51
|
Doeppner TR, Ewert TAS, Tönges L, Herz J, Zechariah A, ElAli A, Ludwig AK, Giebel B, Nagel F, Dietz GPH, Weise J, Hermann DM, Bähr M. Transduction of neural precursor cells with TAT-heat shock protein 70 chaperone: therapeutic potential against ischemic stroke after intrastriatal and systemic transplantation. Stem Cells 2012; 30:1297-310. [PMID: 22593021 DOI: 10.1002/stem.1098] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Novel therapeutic concepts against cerebral ischemia focus on cell-based therapies in order to overcome some of the side effects of thrombolytic therapy. However, cell-based therapies are hampered because of restricted understanding regarding optimal cell transplantation routes and due to low survival rates of grafted cells. We therefore transplanted adult green fluorescence protein positive neural precursor cells (NPCs) either intravenously (systemic) or intrastriatally (intracerebrally) 6 hours after stroke in mice. To enhance survival of NPCs, cells were in vitro protein-transduced with TAT-heat shock protein 70 (Hsp70) before transplantation followed by a systematic analysis of brain injury and underlying mechanisms depending on cell delivery routes. Transduction of NPCs with TAT-Hsp70 resulted in increased intracerebral numbers of grafted NPCs after intracerebral but not after systemic transplantation. Whereas systemic delivery of either native or transduced NPCs yielded sustained neuroprotection and induced neurological recovery, only TAT-Hsp70-transduced NPCs prevented secondary neuronal degeneration after intracerebral delivery that was associated with enhanced functional outcome. Furthermore, intracerebral transplantation of TAT-Hsp70-transduced NPCs enhanced postischemic neurogenesis and induced sustained high levels of brain-derived neurotrophic factor, glial cell line-derived neurotrophic factor, and vascular endothelial growth factor in vivo. Neuroprotection after intracerebral cell delivery correlated with the amount of surviving NPCs. On the contrary, systemic delivery of NPCs mediated acute neuroprotection via stabilization of the blood-brain-barrier, concomitant with reduced activation of matrix metalloprotease 9 and decreased formation of reactive oxygen species. Our findings imply two different mechanisms of action of intracerebrally and systemically transplanted NPCs, indicating that systemic NPC delivery might be more feasible for translational stroke concepts, lacking a need of in vitro manipulation of NPCs to induce long-term neuroprotection.
Collapse
Affiliation(s)
- Thorsten R Doeppner
- Department of Neurology, University of Duisburg-Essen Medical School, Essen, Germany; Department of Neurology, University of Goettingen Medical School, Goettingen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
52
|
Abstract
Acute ischemic stroke causes a disturbance of neuronal circuitry and disruption of the blood-brain barrier that can lead to functional disabilities. At present, thrombolytic therapy inducing recanalization of the occluded vessels in the cerebral infarcted area is a commonly used therapeutic strategy. However, only a minority of patients have timely access to this kind of therapy. Recently, neural stem cells (NSCs) as therapy for stroke have been developed in preclinical studies. NSCs are harbored in the subventricular zone (SVZ) as well as the subgranular zone of the brain. The microenvironment in the SVZ, including intercellular interactions, extracellular matrix proteins, and soluble factors, can promote NSC proliferation, self-renewal, and multipotency. Endogenous neurogenesis responds to insults of ischemic stroke supporting the existence of remarkable plasticity in the mammalian brain. Homing and integration of NSCs to the sites of damaged brain tissue are complex morphological and physiological processes. This review provides an update on current preclinical cell therapies for stroke, focusing on neurogenesis in the SVZ and dentate gyrus and on recruitment cues that promote NSC homing and integration to the site of the damaged brain.
Collapse
Affiliation(s)
- Dah-Ching Ding
- Department of Obstetrics and Gynecology, Buddhist Tzu Chi General Hospital, Tzu Chi University, Hualien, Taiwan, ROC
| | | | | | | |
Collapse
|
53
|
Hicks C, Stevanato L, Stroemer RP, Tang E, Richardson S, Sinden JD. In vivo and in vitro characterization of the angiogenic effect of CTX0E03 human neural stem cells. Cell Transplant 2012; 22:1541-52. [PMID: 23067568 DOI: 10.3727/096368912x657936] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
CTX0E03 is a human neural stem cell line previously reported to reduce sensory motor deficits in a middle cerebral artery occlusion (MCAo) model of stroke. The objective of this study was to investigate if CTX0E03 treatment promotes angiogenesis. As stroke leads to damage of the vasculature in the brain, angiogenesis may contribute to the functional recovery. To test this hypothesis, the angiogenic activity of CTX0E03 was assessed both in vitro and in vivo. In vitro, CTX0E03 expression of trophic and proangiogenic factors was determined by real-time RT-PCR, Western blot, and ELISA, and its angiogenic activity was investigated in well-established angiogenesis assays. In vivo, angiogenesis was investigated in naive mice and MCAo rat brain and was evaluated by immunohistochemistry (IHC) using Von Willebrand factor (VWF), a marker of blood vessel formation, and BrdU/CD31 double labeling in naive mice only. In vitro results showed that CTX0E03-conditioned medium and coculture significantly increased total tubule formation compared with controls (p=0.002 and p=0.0008, respectively). Furthermore, CTX0E03 cells were found to be in direct association with the tubules by ICC. In vivo CTX0E03-treated brains demonstrated a significant increase in areas occupied by VWF-positive microvessels compared with vehicle-treated naive mice (two-way ANOVA, Interaction p<0.05, Treatment p<0.0001, Time p<0.0) and MCAo rat (p=0.001 unpaired t test, Welch's correction). CTX0E03-treated naive mouse brains showed an increase in BrdU/CD31 colabeling. In conclusion, in vitro CTX0E03 cells express proangiogenic factors and may promote angiogenesis by both release of paracrine factors and direct physical interaction. Furthermore, in vivo CTX0E03-treated rodent brains exhibited a significant increase in microvessels at the site of implantation compared with vehicle-injected groups. Taken together these data suggest that CTX0E03 cell therapy may provide significant benefit to stroke patients through upregulation of angiogenesis in the ischemic brain.
Collapse
Affiliation(s)
- Caroline Hicks
- ReNeuron Limited, Surrey Research Park, Guildford, Surrey, UK
| | | | | | | | | | | |
Collapse
|
54
|
dl-3-n-Butylphthalide protects rat bone marrow stem cells against hydrogen peroxide-induced cell death through antioxidation and activation of PI3K-Akt pathway. Neurosci Lett 2012; 516:247-52. [DOI: 10.1016/j.neulet.2012.04.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2012] [Revised: 03/28/2012] [Accepted: 04/01/2012] [Indexed: 11/24/2022]
|
55
|
Hermann DM, Chopp M. Promoting brain remodelling and plasticity for stroke recovery: therapeutic promise and potential pitfalls of clinical translation. Lancet Neurol 2012; 11:369-80. [PMID: 22441198 DOI: 10.1016/s1474-4422(12)70039-x] [Citation(s) in RCA: 258] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recent laboratory findings suggest that it might be possible to promote cerebral plasticity and neurological recovery after stroke by use of exogenous pharmacological or cell-based treatments. Brain microvasculature and glial cells respond in concert to ischaemic stressors and treatment, creating an environment in which successful recovery can ensue. Neurons remote from and adjacent to the ischaemic lesion are enabled to sprout, and neural precursor cells that accumulate with cerebral microvessels in the perilesional tissue further stimulate brain plasticity and neurological recovery. These factors interact in a highly dynamic way, facilitating temporally and spatially orchestrated responses of brain networks. In view of the complexity of the systems involved, stroke treatments that stimulate and amplify these endogenous restorative mechanisms might also provoke unwanted side-effects. In experimental studies, adverse effects have been identified when neurorestorative treatments were administered to animals with severe associated illnesses, after thrombolysis with alteplase, and when therapies were initiated outside appropriate time windows. Balancing the opportunities and possible risks, we provide suggestions for the translation of restorative therapies from the laboratory to the clinic.
Collapse
Affiliation(s)
- Dirk M Hermann
- Department of Neurology, University Hospital Essen, Essen, Germany.
| | | |
Collapse
|
56
|
Cossetti C, Alfaro-Cervello C, Donegà M, Tyzack G, Pluchino S. New perspectives of tissue remodelling with neural stem and progenitor cell-based therapies. Cell Tissue Res 2012; 349:321-9. [PMID: 22322425 DOI: 10.1007/s00441-012-1341-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Accepted: 01/25/2012] [Indexed: 01/06/2023]
Abstract
Compelling evidence exists that neural stem cell-based therapies protect the central nervous system (CNS) from chronic inflammatory degeneration, such as that occurring in experimental autoimmune encephalomyelitis and stroke. It was first assumed that stem cells directly replace lost cells but it is now becoming clearer that they might be able to protect the nervous system through mechanisms other than cell replacement. In immune-mediated experimental demyelination and stroke, transplanted neural stem/precursor cells (NPCs) are able to mediate efficient bystander myelin repair and axonal rescue. This is dependent on multiple capacities that transplanted NPCs exhibit within specific microenvironments after transplantation. However, a comprehensive understanding of the mechanisms by which NPCs exert their therapeutic impact is lacking. Here we will review some of the most recent evidence--and discuss some of the likely mechanisms--that support the remarkable capacity of NPCs to cross-talk with endogenous cells and to remodel the injured nervous system when applied as novel therapeutic regimes. We foresee that the exploitation of the innate mechanisms regulating these modalities of cell-to-cell communication has realistic chances of revolutionizing most of the actual understanding of stem cell biology and its application to regenerative medicine and CNS repair.
Collapse
Affiliation(s)
- Chiara Cossetti
- Department of Clinical Neurosciences, Cambridge Centre for Brain Repair and Cambridge Stem Cell Initiative, University of Cambridge, ED Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK
| | | | | | | | | |
Collapse
|
57
|
Stem cell therapy in stroke: Designing clinical trials. Neurochem Int 2011; 59:367-70. [DOI: 10.1016/j.neuint.2011.03.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Accepted: 03/04/2011] [Indexed: 01/19/2023]
|
58
|
Yuen CM, Sun CK, Lin YC, Chang LT, Kao YH, Yen CH, Chen YL, Tsai TH, Chua S, Shao PL, Leu S, Yip HK. Combination of cyclosporine and erythropoietin improves brain infarct size and neurological function in rats after ischemic stroke. J Transl Med 2011; 9:141. [PMID: 21864394 PMCID: PMC3177906 DOI: 10.1186/1479-5876-9-141] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 08/24/2011] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND This study tested the superiority of combined cyclosporine A (CsA)-erythropoietin (EPO) therapy compared with either one in limiting brain infarction area (BIA) and preserving neurological function in rat after ischemic stroke (IS). METHODS Fifty adult-male SD rats were equally divided into sham control (group 1), IS plus intra-peritoneal physiological saline (at 0.5/24/48 h after IS) (group 2), IS plus CsA (20.0 mg/kg at 0.5/24h, intra-peritoneal) (group 3), IS plus EPO (5,000IU/kg at 0.5/24/48h, subcutaneous) (group 4), combined CsA and EPO (same route and dosage as groups 3 and 4) treatment (group 5) after occlusion of distal left internal carotid artery. RESULTS BIA on day 21 after acute IS was higher in group 2 than in other groups and lowest in group 5 (all p < 0.01). The sensorimotor functional test showed higher frequency of left turning in group 2 than in other groups and lowest in group 5 (all p < 0.05). mRNA and protein expressions of apoptotic markers and number of apoptotic nuclei on TUNEL were higher in group 2 than in other groups and lowest in group 1 and 5, whereas the anti-apoptotic markers exhibited an opposite trend (all p < 0.05). The expressions of inflammatory and oxidized protein were higher in group 2 than in other groups and lowest in group 1 and 5, whereas anti-inflammatory markers showed reversed changes in group 1 and other groups (all p < 0.05). The number of aquaporin-4+ and glial fibrillary acid protein+ stained cells were higher in group 2 as compared to other groups and lowest in groups 1 and 5 (all p < 0.01). CONCLUSION combined treatment with CsA and EPO was superior to either one alone in protecting rat brain from ischemic damage after IS.
Collapse
Affiliation(s)
- Chun-Man Yuen
- Division of Trauma, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Abstract
Poststroke depression (PSD) occurs in approximately one-third of stroke survivors and is one of the serious sequelae of stroke. The onset of PSD causes delayed functional recovery by rehabilitation and also increases cognitive impairment. However, appropriate strategies for the therapy against ischemia-induced depression-like behaviors still remain to be developed. Such behaviors have been associated with a reduced level of brain-derived neurotrophic factor (BDNF). In addition, accumulating evidence indicates the ability of stem cells to improve cerebral ischemia-induced brain injuries. However, it remains to be clarified as to the effect of neural progenitor cells (NPCs) on PSD and the association between BDNF level and PSD. Using NPCs, we investigated the effect of intravenous injection of NPCs on PSD. We showed that injection of NPCs improved ischemia-induced depression-like behaviors in the forced-swimming test and sucrose preference test without having any effect on the viable area between vehicle- and NPC-injected ischemic rats. The injection of NPCs prevented the decrease in the level of BDNF in the ipsilateral hemisphere. The levels of phosphorylated CREB, ERK and Akt, which have been implicated in events downstream of BDNF signaling, were also decreased after cerebral ischemia. NPC injection inhibited these decreases in the phosphorylation of CREB and ERK, but not that of Akt. Our findings provide evidence that injection of NPCs may have therapeutic potential for the improvement of depression-like behaviors after cerebral ischemia and that these effects might be associated with restoring BDNF-ERK-CREB signaling.
Collapse
|
60
|
Intracranial Delivery of Stem Cells. Transl Stroke Res 2011; 2:266-71. [DOI: 10.1007/s12975-011-0095-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Revised: 07/13/2011] [Accepted: 07/13/2011] [Indexed: 11/27/2022]
|
61
|
Hermann DM. Future Perspectives for Brain Pharmacotherapies: Implications of Drug Transport Processes at the Blood-brain Barrier. Ther Adv Neurol Disord 2011; 1:167-79. [PMID: 21180575 DOI: 10.1177/1756285608097775] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Years ago, ischemic stroke was regarded as a model disease for the development of neuroprotective therapies by the pharmacological industry. Results were disappointing. There are still no treatments available allowing the rescue of brain tissue once a stroke has occurred. Study failure is not only a problem in the stroke field. In other neurodegenerative conditions and in non-degenerative brain disorders, progress in drug development was also rather scarce until recently. An important factor in drug failure is the blood-brain barrier, which expresses active transporters that eliminate drugs from the brain. These transporters exhibit strong variations between different animals, which make it difficult to predict brain concentrations of drugs over species barriers. This paper claims that more detailed knowledge about: (1) the biology of blood-brain barrier transporters; (2) their regulation in brain disease, (3) the affinity of transporters to candidate drugs; and (4) the accumulation of drugs in brain tissue is needed for the overall success of clinical trials to be improved. An alternative strategy could be the use of disease-modifying treatments that do not have to enter the brain to exert their function. As such, restorative and anti-inflammatory strategies acting at the blood-brain interface might gain therapeutic potential in the future.
Collapse
Affiliation(s)
- Dirk M Hermann
- Chair of Vascular Neurology and Dementia, Department of Neurology, University Hospital Essen, Essen, Germany
| |
Collapse
|
62
|
Kucharova K, Hefferan MP, Patel P, Marsala S, Nejime T, Miyanohara A, Marsala M, Drummond JC. Transplantation of rat synapsin-EGFP-labeled embryonic neurons into the intact and ischemic CA1 hippocampal region: distribution, phenotype, and axodendritic sprouting. Cell Transplant 2011; 20:1163-78. [PMID: 21669049 DOI: 10.3727/096368910x564544] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
A major limitation of neural transplantation studies is assessing the degree of host-graft interaction. In the present study, rat hippocampal/cortical embryonic neurons (E18) were infected with a lentivirus encoding enhanced green fluorescent protein (GFP) under control of the neuron-specific synapsin promoter, thus permitting robust identification of labeled neurons after in vivo grafting. Two weeks after transient forebrain ischemia or sham-surgery, GFP-expressing neurons were transplanted into CA1 hippocampal regions in immunosuppressed adult Wistar rats. The survival, distribution, phenotype, and axonal projections of GFP-immunoreactive (IR) positive transplanted neurons were evaluated in the sham-operated and ischemia- damaged CA1 hippocampal regions 4, 8, and 12 weeks after transplantation. In both experimental groups, we observed that the main phenotype of host-derived afferents projecting towards grafted GFP-IR neurons as well as transplant-derived GFP-IR efferents were glutamatergic in both animal groups. GFP axonal projections were seen in the nucleus accumbens, septal nuclei, and subiculum-known target areas of CA1 pyramidal neurons. Compared to sham-operated animals, GFP-IR neurons grafted into the ischemia-damaged CA1 migrated more extensively throughout a larger volume of host tissue, particularly in the stratum radiatum. Moreover, enhanced axonal sprouting and neuronal plasticity of grafted cells were evident in the hippocampus, subiculum, septal nuclei, and nucleus accumbens of the ischemia-damaged rats. Our study suggests that the adult rat brain retains some capacity to direct newly sprouting axons of transplanted embryonic neurons to the correct targets and that this capacity is enhanced in previously ischemia-injured forebrain.
Collapse
Affiliation(s)
- K Kucharova
- Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA.
| | | | | | | | | | | | | | | |
Collapse
|
63
|
Acute hepatocyte growth factor treatment induces long-term neuroprotection and stroke recovery via mechanisms involving neural precursor cell proliferation and differentiation. J Cereb Blood Flow Metab 2011; 31:1251-62. [PMID: 21119693 PMCID: PMC3099629 DOI: 10.1038/jcbfm.2010.211] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hepatocyte growth factor (HGF) is an interesting candidate for acute stroke treatment as shown by continuous infusion or gene delivery protocols. However, little is known about HGF-mediated long-term effects. The present study therefore analyzed long-term effects of an acute intrastriatal HGF treatment (5 μg) after a 45-minute stroke, with regard to brain injury and neurologic recovery. Hepatocyte growth factor induced long-term neuroprotection as assessed by infarct volume and neuronal cell death analysis for as long as 4 weeks after stroke, which was associated with sustained neurologic recovery as evidenced by corner-turn and tight-rope tests. Analyzing underlying mechanisms of HGF-induced sustained neuroprotection, enhanced cell proliferation followed by increased neuronal differentiation of neural precursor cells (NPCs) was observed in the ischemic striatum of HGF-treated mice, which persisted for up to 4 weeks. In line with this, HGF promoted neurosphere formation as well as proliferation of NPC and decreased caspase-3-dependent hypoxic injury in vitro. Preservation of blood-brain barrier integrity 24 hours after stroke was furthermore noticed in animals receiving HGF, which was associated with the inhibition of matrix metalloproteases (MMP)-2 and MMP-9 at 4 and 24 hours, respectively. We suggest that sustained recruitment of proliferating cells together with improved neurovascular remodeling provides an explanation for HGF-induced long-term neuroprotection.
Collapse
|
64
|
Kaeser M, Brunet JF, Wyss A, Belhaj-Saif A, Liu Y, Hamadjida A, Rouiller EM, Bloch J. Autologous Adult Cortical Cell Transplantation Enhances Functional Recovery Following Unilateral Lesion of Motor Cortex in Primates: A Pilot Study. Neurosurgery 2011; 68:1405-16; discussion 1416-7. [DOI: 10.1227/neu.0b013e31820c02c0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
65
|
Darsalia V, Allison SJ, Cusulin C, Monni E, Kuzdas D, Kallur T, Lindvall O, Kokaia Z. Cell number and timing of transplantation determine survival of human neural stem cell grafts in stroke-damaged rat brain. J Cereb Blood Flow Metab 2011; 31:235-42. [PMID: 20531461 PMCID: PMC3049487 DOI: 10.1038/jcbfm.2010.81] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2010] [Revised: 04/28/2010] [Accepted: 05/18/2010] [Indexed: 12/29/2022]
Abstract
Neural stem cells (NSCs) derived from human fetal striatum and transplanted as neurospheres survive in stroke-damaged striatum, migrate from the implantation site, and differentiate into mature neurons. Here, we investigated how various steps of neurogenesis are affected by intrastriatal transplantation of human NSCs at different time points after stroke and with different numbers of cells in each implant. Rats were subjected to middle cerebral artery occlusion and then received intrastriatal transplants of NSCs. Transplantation shortly after stroke (48 hours) resulted in better cell survival than did transplantation 6 weeks after stroke, but the delayed transplantation did not influence the magnitude of migration, neuronal differentiation, and cell proliferation in the grafts. Transplanting greater numbers of grafted NSCs did not result in a greater number of surviving cells or increased neuronal differentiation. A substantial number of activated microglia was observed at 48 hours after the insult in the injured striatum, but reached maximum levels 1 to 6 weeks after stroke. Our findings show that the best survival of grafted human NSCs in stroke-damaged brain requires optimum numbers of cells to be transplanted in the early poststroke phase, before the inflammatory response is established. These findings, therefore, have direct clinical implications.
Collapse
Affiliation(s)
- Vladimer Darsalia
- Laboratory of Neural Stem Cell Biology and Therapy, University Hospital, Lund, Sweden
- Lund Stem Cell Center, Lund, Sweden
| | - Susan J Allison
- Laboratory of Neural Stem Cell Biology and Therapy, University Hospital, Lund, Sweden
- Lund Stem Cell Center, Lund, Sweden
| | - Carlo Cusulin
- Laboratory of Neural Stem Cell Biology and Therapy, University Hospital, Lund, Sweden
- Lund Stem Cell Center, Lund, Sweden
| | - Emanuela Monni
- Laboratory of Neural Stem Cell Biology and Therapy, University Hospital, Lund, Sweden
- Lund Stem Cell Center, Lund, Sweden
| | - Daniela Kuzdas
- Laboratory of Neural Stem Cell Biology and Therapy, University Hospital, Lund, Sweden
- Lund Stem Cell Center, Lund, Sweden
| | - Therése Kallur
- Laboratory of Neural Stem Cell Biology and Therapy, University Hospital, Lund, Sweden
- Lund Stem Cell Center, Lund, Sweden
| | - Olle Lindvall
- Laboratory of Neurogenesis and Cell Therapy, Wallenberg Neuroscience Center, University Hospital, Lund, Sweden
- Lund Stem Cell Center, Lund, Sweden
| | - Zaal Kokaia
- Laboratory of Neural Stem Cell Biology and Therapy, University Hospital, Lund, Sweden
- Lund Stem Cell Center, Lund, Sweden
| |
Collapse
|
66
|
Functional recovery after hematic administration of allogenic mesenchymal stem cells in acute ischemic stroke in rats. Neuroscience 2010; 175:394-405. [PMID: 21144885 DOI: 10.1016/j.neuroscience.2010.11.054] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Revised: 11/23/2010] [Accepted: 11/27/2010] [Indexed: 12/19/2022]
Abstract
Hematic administration of bone marrow-derived mesenchymal stem cells (MSCs) in acute ischemic stroke may not only be an effective reparative treatment but also a brain protective therapy that improves neurological recovery. Our purpose was to study whether either i.v. or intracarotid (i.c.) administration of allogenic MSCs during the acute phase were effective in improving neurological recovery and decreasing brain damage in an experimental rat model. In a model of permanent middle cerebral artery occlusion (pMCAO), we analyzed: neurological evaluation; MSCs migration and implantation; interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) levels; lesion volume; cell death; cellular proliferation; vascular endothelial growth factor (VEGF) expression and blood vessel number. Regardless of the administration route, treated groups showed better neurological recovery, without significant differences between the two groups. Migration and implantation of MSCs in the lesion area was observed in animals receiving i.c. but not i.v. treatment. The highest cytokine values were observed in the i.v. MSCs and i.c. control groups, and these levels were significantly different from the corresponding i.v. control and i.c. MSCs groups, respectively. In addition, there were significant differences between the i.v. MSCs and i.c. MSCs groups in IL-6 levels. Neither treatment reduced infarction volume. However, cell death, measured as TUNEL+ cells was decreased with significant differences between control groups. BrdU+ cells were also significantly increased in the peri-infarct zone at 14 days. VEGF expression was significantly higher in the i.c. MSCs group than in the i.c. control group and blood vessel number was significantly higher in treated groups than control groups with significant differences in the peri-infarct zone at 14 days. We conclude that allogenic MSCs administration shows therapeutic efficacy in our acute ischemic stroke model. Both routes demonstrably improved neurological recovery and provided brain protection.
Collapse
|
67
|
Doeppner TR, Hermann DM. Mesenchymal stem cells in the treatment of ischemic stroke: progress and possibilities. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2010; 3:157-63. [PMID: 24198521 PMCID: PMC3781740 DOI: 10.2147/sccaa.s7820] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
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.
Collapse
Affiliation(s)
- Thorsten R Doeppner
- Department of Neurology, University of Duisburg-Essen Medical School, Essen, North Rhine-Westphalia, Germany
| | | |
Collapse
|
68
|
Pendharkar AV, Chua JY, Andres RH, Wang N, Gaeta X, Wang H, De A, Choi R, Chen S, Rutt BK, Gambhir SS, Guzman R. Biodistribution of neural stem cells after intravascular therapy for hypoxic-ischemia. Stroke 2010; 41:2064-70. [PMID: 20616329 DOI: 10.1161/strokeaha.109.575993] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND PURPOSE Intravascular transplantation of neural stem cells represents a minimally invasive therapeutic approach for the treatment of central nervous system diseases. The cellular biodistribution after intravascular injection needs to be analyzed to determine the ideal delivery modality. We studied the biodistribution and efficiency of targeted central nervous system delivery comparing intravenous and intra-arterial (IA) administration of neural stem cells after brain ischemia. METHODS Mouse neural stem cells were transduced with a firefly luciferase reporter gene for bioluminescence imaging (BLI). Hypoxic-ischemia was induced in adult mice and reporter neural stem cells were transplanted IA or intravenous at 24 hours after brain ischemia. In vivo BLI was used to track transplanted cells up to 2 weeks after transplantation and ex vivo BLI was used to determine single organ biodistribution. RESULTS Immediately after transplantation, BLI signal from the brain was 12 times higher in IA versus intravenous injected animals (P<0.0001). After IA injection, 69% of the total luciferase activity arose from the brain early after transplantation and 93% at 1 week. After intravenous injection, 94% of the BLI signal was detected in the lungs (P=0.004) followed by an overall 94% signal loss at 1 week, indicating lack of cell survival outside the brain. Ex vivo single organ analysis showed a significantly higher BLI signal in the brain than in the lungs, liver, and kidneys at 1 week (P<0.0001) and 2 weeks in IA (P=0.007). CONCLUSIONS IA transplantation results in superior delivery and sustained presence of neural stem cells in the ischemic brain in comparison to intravenous infusion.
Collapse
Affiliation(s)
- Arjun V Pendharkar
- Department of Neurosurgery, Stanford University, School of Medicine, Stanford, CA 94305-5327, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Leu S, Lin YC, Yuen CM, Yen CH, Kao YH, Sun CK, Yip HK. Adipose-derived mesenchymal stem cells markedly attenuate brain infarct size and improve neurological function in rats. J Transl Med 2010; 8:63. [PMID: 20584315 PMCID: PMC2913939 DOI: 10.1186/1479-5876-8-63] [Citation(s) in RCA: 174] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 06/28/2010] [Indexed: 12/16/2022] Open
Abstract
Background The therapeutic effect of adipose-derived mesenchymal stem cells (ADMSCs) on brain infarction area (BIA) and neurological status in a rat model of acute ischemic stroke (IS) was investigated. Methods Adult male Sprague-Dawley (SD) rats (n = 30) were divided into IS plus intra-venous 1 mL saline (at 0, 12 and 24 h after IS induction) (control group) and IS plus intra-venous ADMSCs (2.0 × 106) (treated interval as controls) (treatment group) after occlusion of distal left internal carotid artery. The rats were sacrificed and brain tissues were harvested on day 21 after the procedure. Results The results showed that BIA was larger in control group than in treatment group (p < 0.001). The sensorimotor functional test (Corner test) identified a higher frequency of turning movement to left in control group than in treatment group (p < 0.05). mRNA expressions of Bax, caspase 3, interleukin (IL)-18, toll-like receptor-4 and plasminogen activator inhibitor-1 were higher, whereas Bcl-2 and IL-8/Gro were lower in control group than in treatment group (all p < 0.05). Western blot demonstrated a lower CXCR4 and stromal-cell derived factor-1 (SDF-1) in control group than in treatment group (all p < 0.01). Immunohistofluorescent staining showed lower expressions of CXCR4, SDF-1, von Willebran factor and doublecortin, whereas the number of apoptotic nuclei on TUNEL assay was higher in control group than in treatment group (all p < 0.001). Immunohistochemical staining showed that cellular proliferation and number of small vessels were lower but glial fibrillary acid protein was higher in control group than in treatment group (all p < 0.01). Conclusions ADMSC therapy significantly limited BIA and improved sensorimotor dysfunction after acute IS.
Collapse
Affiliation(s)
- Steve Leu
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | | | | | | | | | | | | |
Collapse
|
70
|
Bifari F, Pacelli L, Krampera M. Immunological properties of embryonic and adult stem cells. World J Stem Cells 2010; 2:50-60. [PMID: 21607122 PMCID: PMC3097923 DOI: 10.4252/wjsc.v2.i3.50] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 03/17/2010] [Accepted: 03/24/2010] [Indexed: 02/06/2023] Open
Abstract
The possibility of treating degenerative diseases by stem cell-based approaches is a promising therapeutical option. Among major concerns for the clinical application of stem cells, some derive from the possibility that stem cells may be rejected by the immune system as a consequence of histoincompatibility and that stem cells themselves may interfere with the normal functions of host immune response. Therefore, the immunogenicity and the immunomodulatory properties of stem cells must be carefully addressed. Although these properties are common features of different stem cell types, some peculiarities can be recognized and characterized for their proper clinical use.
Collapse
Affiliation(s)
- Francesco Bifari
- Francesco Bifari, Luciano Pacelli, Mauro Krampera, Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Pz.le Scuro 10, 37134 Verona, Italy
| | | | | |
Collapse
|
71
|
Doeppner TR, El Aanbouri M, Dietz GPH, Weise J, Schwarting S, Bähr M. Transplantation of TAT-Bcl-xL-transduced neural precursor cells: long-term neuroprotection after stroke. Neurobiol Dis 2010; 40:265-76. [PMID: 20554038 DOI: 10.1016/j.nbd.2010.05.033] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Revised: 05/14/2010] [Accepted: 05/30/2010] [Indexed: 01/19/2023] Open
Abstract
Neural precursor cells (NPC) are an interesting tool in experimental stroke research, but their therapeutic potential is limited due to poor long-term survival. We therefore in vitro transduced subventricular zone-(SVZ)-derived NPC with the anti-apoptotic fusion protein TAT-Bcl-x(L) and analyzed NPC survival, differentiation, and post-stroke functional deficits after experimental ischemia in mice. Survival of TAT-Bcl-x(L)-transduced NPC, which were injected at day 7 post-stroke into the ischemic striatum, was significantly increased at 4 weeks after stroke. Increased survival of NPC was associated with reduced infarct injury and decreased post-stroke functional deficits. Animals grafted with TAT-Bcl-x(L)-transduced NPC showed an increased number of immature cells expressing the neuronal marker doublecortin. Since mature neuronal differentiation of NPC was not observed, reduced post-stroke injury cannot be attributed to enhanced neuronal regeneration, but rather to indirect by-stander effects of grafted NPC. In line with this, NPC-mediated neuroprotection of cortical neurons in vitro was associated with increased secretion of growth factors. Thus, in vitro transduction of cultivated NPC with TAT-Bcl-x(L) results in enhanced resistance of transplanted NPC followed by long-term neuroprotection and ameliorated functional deficits after transient focal cerebral ischemia in mice.
Collapse
Affiliation(s)
- Thorsten R Doeppner
- Department of Neurology, University of Goettingen Medical School, 37075 Goettingen, Germany.
| | | | | | | | | | | |
Collapse
|
72
|
Naegele JR, Maisano X, Yang J, Royston S, Ribeiro E. Recent advancements in stem cell and gene therapies for neurological disorders and intractable epilepsy. Neuropharmacology 2010; 58:855-64. [PMID: 20146928 PMCID: PMC2838966 DOI: 10.1016/j.neuropharm.2010.01.019] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2009] [Revised: 01/22/2010] [Accepted: 01/26/2010] [Indexed: 12/11/2022]
Abstract
The potential applications of stem cell therapies for treating neurological disorders are enormous. Many laboratories are focusing on stem cell treatments for CNS diseases, including spinal cord injury, Amyotrophic lateral sclerosis, Parkinson's disease, Huntington's disease, multiple sclerosis, stroke, traumatic brain injury, and epilepsy. Among the many stem cell types under testing for neurological treatments, the most common are fetal and adult brain stem cells, embryonic stem cells, induced pluripotent stem cells, and mesenchymal stem cells. An expanding toolbox of molecular probes is now available to allow analyses of neural stem cell fates prior to and after transplantation. Concomitantly, protocols are being developed to direct the fates of stem cell-derived neural progenitors, and also to screen stem cells for tumorigenicity and aneuploidy. The rapid progress in the field suggests that novel stem cell and gene therapies for neurological disorders are in the pipeline.
Collapse
Affiliation(s)
- Janice R Naegele
- Department of Biology and Program in Neuroscience and Behavior, Hall Atwater Laboratory, 52 Lawn Avenue, Wesleyan University, Middletown, CT 06459, USA.
| | | | | | | | | |
Collapse
|
73
|
Kubinová S, Syková E. Nanotechnology for treatment of stroke and spinal cord injury. Nanomedicine (Lond) 2010; 5:99-108. [PMID: 20025468 DOI: 10.2217/nnm.09.93] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The use of nanotechnology in cell therapy and tissue engineering offers promising future perspectives for brain and spinal cord injury treatment. Stem cells have been shown to selectively target injured brain and spinal cord tissue and improve functional recovery. To allow cell detection, superparamagnetic iron-oxide nanoparticles can be used to label transplanted cells. MRI is then a suitable method for the in vivo tracking of grafted cells in the host organism. CNS, and particularly spinal cord, injury is accompanied by tissue damage and the formation of physical and biochemical barriers that prevent axons from regenerating. One aspect of nanomedicine is the development of biologically compatible nanofiber scaffolds that mimic the structure of the extracellular matrix and can serve as a permissive bridge for axonal regeneration or as a drug-delivery system. The incorporation of biologically active epitopes and/or the utilization of these scaffolds as stem cell carriers may further enhance their therapeutic efficacy.
Collapse
Affiliation(s)
- Sárka Kubinová
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídenská 1083, 142 20 Prague, Czech Republic
| | | |
Collapse
|
74
|
Li L, Jiang Q, Ding G, Zhang L, Zhang ZG, Li Q, Panda S, Lu M, Ewing JR, Chopp M. Effects of administration route on migration and distribution of neural progenitor cells transplanted into rats with focal cerebral ischemia, an MRI study. J Cereb Blood Flow Metab 2010; 30:653-62. [PMID: 19888287 PMCID: PMC2844252 DOI: 10.1038/jcbfm.2009.238] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We tested the hypotheses that administration routes affect the migration and distribution of grafted neural progenitor cells (NPCs) in the ischemic brain and that the ischemic lesion plays a role in mediating the grafting process. Male Wistar rats (n=41) were subjected to 2-h middle cerebral artery occlusion (MCAo), followed 1 day later by administration of magnetically labeled NPCs. Rats with MCAo were assigned to one of three treatment groups targeted for cell transplantation intra-arterially (IA), intracisternally (IC), or intravenously (IV). MRI measurements consisting of T2-weighted imaging and three-dimensional (3D) gradient echo imaging were performed 24 h after MCAo, 4 h after cell injection, and once a day for 4 days. Prussian blue staining was used to identify the labeled cells, 3D MRI to detect cell migration and distribution, and T2 map to assess lesion volumes. Intra-arterial (IA) administration showed significantly increased migration, a far more diffuse distribution pattern, and a larger number of transplanted NPCs in the target brain than IC or IV administration. However, high mortality with IA delivery (IA: 41%; IC: 17%; IV: 8%) poses a serious concern for using this route of administration. Animals with smaller lesions at the time of transplantation have fewer grafted cells in the parenchyma.
Collapse
Affiliation(s)
- Lian Li
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan 48202, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Lindvall O, Kokaia Z. Stem cells in human neurodegenerative disorders--time for clinical translation? J Clin Invest 2010; 120:29-40. [PMID: 20051634 PMCID: PMC2798697 DOI: 10.1172/jci40543] [Citation(s) in RCA: 464] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Stem cell-based approaches have received much hype as potential treatments for neurodegenerative disorders. Indeed, transplantation of stem cells or their derivatives in animal models of neurodegenerative diseases can improve function by replacing the lost neurons and glial cells and by mediating remyelination, trophic actions, and modulation of inflammation. Endogenous neural stem cells are also potential therapeutic targets because they produce neurons and glial cells in response to injury and could be affected by the degenerative process. As we discuss here, however, significant hurdles remain before these findings can be responsibly translated to novel therapies. In particular, we need to better understand the mechanisms of action of stem cells after transplantation and learn how to control stem cell proliferation, survival, migration, and differentiation in the pathological environment.
Collapse
Affiliation(s)
- Olle Lindvall
- Address correspondence to: Olle Lindvall, Laboratory of Neurogenesis and Cell Therapy, Wallenberg Neuroscience Center, University Hospital, SE-221 84, Lund, Sweden. Phone: 46-46-222-0543; Fax: 46-46-222-0560; E-mail:
| | - Zaal Kokaia
- Address correspondence to: Olle Lindvall, Laboratory of Neurogenesis and Cell Therapy, Wallenberg Neuroscience Center, University Hospital, SE-221 84, Lund, Sweden. Phone: 46-46-222-0543; Fax: 46-46-222-0560; E-mail:
| |
Collapse
|
76
|
Farin A, Liu CY, Langmoen IA, Apuzzo ML. BIOLOGICAL RESTORATION OF CENTRAL NERVOUS SYSTEM ARCHITECTURE AND FUNCTION. Neurosurgery 2009; 65:831-59; discussion 859. [DOI: 10.1227/01.neu.0000351721.81175.0b] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
77
|
Koch P, Kokaia Z, Lindvall O, Brüstle O. Emerging concepts in neural stem cell research: autologous repair and cell-based disease modelling. Lancet Neurol 2009; 8:819-29. [PMID: 19679274 DOI: 10.1016/s1474-4422(09)70202-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The increasing availability of human pluripotent stem cells provides new prospects for neural-replacement strategies and disease-related basic research. With almost unlimited potential for self-renewal, the use of human embryonic stem cells (ESCs) bypasses the restricted supply and expandability of primary cells that has been a major bottleneck in previous neural transplantation approaches. Translation of developmental patterning and cell-type specification techniques to human ESC cultures enables in vitro generation of various neuronal and glial cell types. The derivation of stably proliferating neural stem cells from human ESCs further facilitates standardisation and circumvents the problem of batch-to-batch variations commonly encountered in "run-through" protocols, which promote terminal differentiation of pluripotent stem cells into somatic cell types without defined intermediate precursor stages. The advent of cell reprogramming offers an opportunity to translate these advances to induced pluripotent stem cells, thereby enabling the generation of neurons and glia from individual patients. Eventually, reprogramming could provide a supply of autologous neural cells for transplantation, and could lead to the establishment of cellular model systems of neurological diseases.
Collapse
Affiliation(s)
- Philipp Koch
- Institute of Reconstructive Neurobiology, Life & Brain Center, University of Bonn and Hertie Foundation, Bonn, Germany
| | | | | | | |
Collapse
|
78
|
Bordet R, Ouk T, Onténiente B, Charriaut-Marlangue C, Heurteaux C. Ischémie cérébrale. Med Sci (Paris) 2009; 25:847-54. [DOI: 10.1051/medsci/20092510847] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
79
|
Zhang P, Li J, Liu Y, Chen X, Kang Q, Zhao J, Li W. Human neural stem cell transplantation attenuates apoptosis and improves neurological functions after cerebral ischemia in rats. Acta Anaesthesiol Scand 2009; 53:1184-91. [PMID: 19650809 DOI: 10.1111/j.1399-6576.2009.02024.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Neuroprotection is a major therapeutic approach for ischemic brain injury. We investigated the neuroprotective effects induced by transplantation of human embryonic neural stem cells (NSCs) into the cortical penumbra 24 h after focal cerebral ischemia. METHODS NSCs were prepared from human embryonic brains obtained at 8 weeks of gestation. Focal cerebral ischemia was induced in adult rats by permanent occlusion of the middle cerebral artery. Animals were randomly divided into two groups: NSCs-grafted group and medium-grafted group (control). Infarct size was assessed 28 days after transplantation by hematoxylin and eosin staining. Neurological severity scores were evaluated before ischemia and at 1, 7, 14, and 28 days after transplantation. The terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay and immunohistochemical analysis of Bcl-2 and Bax were performed at 7, 14, and 28 days after transplantation. RESULTS Physiological parameters of the two groups were comparable, but not significantly different. NSC transplantation significantly improved neurological function (P<0.05) but did not reduce the infarct size significantly (P>0.05). Compared with the control, NSC transplantation significantly reduced the number of TUNEL- and Bax-positive cells in the penumbra at 7 days. Interestingly, the number of Bcl-2-positive cells in the penumbra after NSC transplantation was significantly higher than that after medium transplantation (P<0.05). CONCLUSIONS The results indicate that NSC transplantation has anti-apoptotic activity and can improve the neurological function; these effects are mediated by the up-regulation of Bcl-2 expression in the penumbra.
Collapse
Affiliation(s)
- P Zhang
- Institute of Neurobiology, National Key Academic Subject of Physiology, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | | | | | | | | | | | | |
Collapse
|
80
|
Huang G, Cao X, Zhang X, Chang H, Yang Y, Du W, Wilson JX. Effects of soybean isoflavone on the notch signal pathway of the brain in rats with cerebral ischemia. J Nutr Sci Vitaminol (Tokyo) 2009; 55:326-31. [PMID: 19763033 DOI: 10.3177/jnsv.55.326] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The aim of this study was to test the hypothesis that gastric lavage with soybean isoflavone activates the Notch signaling pathway and prevents apoptosis in the cerebral cortex during experimental strokes. Sprague-Dawley rats were randomly assigned to four groups of 10 rats each: sham operation plus vehicle (Sham), middle cerebral artery occlusion plus vehicle (MCAO), MCAO plus low dose soybean isoflavone (10 mg/(kg.d)) and MCAO plus high dose soybean isoflavone (50 mg/(kg.d)). The vehicle (saline, 10 mL/(kg.d)) and soybean isoflavone were administered by gastric lavage for 28 d prior to sham or MCAO operation and for 7 d afterward. The mRNA and protein expression levels of components of the Notch signaling system (Notch1 and Hes5) were measured by in situ hybridization and western blotting, respectively, whereas apoptosis was quantified by TUNEL assay. The results showed that MCAO stimulated expression of Notch1 and Hes5, at both the mRNA and protein levels, and also increased apoptosis. Soybean isoflavone dose-dependently augmented the stimulatory effect of MCAO on Notch1 and Hes5 expression levels but decreased apoptosis. These results identify a possible mechanism by which soybean isoflavone confers neuroprotection in strokes.
Collapse
Affiliation(s)
- Guowei Huang
- Tianjin Key Laboratory of Food Nutrition and Safety, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, China.
| | | | | | | | | | | | | |
Collapse
|
81
|
Bacigaluppi M, Pluchino S, Peruzzotti-Jametti L, Jametti LP, Kilic E, Kilic U, Salani G, Brambilla E, West MJ, Comi G, Martino G, Hermann DM. Delayed post-ischaemic neuroprotection following systemic neural stem cell transplantation involves multiple mechanisms. ACTA ACUST UNITED AC 2009; 132:2239-51. [PMID: 19617198 DOI: 10.1093/brain/awp174] [Citation(s) in RCA: 277] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Recent evidence suggests that neural stem/precursor cells (NPCs) promote recovery in animal models with delayed neuronal death via a number of indirect bystander effects. A comprehensive knowledge of how transplanted NPCs exert their therapeutic effects is still lacking. Here, we investigated the effects of a delayed transplantation of adult syngenic NPCs--injected intravenously 72 h after transient middle cerebral artery occlusion--on neurological recovery, histopathology and gene expression. NPC-transplanted mice showed a significantly improved recovery from 18 days post-transplantation (dpt) onwards, which persisted throughout the study. A small percentage of injected NPCs accumulated in the brain, integrating mainly in the infarct boundary zone, where most of the NPCs remained undifferentiated up to 30 dpt. Histopathological analysis revealed a hitherto unreported very delayed neuroprotective effect of NPCs, becoming evident at 10 and 30 dpt. Tissue survival was associated with downregulation of markers of inflammation, glial scar formation and neuronal apoptotic death at both mRNA and protein levels. Our data highlight the relevance of very delayed degenerative processes in the stroke brain that are intimately associated with inflammatory and glial responses. These processes may efficaciously be antagonized by (stem) cell-based strategies at time-points far beyond established therapeutic windows for pharmacological neuroprotection.
Collapse
Affiliation(s)
- Marco Bacigaluppi
- Department of Neurology, University Hospital Zurich, Frauenklinikstrasse 26, CH-8091 Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Zhang P, Li J, Liu Y, Chen X, Kang Q. Transplanted human embryonic neural stem cells survive, migrate, differentiate and increase endogenous nestin expression in adult rat cortical peri-infarction zone. Neuropathology 2009; 29:410-21. [PMID: 19170896 DOI: 10.1111/j.1440-1789.2008.00993.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Transplantation of stem cells is a potential therapeutic strategy for stroke damage. The survival, migration, and differentiation of transplanted human embryonic neural stem cells in the acute post-ischemic environment were characterized and endogenous nestin expression after transplantation was investigated. Human embryonic neural stem cells obtained from the temporal lobe cortex were cultured and labeled with fluorescent 1,1'-dioctadecy-6,6'-di (4-sulfopheyl)-3,3,3',3'-tetramethylindocarbocyanin (DiI) in vitro. Labeled cells were transplanted into cortical peri-infarction zones of adult rats 24 h after permanent middle cerebral artery occlusion. Survival, migration, and differentiation of grafted cells were quantified in immunofluorescence-stained sections from rats sacrificed at 7, 14, and 28 days after transplantation. Endogenous nestin-positive cells in the cortical peri-infarction zone were counted at serial time points. The cells transplanted into the cortical peri-infarction zone displayed the morphology of living cells and became widely located around the ischemic area. Moreover, some of the transplanted cells expressed nestin, GFAP, or NeuN in the peri-infarction zone. Furthermore, compared with the control group, endogenous nestin-positive cells in the peri-infarction zone had increased significantly 7 days after cell transplantation. These results confirm the survival, migration, and differentiation of transplanted cells in the acute post-ischemic environment and enhanced endogenous nestin expression within a brief time window. These findings indicate that transplantation of neural stem cells into the peri-infarction zone may be performed as early as 24 h after ischemia.
Collapse
Affiliation(s)
- Pengbo Zhang
- Institute of Neurobiology, National Key Academic Subject of Physiology, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | | | | | | | | |
Collapse
|
83
|
Dharmasaroja P. Bone marrow-derived mesenchymal stem cells for the treatment of ischemic stroke. J Clin Neurosci 2008; 16:12-20. [PMID: 19017556 DOI: 10.1016/j.jocn.2008.05.006] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2008] [Revised: 05/12/2008] [Accepted: 05/17/2008] [Indexed: 02/01/2023]
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs) have great potential as therapeutic agents in stroke management, since they are easily accessible and can be rapidly expanded ex vivo for autologous transplantation. Increasing evidence suggests that bone marrow cells migrate throughout the brain and differentiate into neurons and glial cells. Both non-human and human MSCs have been used to treat stroke in murine models with satisfactory results. Several factors, such as transdifferentiation, induction of neurogenesis and angiogenesis, neuroprotection, and activation of endogenous neurorestorative processes, contribute to the benefits of MSCs in the ischemic brain. Many variables, including types of MSCs, cell dose, timing of treatment, route of cell delivery, and characteristics of stroke patients, influence the efficacy of MSC treatment of stroke. Although the first trials of autologous MSC therapy in stroke patients showed promising results, the optimal approach for different clinical settings has yet to be determined. The fundamental properties of MSCs and their potential short-term and long-term toxicities also need to be determined before moving forward to use of these cells in clinical practice.
Collapse
Affiliation(s)
- Permphan Dharmasaroja
- Department of Anatomy and Center for Neuroscience, Faculty of Science, Mahidol University, Rama VI Road, Rajthevi, Bangkok 10400, Thailand.
| |
Collapse
|