PubMed] [Google Scholar]
  • 49. Chen J, Crawford R, Chen C, Xiao Y. The key regulatory roles of the PI3K/Akt signaling pathway in the functionalities of mesenchymal stem cells and applications in tissue regeneration. Tissue Eng Part B Rev. 2013;19(6):516–528. [DOI] [PubMed] [Google Scholar]
  • 50. Tsai MJ, Tsai SK, Hu BR, Liou DY, Huang SL, Huang MC, Huang WC, Cheng H, Huang SS. Recovery of neurological function of ischemic stroke by application of conditioned medium of bone marrow mesenchymal stem cells derived from normal and cerebral ischemia rats. J Biomed Sci. 2014;21:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Bao X, Wei J, Feng M, Lu S, Li G, Dou W, Ma W, Ma S, An Y, Qin C, Zhao RC, Wang R. Transplantation of human bone marrow-derived mesenchymal stem cells promotes behavioral recovery and endogenous neurogenesis after cerebral ischemia in rats. Brain research. 2011;1367:103–113. [DOI] [PubMed] [Google Scholar]
  • 52. Hsieh JY, Wang HW, Chang SJ, Liao KH, Lee IH, Lin WS, Wu CH, Lin WY, Cheng SM. Mesenchymal stem cells from human umbilical cord express preferentially secreted factors related to neuroprotection, neurogenesis, and angiogenesis. PLoS One. 2013;8(8):e72604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Bronckaers A, Hilkens P, Martens W, Gervois P, Ratajczak J, Struys T, Lambrichts I. Mesenchymal stem/stromal cells as a pharmacological and therapeutic approach to accelerate angiogenesis. Pharmacol Ther. 2014;143(2):181–196. [DOI] [PubMed] [Google Scholar]
  • 54. Matsuda-Hashii Y, Takai K, Ohta H, Fujisaki H, Tokimasa S, Osugi Y, Ozono K, Matsumoto K, Nakamura T, Hara J. Hepatocyte growth factor plays roles in the induction and autocrine maintenance of bone marrow stromal cell IL-11, SDF-1 alpha, and stem cell factor. Exp Hematol. 2004;32(10):955–961. [DOI] [PubMed] [Google Scholar]
  • 55. Lin YC, Ko TL, Shih YH, Lin MY, Fu TW, Hsiao HS, Hsu JY, Fu YS. Human umbilical mesenchymal stem cells promote recovery after ischemic stroke. Stroke. 2011;42(7):2045–20s53. [DOI] [PubMed] [Google Scholar]
  • 56. Wang J, Fu X, Jiang C, Yu L, Wang M, Han W, Liu L, Wang J. Bone marrow mononuclear cell transplantation promotes therapeutic angiogenesis via upregulation of the VEGF-VEGFR2 signaling pathway in a rat model of vascular dementia. Behav Brain Res. 2014; 265:171–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Du S, Guan J, Mao G, Liu Y, Ma S, Bao X, Gao J, Feng M, Li G, Ma W, Yang Y, Zhao RC, Wang R. Intra-arterial delivery of human bone marrow mesenchymal stem cells is a safe and effective way to treat cerebral ischemia in rats. Cell Transplant. 2014;23(Suppl 1):s73–s82. [DOI] [PubMed] [Google Scholar]
  • 58. Gutiérrez-Fernández M, Rodríguez-Frutos B, Ramos-Cejudo J, Teresa Vallejo-Cremades M, Fuentes B, Cerdán S, Díez-Tejedor E. Effects of intravenous administration of allogenic bone marrow- and adipose tissue-derived mesenchymal stem cells on functional recovery and brain repair markers in experimental ischemic stroke. Stem Cell Res Ther. 2013;4(1):11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Zhang YL, Michael C, Meng YL, Katakowski M, Xin H, Mahmood A, Xiong Y. Effect of exosomes derived from multipluripotent mesenchymal stromal cells on functional recovery and neurovascular plasticity in rats after traumatic brain injury. J Neurosurg. 2015;122(4):856–867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Xin H, Li Y, Liu Z, Wang X, Shang X, Cui Y, Zhang ZG, Chopp M. MiR-133b promotes neural plasticity and functional recovery after treatment of stroke with multipotent mesenchymal stromal cells in rats via transfer of exosome-enriched extracellular particles. Stem Cells. 2013;31(12):2737–2746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Van Velthoven CT, Sheldon RA, Kavelaars A, Derugin N, Vexler ZS, Willemen HL, Maas M, Heijnen CJ, Ferriero DM. Mesenchymal stem cell transplantation attenuates brain injury after neonatal stroke. Stroke. 2013;44(5):1426–1432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Kurozumi K, Nakamura K, Tamiya T, Kawano Y, Ishii K, Kobune M, Hirai S, Uchida H, Sasaki K, Ito Y, Kato K, Honmou O, Houkin K, Date I, Hamada H. Mesenchymal stem cells that produce neurotrophic factors reduce ischemic damage in the rat middle cerebral artery occlusion model. Mol Ther. 2005;11(1):96–104. [DOI] [PubMed] [Google Scholar]
  • 63. Miki Y, Nonoguchi N, Ikeda N, Coffin RS, Kuroiwa T, Miyatake S. Vascular endothelial growth factor gene-transferred bone marrow stromal cells engineered with a herpes simplex virus type 1 vector can improve neurological deficits and reduce infarction volume in rat brain ischemia. Neurosurgery. 2007;61(3):586–594. [DOI] [PubMed] [Google Scholar]
  • 64. Onda T, Honmou O, Harada K, Houkin K, Hamada H, Kocsis JD. Therapeutic benefits by human mesenchymal stem cells (hMSCs) and Ang-1 gene-modified hMSCs after cerebral ischemia. J Cereb Blood Flow Metab. 2008;28(2):329–340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Islam MN, Das SR, Emin MT, Wei M, Sun L, Westphalen K, Rowlands DJ, Quadri SK, Bhattacharya S, Bhattacharya J. Mitochondrial transfer from bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat Med. 2012;18(5):759–765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Li X, Zhang Y, Yeung SC, Liang Y, Liang X, Ding Y, Ip MS, Tse HF, Mak JC, Lian Q. Mitochondrial transfer of induced pluripotent stem cell-derived mesenchymal stem cells to airway epithelial cells attenuates cigarette smoke-induced damage. Am J Respir Cell Mol Biol. 2014;51(3):455–465. [DOI] [PubMed] [Google Scholar]
  • 67. Han H, Hu J, Yan Q, Zhu J, Zhu Z, Chen Y, Sun J, Zhang R. Bone marrow-derived mesenchymal stem cells rescue injured H9c2 cells via transferring intact mitochondria through tunneling nanotubes in an in vitro simulated ischemia/reperfusion model. Mol Med Rep. 2016;13(2):1517–1524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Liu K, Ji K, Guo L, Wu W, Lu H, Shan P, Yan C. Mesenchymal stem cells rescue injured endothelial cells in an in vitro ischemia-reperfusion model via tunneling nanotube like structure-mediated mitochondrial transfer. Microvasc Res. 2014;92:10–18. [DOI] [PubMed] [Google Scholar]
  • 69. Koga K, Matsumoto K, Akiyoshi T, Kubo M, Yamanaka N, Tasaki A, Nakashima H, Nakamura M, Kuroki S, Tanaka M, Katano M. Purification, characterization and biological significance of tumor-derived exosomes. Anticancer Res. 2005; 25(6):3703–3707. [PubMed] [Google Scholar]
  • 70. O’Driscoll L. Expanding on exosomes and ectosomes in cancer. N Engl J Med. 2015;372(24):2359–2362. [DOI] [PubMed] [Google Scholar]
  • 71. Record M, Subra C, Silvente-Poirot S, Poirot M. Exosomes as intercellular signalosomes and pharmacological effectors. Biochem Pharmacol. 2011;81(10):1171–1182. [DOI] [PubMed] [Google Scholar]
  • 72. Xin H, Li Y, Buller B, Katakowski M, Zhang Y, Wang X, Shang X, Zhang ZG, Chopp M. Exosome-mediated transfer of miR-133b from multipotent mesenchymal stromal cells to neural cells contributes to neurite outgrowth. Stem Cells. 2012;30(7):1556–1564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Xin H, Li Y, Chopp M. Exosomes/miRNAs as mediating cell-based therapy of stroke. Front Cell Neurosci. 2014;8:377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Gresch O, Engel F B, Nesic D, Tran TT, England HM, Hickman ES, Körner I, Gan L, Chen S, Castro-Obregon S, Hammermann R, Wolf J, Müller-Hartmann H, Nix M, Siebenkotten G, Kraus G, Lun K. New non-viral method for gene transfer into primary cells. Methods. 2004;33(2):151–163. [DOI] [PubMed] [Google Scholar]
  • 75. Liew A, André FM, Lesueur LL, De Ménorval MA, O’Brien T, Mir LM. Robust, efficient, and practical electrogene transfer method for human mesenchymal stem cells using square electric pulses. Hum Gene Ther Methods. 2013;24(5):289–297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Muroski ME, Morgan TJ, Jr, Levenson CW, Strouse GF. A gold nanoparticle pentapeptide: Gene fusion to induce therapeutic gene expression in mesenchymal stem cells. J Am Chem Soc. 2014;136(42):14763–14771. [DOI] [PubMed] [Google Scholar]
  • 77. Haber T, Baruch L, Machluf M. Ultrasound-mediated mesenchymal stem cells transfection as a targeted cancer therapy platform. Sci Rep. 2017;7:42046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Locatelli P, Olea FD, Hnatiuk A, Sepúlveda D, Pérez Sáez JM, Argüello R, Crottogini A. Efficient plasmid-mediated gene transfection of ovine bone marrow mesenchymal stromal cells. Cytotherapy. 2013;15(2):163–170. [DOI] [PubMed] [Google Scholar]
  • 79. Cao X, Deng W, Wei Y, Su W, Yang Y, Wei Y, Yu J, Xu X. Encapsulation of plasmid DNA in calcium phosphate nanoparticles: Stem cell uptake and gene transfer efficiency. Int J Nanomedicine. 2011;6:3335–3349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Wu C, Li J, Pang P, Liu J, Zhu K, Li D, Cheng D, Chen J, Shuai X, Shan H. Polymeric vector-mediated gene transfection of MSCs for dual bioluminescent and MRI tracking in vivo. Biomaterials. 2014;35(28):8249–8260. [DOI] [PubMed] [Google Scholar]
  • 81. Thakor DK, Teng YD, Obata H, Nagane K, Saito S, Tabata Y. Nontoxic genetic engineering of mesenchymal stem cells using serum-compatible pullulan-spermine/DNA anioplexes. Tissue Eng Part C Methods. 2011;17(2):131–144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Shan CL, Huang B, You J, Yuan H, Gao JQ, Hu FQ, Du YZ. High efficiency intracellular transport of cationic peptide stearate for gene delivery in tumor cells and multipotent stem cells. J Biomed Nanotechnol. 2014;10(11):3231–3243. [DOI] [PubMed] [Google Scholar]
  • 83. Santos JL, Pandita D, Rodrigues J, Pêgo AP, Granja PL, Tomás H. Non-viral gene delivery to mesenchymal stem cells: Methods, strategies and application in bone tissue engineering and regeneration. Curr Gene Ther. 2011;11(1):46–57. [DOI] [PubMed] [Google Scholar]
  • 84. Wang W, Xu X, Li Z, Lendlein A, Ma N. Genetic engineering of mesenchymal stem cells by non-viral gene delivery. Clin Hemorheol Microcirc. 2014;58(1):19–48. [DOI] [PubMed] [Google Scholar]
  • 85. Nowakowski A, Andrzejewska A, Janowski M, Walczak P, Lukomska B. Genetic engineering of stem cells for enhanced therapy. Acta Neurobiol Exp. 2013;73(1):1–18. [DOI] [PubMed] [Google Scholar]
  • 86. Huang D, Zhang Z, Chen B, Wu X, Wang N, Zhang Y. Therapeutic efficacy of lentiviral vector mediated BDNF gene-modified MSCs in cerebral infarction. Sheng Wu Gong Cheng Xue Bao. 2008;24(7):1174–1179. [PubMed] [Google Scholar]
  • 87. Zhao MZ, Nonoguchi N, Ikeda N, Watanabe T, Furutama D, Miyazawa D, Funakoshi H, Kajimoto Y, Nakamura T, Dezawa M, Shibata MA, Otsuki Y, Coffin RS, Liu WD, Kuroiwa T, Miyatake S. Novel therapeutic strategy for stroke in rats by bone marrow stromal cells and ex vivo HGF gene transfer with HSV-1 vector. J Cereb Blood Flow Metab. 2006;26(9):1176–1188. [DOI] [PubMed] [Google Scholar]
  • 88. Vu Q, Xie K, Eckert M, Zhao W, Cramer SC. Meta-analysis of preclinical studies of mesenchymal stromal cells for ischemic stroke. Neurology. 2014;82(14):1277–1286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Ge J, Guo L, Wang S, Zhang Y, Cai T, Zhao RC, Wu Y. The size of mesenchymal stem cells is a significant cause of vascular obstructions and stroke. Stem Cell Rev. 2014;10(2):295–303. [DOI] [PubMed] [Google Scholar]
  • 90. Li H, Fan X, Kovi RC, Jo Y, Moquin B, Konz R, Stoicov C, Kurt-Jones E, Grossman SR, Lyle S, Rogers AB, Montrose M, Houghton J. Spontaneous expression of embryonic factors and p53 point mutations in aged mesenchymal stem cells: a model of age-related tumorigenesis in mice. Cancer Res. 2007;67(22):10889–10898. [DOI] [PubMed] [Google Scholar]
  • 91. Mitkari B, Kerkelä E, Nystedt J, Korhonen M, Jolkkonen J. Unexpected complication in a rat stroke model: exacerbation of secondary pathology in the thalamus by subacute intraarterial administration of human bone marrow-derived mesenchymal stem cells. J Cereb Blood Flow Metab. 2015;35(3):363–366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Yavagal DR, Lin B, Raval AP, Garza PS, Dong C, Zhao W, Rangel EB, McNiece I, Rundek T, Sacco RL, Perez-Pinzon M, Hare JM. Efficacy and dose-dependent safety of intra-arterial delivery of mesenchymal stem cells in a rodent stroke model. PLoS One. 2014;9(5):e93735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Wang LQ, Lin ZZ, Zhang HX, Shao B, Xiao L, Jiang HG, Zhuge QC, Xie LK, Wang B, Su DM, Jin KL. Timing and dose regimens of marrow mesenchymal stem cell transplantation affect the outcomes and neuroinflammatory response after ischemic stroke. CNS Neurosci Ther. 2014;20(4):317–326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Parys M, Nelson N, Koehl K, Miller R, Kaneene JB, Kruger JM, Yuzbasiyan-Gurkan V. Safety of intraperitoneal injection of adipose tissue-derived autologous mesenchymal stem cells in cats. J Vet Intern Med. 2016;30(1):157–163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Donders R, Vanheusden M, Bogie JF, Ravanidis S, Thewissen K, Stinissen P, Gyselaers W, Hendriks JJ, Hellings N. Human Wharton’s Jelly-derived stem cells display immunomodulatory properties and transiently improve rat experimental autoimmune encephalomyelitis. Cell Transplant. 2015;24(10):2077–2098. [DOI] [PubMed] [Google Scholar]
  • 96. Honmou O, Houkin K, Matsunaga T, Niitsu Y, Ishiai S, Onodera R, Waxman SG, Kocsis JD. Intravenous administration of auto serum-expanded autologous mesenchymal stem cells in stroke. Brain. 2011;134(Pt 6):1790–1807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Rodríguez-Frutos B, Otero-Ortega L, Gutiérrez-Fernández M, Fuentes B, Ramos-Cejudo J, Díez-Tejedor E. Stem cell therapy and administration routes after stroke. Transl Stroke Res. 2016;7(5):378–387. [DOI] [PubMed] [Google Scholar]
  • 98. Bang OY, Lee JS, Lee PH, Lee G. Autologous mesenchymal stem cell transplantation in stroke patients. Ann Neurol. 2005;57(6):874–82. [DOI] [PubMed] [Google Scholar]
  • 99. Lee JS, Hong JM, Choi JY, Lee PH, Ahn YH, Bang OY; STARTING collaborators. A long-term follow-up study of intravenous autologous mesenchymal stem cell transplantation in patients with ischemic stroke. Stem Cells. 2010;28(6):1099–1106. [DOI] [PubMed] [Google Scholar]
  • 100. Lalu MM, McIntyre L, Pugliese C, Fergusson D, Winston BW, Marshall JC, Granton J, Stewart DJ; Canadian Critical Care Trials Group. Safety of cell therapy with mesenchymal stromal cells (SafeCell): A systematic review and meta-analysis of clinical trials. PLoS One. 2012;7(10):e47559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Prasad K, Sharma A, Garg A, Mohanty S, Bhatnagar S, Johri S, Singh KK, Nair V, Sarkar RS, Gorthi SP, Hassan KM, Prabhakar S, Marwaha N, Khandelwal N, Misra UK, Kalita J, Nityanand S; InveST Study Group. Intravenous autologous bone marrow mononuclear stem cell therapy for ischemic stroke. A multicentric, randomized trial. Stroke. 2014; 45(12):3618–3624. [DOI] [PubMed] [Google Scholar]
  • 102. Napoli E, Borlongan CV. Recent advances in stem cell-based therapeutics for stroke. Transl Stroke Res. 2016;7(6):452–457. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cell Transplantation are provided here courtesy of SAGE Publications

RESOURCES