301
|
Delayed functional maturation of human neuronal progenitor cells in vitro. Mol Cell Neurosci 2011; 47:36-44. [PMID: 21362477 DOI: 10.1016/j.mcn.2011.02.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 02/19/2011] [Accepted: 02/21/2011] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION Differentiation of neuronal progenitor cells (NPCs) in vitro into functional neurons is dependent on a complex cascade of molecular signaling pathways, many of which remain unknown. More specifically, in human NPCs the relationship between the expression of typical neuronal marker proteins and functional properties, such as firing action potential and synaptic transmission, is not well understood. In the present report, the immunocytochemical, morphological and electrophysiological changes that human NPCs undergo during neuronal differentiation in vitro were investigated. METHODS Human NPCs were differentiated toward a neuronal phenotype. The time course of the expression of neuronal markers and morphological cell changes was mapped and passive and active electrophysiological membrane properties assessed, throughout the neuronal maturation process. RESULTS The acquisition of neuronal markers preceded functional physiological maturation by several weeks. Cell input resistance decreased in the first 2 weeks as cells became less sensitive to input current, while cell capacitance progressively increased with continued neuronal process growth. Functional maturation was observed only by the fifth/sixth week, preceded by a marked increase in Na+ and K+ currents. In contrast, electrophysiological maturation of rodent precursor cells was observed at the end of the first week in vitro. Functionally, human neuronal cells became capable of firing action potentials and forming active synaptic contacts. Many features of the firing pattern however remained immature. CONCLUSIONS The results showed that human NPCs develop remarkably slowly and retain immature neuronal features for a prolonged period. The importance of Na-dependent activity for proper neuronal maturation is emphasized.
Collapse
|
302
|
Lee HJ, Lim IJ, Lee MC, Kim SU. Human neural stem cells genetically modified to overexpress brain-derived neurotrophic factor promote functional recovery and neuroprotection in a mouse stroke model. J Neurosci Res 2011; 88:3282-94. [PMID: 20818776 DOI: 10.1002/jnr.22474] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intracerebral hemorrhage (ICH) is a lethal stroke type; mortality approaches 50%, and current medical therapy against ICH shows only limited effectiveness, so an alternative approach is required, such as stem cell-based cell therapy. Previously we have shown that intravenously transplanted human neural stem cells (NSCs) selectively migrate to the brain and promote functional recovery in rat ICH model, and others have shown that intracerebral infusion of brain-derived neurotrophic factor (BDNF) results in improved structural and functional outcome from cerebral ischemia. We postulated that human NSCs overexpressing BDNF transplanted into cerebral cortex overlying ICH lesion could provide improved survival of grafted NSCs and increased angiogenesis and behavioral recovery in mouse ICH model. ICH was induced in adult mice by injection of bacterial collagenase into striatum. The HB1.F3.BDNF (F3.BDNF) human NSC line produces sixfold higher amounts of BDNFF over the parental F3 cell line in vitro, induces behavioral improvement, and produces a threefold increase in cell survival at 2 weeks and 8 weeks posttransplantation. Brain transplantation of human NSCs overexpressing BDNF provided differentiation and survival of grafted human NSCs and renewed angiogenesis of host brain and functional recovery of ICH animals. These results indicate that the F3.BDNF human NSCs should be of great value as a cellular source for experimental studies involving cellular therapy for human neurological disorders, including ICH.
Collapse
Affiliation(s)
- Hong J Lee
- Medical Research Institute, Chung-Ang University College of Medicine, Seoul, Korea
| | | | | | | |
Collapse
|
303
|
Cord blood transplantation and stem cell regenerative potential. Exp Hematol 2011; 39:393-412. [PMID: 21238533 DOI: 10.1016/j.exphem.2011.01.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Revised: 01/06/2011] [Accepted: 01/08/2011] [Indexed: 02/06/2023]
Abstract
The past 20 years of experience with umbilical cord blood transplantation have demonstrated that cord blood is effective in the treatment of a spectrum of diseases, including hematological malignancies, bone marrow failure, hemoglobinopathies, and inborn errors of metabolism. Cord blood can be obtained with ease and then safely cryopreserved for either public or private use without loss of viability. As compared to other unrelated donor cell sources, cord blood transplantation allows for greater human leukocyte antigen disparity without a corresponding increase in graft-vs.-host disease. Moreover, cord blood has a lower risk of transmitting infections by latent viruses and is less likely to carry somatic mutations than other adult cells. Recently, multiple populations of stem cells with primitive stem cell properties have been identified from cord blood. Meanwhile, there is an increasing interest in applying cord blood mononuclear cells or enriched stem cell populations to regenerative therapies. Accumulating evidence has suggested functional improvements after cord blood transplantation in various animal models for treatments of cardiac infarction, diabetes, neurological diseases, etc. In this review, we will summarize the most recent updates on clinical applications of cord blood transplantation and the promises and limitations of cell-based therapies for tissue repair and regeneration.
Collapse
|
304
|
Enhancing Stroke Recovery with Cellular Therapies. Stroke 2011. [DOI: 10.1016/b978-1-4160-5478-8.10057-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
305
|
Banerjee S, Williamson D, Habib N, Gordon M, Chataway J. Human stem cell therapy in ischaemic stroke: a review. Age Ageing 2011; 40:7-13. [PMID: 21071454 DOI: 10.1093/ageing/afq133] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Stroke is a leading cause of death and disability. Globally, 15 million people suffer a stroke each year, of whom more than 5 million die, and a further 5 million are left permanently disabled. Current treatment options offer modest benefits, and there is a pressing need for new and effective treatments. Stem cell therapy is a well-established treatment modality for various haematological diseases, with its use now being explored in different disease processes, including various neurological diseases, as well as vascular conditions such as ischaemic heart disease and peripheral vascular disease. Promising results have been seen in animal models of stroke, with evidence of significant functional benefits. Translation to the bedside, however, is in its early stages. This review will discuss the scientific background to stem cell therapy in ischaemic stroke, including evidence from current clinical trials.
Collapse
Affiliation(s)
- Soma Banerjee
- Department of Neurology, St Mary's Hospital, Imperial College Healthcare NHS Trust, London W21NY, UK.
| | | | | | | | | |
Collapse
|
306
|
Onteniente B, Polentes J. Regenerative Medicine for Stroke – Are We There Yet. Cerebrovasc Dis 2011; 31:544-51. [DOI: 10.1159/000324325] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Accepted: 01/11/2011] [Indexed: 01/01/2023] Open
|
307
|
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
|
308
|
Hartman NW, Carpentino JE, LaMonica K, Mor DE, Naegele JR, Grabel L. CXCL12-mediated guidance of migrating embryonic stem cell-derived neural progenitors transplanted into the hippocampus. PLoS One 2010; 5:e15856. [PMID: 21209827 PMCID: PMC3013129 DOI: 10.1371/journal.pone.0015856] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 11/26/2010] [Indexed: 01/19/2023] Open
Abstract
Stem cell therapies for neurodegenerative disorders require accurate delivery of the transplanted cells to the sites of damage. Numerous studies have established that fluid injections to the hippocampus can induce lesions in the dentate gyrus (DG) that lead to cell death within the upper blade. Using a mouse model of temporal lobe epilepsy, we previously observed that embryonic stem cell-derived neural progenitors (ESNPs) survive and differentiate within the granule cell layer after stereotaxic delivery to the DG, replacing the endogenous cells of the upper blade. To investigate the mechanisms for ESNP migration and repair in the DG, we examined the role of the chemokine CXCL12 in mice subjected to kainic acid-induced seizures. We now show that ESNPs transplanted into the DG show extensive migration through the upper blade, along the septotemporal axis of the hippocampus. Seizures upregulate CXCL12 and infusion of the CXCR4 antagonist AMD3100 by osmotic minipump attenuated ESNP migration. We also demonstrate that seizures promote the differentiation of transplanted ESNPs toward neuronal rather than astrocyte fates. These findings suggest that ESNPs transplanted into the adult rodent hippocampus migrate in response to cytokine-mediated signals.
Collapse
Affiliation(s)
- Nathaniel W Hartman
- Department of Biology, Wesleyan University, Middletown, Connecticut, United States of America.
| | | | | | | | | | | |
Collapse
|
309
|
Lee IS, Jung K, Kim M, Park KI. Neural stem cells: properties and therapeutic potentials for hypoxic-ischemic brain injury in newborn infants. Pediatr Int 2010; 52:855-65. [PMID: 21029253 DOI: 10.1111/j.1442-200x.2010.03266.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Neural stem cells (NSCs) are defined by their ability to self-renew, to differentiate into cells of all glial and neuronal lineages throughout the neuraxis, and to populate developing or degenerating central nervous system (CNS) regions. The recognition that NSCs propagated in culture could be reimplanted into the mammalian brain, where they might integrate appropriately throughout the mammalian CNS and stably express foreign genes, has unveiled a new role for neural transplantation and gene therapy and a possible strategy for addressing the CNS manifestations of diseases that hitherto had been refractory to intervention. An intriguing phenomenon with possible therapeutic potentials has begun to emerge from our observations of the behavior of NSCs in animal models of neonatal hypoxic-ischemic (HI) brain injury. During phases of active neurodegeneration, factors seem to be transiently elaborated to which NSCs may respond by migrating to degenerating regions and differentiating specifically towards replacement of dying neural cells. NSCs may attempt to repopulate and reconstitute ablated regions. These 'repair mechanisms' may actually reflect the reexpression of basic developmental principles that may be harnessed for therapeutic ends. In addition, NSCs may serve as vehicles for gene delivery and appear capable of simultaneous neural cell replacement and gene therapy (e.g. with factors that might enhance neuronal differentiation, neurites outgrowth, proper connectivity, and/or neuroprotection). When combined with certain synthetic biomaterials, NSCs may be even more effective in 'engineering' the damaged CNS towards reconstitution. We have also cultured human NSCs or progenitors as neurospheres which were derived from fetal cadavers at 13 weeks of gestation, and transplanted them into HI-injured immature brains to investigate their therapeutic potentials in this type of model.
Collapse
Affiliation(s)
- Il-Shin Lee
- Department of Pediatrics and BK21 Project for Medical Sciences, Yonsei University College of Medicine, Severance Children's Hospital, Seoul, Korea
| | | | | | | |
Collapse
|
310
|
Jin K, Mao X, Xie L, Greenberg RB, Peng B, Moore A, Greenberg MB, Greenberg DA. Delayed transplantation of human neural precursor cells improves outcome from focal cerebral ischemia in aged rats. Aging Cell 2010; 9:1076-83. [PMID: 20883527 PMCID: PMC2980591 DOI: 10.1111/j.1474-9726.2010.00638.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Neural precursor cell (NPC) transplantation may have a role in restoring brain function after stroke, but how aging might affect the brain's receptivity to such transplants is unknown. We reported previously that transplantation of human embryonic stem cell (hESC)-derived NPCs together with biomaterial (Matrigel) scaffolding into the brains of young adult Sprague-Dawley rats 3 weeks after distal middle cerebral artery occlusion (MCAO) reduced infarct volume and improved neurobehavioral performance. In this study, we compared the effect of NPC and Matrigel transplants in young adult (3-month-old) and aged (24-month-old) Fisher 344 rats from the National Institute on Aging's aged rodent colony. Distal MCAO was induced by electrocoagulation, and hESC-derived NPCs were transplanted into the infarct cavity 3 weeks later. Aged rats developed larger infarcts, but infarct volume and performance on the cylinder and elevated body swing tests, measured 6-8 weeks post-transplant, were improved by transplantation. We conclude that advanced age does not preclude a beneficial response to NPC transplantation following experimental stroke.
Collapse
Affiliation(s)
- Kunlin Jin
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945
| | - XiaoOu Mao
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945
| | - Lin Xie
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945
| | - Rose B. Greenberg
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945
| | - Botao Peng
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945
| | - Alexander Moore
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945
| | - Maeve B. Greenberg
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945
| | - David A. Greenberg
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945
| |
Collapse
|
311
|
Rota Nodari L, Ferrari D, Giani F, Bossi M, Rodriguez-Menendez V, Tredici G, Delia D, Vescovi AL, De Filippis L. Long-term survival of human neural stem cells in the ischemic rat brain upon transient immunosuppression. PLoS One 2010; 5:e14035. [PMID: 21124963 PMCID: PMC2988794 DOI: 10.1371/journal.pone.0014035] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Accepted: 10/29/2010] [Indexed: 12/20/2022] Open
Abstract
Understanding the physiology of human neural stem cells (hNSCs) in the context of cell therapy for neurodegenerative disorders is of paramount importance, yet large-scale studies are hampered by the slow-expansion rate of these cells. To overcome this issue, we previously established immortal, non-transformed, telencephalic-diencephalic hNSCs (IhNSCs) from the fetal brain. Here, we investigated the fate of these IhNSC's immediate progeny (i.e. neural progenitors; IhNSC-Ps) upon unilateral implantation into the corpus callosum or the hippocampal fissure of adult rat brain, 3 days after global ischemic injury. One month after grafting, approximately one fifth of the IhNSC-Ps had survived and migrated through the corpus callosum, into the cortex or throughout the dentate gyrus of the hippocampus. By the fourth month, they had reached the ipsilateral subventricular zone, CA1-3 hippocampal layers and the controlateral hemisphere. Notably, these results could be accomplished using transient immunosuppression, i.e administering cyclosporine for 15 days following the ischemic event. Furthermore, a concomitant reduction of reactive microglia (Iba1+ cells) and of glial, GFAP+ cells was also observed in the ipsilateral hemisphere as compared to the controlateral one. IhNSC-Ps were not tumorigenic and, upon in vivo engraftment, underwent differentiation into GFAP+ astrocytes, and β-tubulinIII+ or MAP2+ neurons, which displayed GABAergic and GLUTAmatergic markers. Electron microscopy analysis pointed to the formation of mature synaptic contacts between host and donor-derived neurons, showing the full maturation of the IhNSC-P-derived neurons and their likely functional integration into the host tissue. Thus, IhNSC-Ps possess long-term survival and engraftment capacity upon transplantation into the globally injured ischemic brain, into which they can integrate and mature into neurons, even under mild, transient immunosuppressive conditions. Most notably, transplanted IhNSC-P can significantly dampen the inflammatory response in the lesioned host brain. This work further supports hNSCs as a reliable and safe source of cells for transplantation therapy in neurodegenerative disorders.
Collapse
Affiliation(s)
- Laura Rota Nodari
- Department of Biotechnologies and Biosciences, University Milano Bicocca, Milan, Italy
| | - Daniela Ferrari
- Department of Biotechnologies and Biosciences, University Milano Bicocca, Milan, Italy
| | - Fabrizio Giani
- Department of Biotechnologies and Biosciences, University Milano Bicocca, Milan, Italy
| | - Mario Bossi
- Department of Neurosciences and Biomedical Technologies, University Milano Bicocca, Milan, Italy
| | | | - Giovanni Tredici
- Department of Neurosciences and Biomedical Technologies, University Milano Bicocca, Milan, Italy
| | - Domenico Delia
- Department of Experimental Oncology, Fondazione IRCSS Istituto Nazionale Tumori, Milan, Italy
| | - Angelo Luigi Vescovi
- Department of Biotechnologies and Biosciences, University Milano Bicocca, Milan, Italy
- IRCCS Casa Sollievo della Sofferenza, Opera di San Pio da Pietralcina, San Giovanni Rotondo, Italy
- * E-mail: (LDF); (ALV)
| | - Lidia De Filippis
- Department of Biotechnologies and Biosciences, University Milano Bicocca, Milan, Italy
- * E-mail: (LDF); (ALV)
| |
Collapse
|
312
|
Abstract
MicroRNAs play roles in developmental switching; however, their roles in human neural progenitor cells (hNPCs) is poorly understood. In this issue of Cell Stem Cell, Delaloy et al. (2010) report that proliferation and migration choices in hNPCs are regulated by miR-9.
Collapse
Affiliation(s)
- Nobuko Uchida
- StemCells Inc., 3155 Porter Drive, Palo Alto, CA 94301, USA.
| |
Collapse
|
313
|
Yu H, Cao B, Feng M, Zhou Q, Sun X, Wu S, Jin S, Liu H, Lianhong J. Combinated transplantation of neural stem cells and collagen type I promote functional recovery after cerebral ischemia in rats. Anat Rec (Hoboken) 2010; 293:911-7. [PMID: 20191618 DOI: 10.1002/ar.20941] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Using tissue engineering, a complex of neural stem cells (NSCs) and collagen type I was transplanted for the therapy of cerebral ischemic injury. NSCs from E14 d rats were dissociated and cultured by neurosphere formation in serum-free medium in the presence of basic fibroblast growth factor (bFGF), then seeded onto collagen to measure cell adhesive ability. BrdU was added to the culture medium to label the NSCs. Wistar rats (n=100) were subjected to 2-hour middle cerebral artery occlusion. After 24 hours of reperfusion, rats were assigned randomly to five groups: NSCs-collagen repair group, NSCs repair group, unseeded collagen repair group, MCAO medium group, and sham group. Neurological, immunohistological and electronic microscope assessments were performed to examine the effects of these treatments. Scanning electronic microscopy showed that NSCs assemble in the pores of collagen. At 3, 7, 15, and 30 d after transplantation of the NSC-collagen complex, some of the engrafted NSCs survive, differentiate and form synapses in the brains of rats subjected to cerebral ischemia. Six d after transplantation of the NSC-collagen complex into the brains of ischemic rats, the collagen began to degrade; 30 d after transplantation, the collagen had degraded completely. The implantation of NSCs and type I collagen facilitated the structural and functional recovery of neural tissue following ischemic injury.
Collapse
Affiliation(s)
- Hongwei Yu
- Department of Histology and Embryology, Harbin Medical University, Harbin, China
| | | | | | | | | | | | | | | | | |
Collapse
|
314
|
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
|
315
|
Hematopoietic stem cell transplantation protects mice from lethal stroke. Exp Neurol 2010; 225:284-93. [PMID: 20547154 DOI: 10.1016/j.expneurol.2010.06.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 05/26/2010] [Accepted: 06/07/2010] [Indexed: 01/19/2023]
Abstract
Stroke is a major cause of mortality and morbidity in the United States. The ideal therapeutic approach would minimize cell death and regenerate brain tissue. In order to investigate some questions that are related to such an approach, we have generated a mouse model in which we induce a stroke using the middle cerebral artery occlusion method. After 2h occlusion followed by reperfusion, 99% of mice died within 8 days of stroke. Total bone marrow cell transplantation by intravenous injection revealed an optimal timing of cell transfer in two doses on days 1 (same day of surgery) and 2 after surgery. Moreover, intravenous injection of Sca1+ bone marrow cells (enriched in hematopoietic stem cells) showed a dose-response effect on survival. Surviving mice also had no signs of apparent paralysis or weakness. Tracking analysis using donor stem cells expressing LacZ revealed only few donor cells in the brain. We conclude that hematopoietic stem cell-rich Sca1+ bone marrow cell transplantation after stroke protects the brain of a sizeable portion of mice subjected to stroke and alleviate remarkably the resulting neurological morbidity in surviving mice.
Collapse
|
316
|
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
|
317
|
Yilmaz G, Alexander JS, Erkuran Yilmaz C, Granger DN. Induction of neuro-protective/regenerative genes in stem cells infiltrating post-ischemic brain tissue. EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2010; 2:11. [PMID: 20509949 PMCID: PMC2893124 DOI: 10.1186/2040-7378-2-11] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Accepted: 05/28/2010] [Indexed: 12/25/2022]
Abstract
BACKGROUND Although the therapeutic potential of bone marrow-derived stromal stem cells (BMSC) has been demonstrated in different experimental models of ischemic stroke, it remains unclear how stem cells (SC) induce neuroprotection following stroke. In this study, we describe a novel method for isolating BMSC that infiltrate postischemic brain tissue and use this method to identify the genes that are persistently activated or depressed in BMSC that infiltrate brain tissue following ischemic stroke. METHODS Ischemic strokes were induced in C57BL/6 mice by middle cerebral artery occlusion for 1 h, followed by reperfusion. BMSC were isolated from H-2 Kb-tsA58 (immortomouse) mice, and were administered (i.v.) 24 h after reperfusion. At the peak of therapeutic improvement (14 days after the ischemic insult), infarcted brain tissue was isolated, and the BMSC were isolated by culturing at 33 degrees C. Microarray analysis and RT-PCR were performed to compare differential gene expression between naïve and infiltrating BMSC populations. RESULTS Z-scoring revealed dramatic differences in the expression of extracellular genes between naïve and infiltrating BMSC. Pair-wise analysis detected 80 extracellular factor genes that were up-regulated (>/= 2 fold, P < 0.05, Benjamini-Hochberg correction) between naïve and infiltrated BMSC. Although several anticipated neuroregenerative, nerve guidance and angiogenic factor (e.g., bFGF, bone morphogenetic protein, angiopoietins, neural growth factor) genes exhibited an increased expression, a remarkable induction of genes for nerve guidance survival (e.g., cytokine receptor-like factor 1, glypican 1, Dickkopf homolog 2, osteopontin) was also noted. CONCLUSIONS BMSC infiltrating the post-ischemic brain exhibit persistent epigenetic changes in gene expression for numerous extracellular genes, compared to their naïve counterparts. These genes are relevant to the neuroprotection, regeneration and angiogenesis previously described following stem cell therapy in animal models of ischemic stroke.
Collapse
Affiliation(s)
- Gokhan Yilmaz
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, Shreveport, LA, USA.
| | | | | | | |
Collapse
|
318
|
Lundberg J, Jonsson S, Holmin S. New endovascular method for transvascular exit of arteries and veins: developed in simulator, in rat and in rabbit with full clinical integration. PLoS One 2010; 5:e10449. [PMID: 20454668 PMCID: PMC2862716 DOI: 10.1371/journal.pone.0010449] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2010] [Accepted: 04/07/2010] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Endovascular technique has benefits vis-a-vis surgical access to organs with less accessible anatomical locations. To minimize surgical risk we propose a novel endovascular technique, to create parenchymal access through endovascular methods. METHODOLOGY/PRINCIPAL FINDINGS We have developed, manufactured and tested an endovascular catheter with a depth limiting collar and a penetrating tip that is used to perforate vessels, thereby creating a working channel to the extra-vascular space. Computer simulations and subsequent interventions have been performed ex vivo and in vivo in both small and large animals by testing different prototypes. All tests were designed for testing extravascular hemostasis and absence of thrombo-embolic complications when exiting the vessels from the inside to the extra vascular space. We have deposited prototypes after intervention in vascular walls over a period of 14 days in rat with no impairment on blood flow and no signs of thrombo-embolic complications upon re-exploration (n = 7). We have also incorporated the catheter system with clinically available systems both in an ex vivo simulator setting and in a full scale clinical angiographical setting in rabbit were no bleeding (0%) in any of the interventions performed (n = 40). To prevent hemorrhage during termination of the procedure, a hollow electrolysis detachment-zone leaves the distal tip in the vessel-wall after the intervention. This has also been tested with absolute hemostasis in large animals (n = 6). CONCLUSIONS/SIGNIFICANCE We have developed and tested a new system for transvascular tissue access in simulations, ex vivo and in vivo in small and large animals, integrating it with standard clinical catheters and angiographical environment, with absolute hemostasis and without thromboembolic complications. In a clinical setting for stem cell transplantation, local substance administration or tissue sampling, the benefit should be greatest in organs that are difficult or high-risk to access with other techniques, such as the pancreas, the central nervous system (CNS) and the heart.
Collapse
Affiliation(s)
- Johan Lundberg
- Department of Clinical Neuroscience, Karolinska Institutet and Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Stefan Jonsson
- Department of Materials Science and Engineering, Royal Institute of Technology, Stockholm, Sweden
| | - Staffan Holmin
- Department of Clinical Neuroscience, Karolinska Institutet and Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
319
|
Zhong J, Chan A, Morad L, Kornblum HI, Fan G, Carmichael ST. Hydrogel matrix to support stem cell survival after brain transplantation in stroke. Neurorehabil Neural Repair 2010; 24:636-44. [PMID: 20424193 DOI: 10.1177/1545968310361958] [Citation(s) in RCA: 167] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Stroke is a leading cause of adult disability. Stem/progenitor cell transplantation improves recovery after stroke in rodent models. These studies have 2 main limitations to clinical translation. First, most of the cells in stem/progenitor transplants die after brain transplantation. Second, intraparenchymal approaches target transplants to normal brain adjacent to the stroke, which is the site of the most extensive natural recovery in humans. Transplantation may damage this tissue. The stroke cavity provides an ideal target for transplantation because it is a compartmentalized region of necrosis, can accept a high volume transplant without tissue damage, and lies directly adjacent to the most plastic brain area in stroke. However, direct transplantation into the stroke cavity has caused massive death in the transplant. To overcome these limitations, the authors tested stem/progenitor transplants within a specific biopolymer hydrogel matrix to create a favorable environment for transplantation into the infarct cavity after stroke, and they tested this in comparison to stem cell injection without hydrogel support. A biopolymer hydrogel composed of cross-linked hyaluronan and heparin sulfate significantly promoted the survival of 2 different neural progenitor cell lines in vitro in conditions of stress and in vivo into the infarct cavity. Quantitative analysis of the transplant and surrounding tissue indicates diminished inflammatory infiltration of the graft with the hydrogel transplant. This result indicates that altering the local environment in stem cell transplantation enhances survival and diminishes cell stress. Stem cell transplantation into the infarct cavity within a pro-survival hydrogel matrix may provide a translational therapy for stroke recovery.
Collapse
Affiliation(s)
- Jin Zhong
- David Geffen School of Medicine at UCLA, Los Angeles, CA 98895, USA
| | | | | | | | | | | |
Collapse
|
320
|
Gera A, Steinberg GK, Guzman R. In vivo neural stem cell imaging: current modalities and future directions. Regen Med 2010; 5:73-86. [PMID: 20017696 DOI: 10.2217/rme.09.79] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Neural stem cells have been proposed as a promising therapy for treating a wide variety of neuropathologies. While several studies have demonstrated the therapeutic benefits of neural stem cells, the exact mechanism remains elusive. In order to facilitate research efforts to understand these mechanisms, and before neural stem cell-based therapies can be utilized in a clinical context, we must develop means of monitoring these cells in vivo. However, because of tissue depth and the blood-brain barrier, in vivo imaging of neural stem cells in the brain has unique challenges that do not apply to stem cells for other purposes. In this paper, we review contemporary methods for in vivo neural stem cell imaging, including MRI, PET and optical imaging techniques.
Collapse
Affiliation(s)
- Atul Gera
- Department of Neurosurgery and Stanford Stroke Center, Stanford University School of Medicine, 300 Pasteur Drive, R2111, Stanford, CA 94305-95327, USA
| | | | | |
Collapse
|
321
|
Neri M, Maderna C, Ferrari D, Cavazzin C, Vescovi AL, Gritti A. Robust generation of oligodendrocyte progenitors from human neural stem cells and engraftment in experimental demyelination models in mice. PLoS One 2010; 5:e10145. [PMID: 20405042 PMCID: PMC2853578 DOI: 10.1371/journal.pone.0010145] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Accepted: 03/16/2010] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Cell-based therapy holds great promises for demyelinating diseases. Human-derived fetal and adult oligodendrocyte progenitors (OPC) gave encouraging results in experimental models of dysmyelination but their limited proliferation in vitro and their potential immunogenicity might restrict their use in clinical applications. Virtually unlimited numbers of oligodendroglial cells could be generated from long-term self-renewing human (h)-derived neural stem cells (hNSC). However, robust oligodendrocyte production from hNSC has not been reported so far, indicating the need for improved understanding of the molecular and environmental signals controlling hNSC progression through the oligodendroglial lineage. The aim of this work was to obtain enriched and renewable cultures of hNSC-derived oligodendroglial cells by means of epigenetic manipulation. METHODOLOGY/PRINCIPAL FINDINGS We report here the generation of large numbers of hNSC-derived oligodendroglial cells by concurrent/sequential in vitro exposure to combinations of growth factors (FGF2, PDGF-AA), neurotrophins (NT3) and hormones (T3). In particular, the combination FGF2+NT3+PDGF-AA resulted in the maintenance and enrichment of an oligodendroglial cell population displaying immature phenotype (i.e., proliferation capacity and expression of PDGFRalpha, Olig1 and Sox10), limited self-renewal and increased migratory activity in vitro. These cells generate large numbers of oligodendroglial progeny at the early stages of maturation, both in vitro and after transplantation in models of CNS demyelination. CONCLUSIONS/SIGNIFICANCE We describe a reliable method to generate large numbers of oligodendrocytes from a renewable source of somatic, non-immortalized NSC from the human foetal brain. We also provide insights on the mechanisms underlying the pro-oligodendrogenic effect of the treatments in vitro and discuss potential issues responsible for the limited myelinating capacity shown by hNSC-derived oligodendrocytes in vivo.
Collapse
Affiliation(s)
- Margherita Neri
- San Raffaele Scientific Institute, San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Milano, Italy
- Vita-Salute San Raffaele University, Milano, Italy
| | - Claudio Maderna
- San Raffaele Scientific Institute, San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Milano, Italy
| | - Daniela Ferrari
- Bioscience and Biotechnology Department, University of Milano-Bicocca, Milano, Italy
| | - Chiara Cavazzin
- San Raffaele Scientific Institute, San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Milano, Italy
| | - Angelo L. Vescovi
- Bioscience and Biotechnology Department, University of Milano-Bicocca, Milano, Italy
| | - Angela Gritti
- San Raffaele Scientific Institute, San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Milano, Italy
| |
Collapse
|
322
|
Kelsen J, Larsen MH, Sørensen JC, Møller A, Frøkiaer J, Nielsen S, Nyengaard JR, Mikkelsen JD, Rønn LCB. Neuronal precursor cell proliferation in the hippocampus after transient cerebral ischemia: a comparative study of two rat strains using stereological tools. EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2010; 2:8. [PMID: 20370927 PMCID: PMC2868803 DOI: 10.1186/2040-7378-2-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2009] [Accepted: 04/06/2010] [Indexed: 01/19/2023]
Abstract
Background We are currently investigating microglial activation and neuronal precursor cell (NPC) proliferation after transient middle cerebral artery occlusion (tMCAo) in rats. This study aimed: (1) to investigate differences in hippocampal NPC proliferation in outbred male spontaneously hypertensive rats (SHRs) and Sprague-Dawley rats (SDs) one week after tMCAo; (2) to present the practical use of the optical fractionator and 2D nucleator in stereological brain tissue analyses; and (3) to report our experiences with an intraluminal tMCAo model where the occluding filament is advanced 22 mm beyond the carotid bifurcation and the common carotid artery is clamped during tMCAo. Methods Twenty-three SDs and twenty SHRs were randomized into four groups subjected to 90 minutes tMCAo or sham. BrdU (50 mg/kg) was administered intraperitoneally twice daily on Day 4 to 7 after surgery. On Day 8 all animals were euthanized. NeuN-stained tissue sections were used for brain and infarct volume estimation with the 2D nucleator and Cavalieri principle. Brains were studied for the presence of activated microglia (ED-1) and hippocampal BrdU incorporation using the optical fractionator. Results We found no significant difference or increase in post-ischemic NPC proliferation between the two strains. However, the response to remote ischemia may differ between SDs and SHRs. In three animals increased post-stroke NPC proliferation was associated with hippocampal ischemic injury. The mean infarct volume was 89.2 ± 76.1 mm3 in SHRs and 16.9 ± 22.7 mm3 in SDs (p < 0.005). Eight out of eleven SHRs had ischemic neocortical damage in contrast to only one out of 12 SDs. We observed involvement of the anterior choroidal and hypothalamic arteries in several animals from both strains and the anterior cerebral artery in two SHRs. Conclusions We found no evidence of an early hippocampal NPC proliferation one week after tMCAo in both strains. Infarction within the anterior choroidal artery could induce hippocampal ischemia and increase NPC proliferation profoundly. NPC proliferation was not aggravated by the presence of activated microglia. Intraluminal tMCAo in SHRs gave a more reliable infarct with neocortical involvement, but affected territories supplied by the anterior cerebral, anterior choroidal and hypothalamic arteries.
Collapse
Affiliation(s)
- Jesper Kelsen
- The Water and Salt Research Centre, University of Aarhus, DK-8000 Aarhus C, Denmark.
| | | | | | | | | | | | | | | | | |
Collapse
|
323
|
Jin K, Mao X, Xie L, Galvan V, Lai B, Wang Y, Gorostiza O, Wang X, Greenberg DA. Transplantation of human neural precursor cells in Matrigel scaffolding improves outcome from focal cerebral ischemia after delayed postischemic treatment in rats. J Cereb Blood Flow Metab 2010; 30:534-44. [PMID: 19826433 PMCID: PMC2831107 DOI: 10.1038/jcbfm.2009.219] [Citation(s) in RCA: 140] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Transplantation of neural cells is a potential approach for stroke treatment, but disruption of tissue architecture may limit transplant efficacy. One strategy for enhancing the ability of transplants to restore brain structure and function is to administer cells together with biomaterial scaffolding. We electrocoagulated the distal middle cerebral artery in adult rats and, 3 weeks later, injected one of the following into the infarct cavity: artificial cerebrospinal fluid, Matrigel scaffolding, human embryonic stem cell-derived neuronal precursor cells, scaffolding plus cells, or cells cultured in and administered together with scaffolding. Five weeks after transplantation, the latter two groups showed approximately 50% and approximately 60% reductions, respectively, in infarct cavity volume. Rats given cells cultured in and administered together with scaffolding also showed (1) survival and neuronal differentiation of transplanted cells shown by immunostaining for neuronal marker proteins and cleaved caspase-3, and by patch-clamp recording, 8 weeks after transplantation and (2) improved outcome on tests of sensorimotor and cognitive functions, 4 to 9 weeks after transplantation. These results indicate that transplantation of human neural cells together with biomaterial scaffolding has the potential to improve the outcome from stroke, even when treatment is delayed for several weeks after the ischemic event.
Collapse
Affiliation(s)
- Kunlin Jin
- Buck Institute for Age Research, Novato, California 94945, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
324
|
Abstract
Although initially considered as primarily a cell-replacement strategy, researchers today attribute a combination of both cellular repair and trophic support from transplanted cells as mediators of recovery.
Collapse
|
325
|
Huang H, Chen L, Sanberg P. Cell Therapy From Bench to Bedside Translation in CNS Neurorestoratology Era. CELL MEDICINE 2010; 1:15-46. [PMID: 21359168 PMCID: PMC3043378 DOI: 10.3727/215517910x516673] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recent advances in cell biology, neural injury and repair, and the progress towards development of neurorestorative interventions are the basis for increased optimism. Based on the complexity of the processes of demyelination and remyelination, degeneration and regeneration, damage and repair, functional loss and recovery, it would be expected that effective therapeutic approaches will require a combination of strategies encompassing neuroplasticity, immunomodulation, neuroprotection, neurorepair, neuroreplacement, and neuromodulation. Cell-based restorative treatment has become a new trend, and increasing data worldwide have strongly proven that it has a pivotal therapeutic value in CNS disease. Moreover, functional neurorestoration has been achieved to a certain extent in the CNS clinically. Up to now, the cells successfully used in preclinical experiments and/or clinical trial/treatment include fetal/embryonic brain and spinal cord tissue, stem cells (embryonic stem cells, neural stem/progenitor cells, hematopoietic stem cells, adipose-derived adult stem/precursor cells, skin-derived precursor, induced pluripotent stem cells), glial cells (Schwann cells, oligodendrocyte, olfactory ensheathing cells, astrocytes, microglia, tanycytes), neuronal cells (various phenotypic neurons and Purkinje cells), mesenchymal stromal cells originating from bone marrow, umbilical cord, and umbilical cord blood, epithelial cells derived from the layer of retina and amnion, menstrual blood-derived stem cells, Sertoli cells, and active macrophages, etc. Proof-of-concept indicates that we have now entered a new era in neurorestoratology.
Collapse
Affiliation(s)
- Hongyun Huang
- *Center for Neurorestoratology, Beijing Rehabilitation Center, Beijing, P.R. China
| | - Lin Chen
- *Center for Neurorestoratology, Beijing Rehabilitation Center, Beijing, P.R. China
| | - Paul Sanberg
- †Department of Neurosurgery, University of South Florida, Tampa, FL, USA
| |
Collapse
|
326
|
Urbaniak Hunter K, Yarbrough C, Ciacci J. Stem cells in the treatment of stroke. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 671:105-16. [PMID: 20455499 DOI: 10.1007/978-1-4419-5819-8_9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Stroke is an often devastating insult resulting in neurological deficit lasting greater than 24 hours. In the United States, stroke is the third leading cause of death. In those who do not succumb, any outcome from total recovery over a period of weeks to months to persistent profound neurological deficits is possible. Present treatment centers on the decision to administer tissue plasminogen activator, subsequent medical stabilization and early intervention with rehabilitation and risk factor management. The advent of stem cell therapy presents an exciting new frontier for research in stroke treatment, with the potential to cause a paradigm shift from symptomatic control and secondary prevention to reconstitution of neural networks and prevention of neuronal cell death after neurologic injury.
Collapse
Affiliation(s)
- Klaudia Urbaniak Hunter
- University of Michigan, Department of Radiation Oncology, UH B2C490, 1500 E. Medical Center Dr., Ann Arbor, Michigan, USA.
| | | | | |
Collapse
|
327
|
Hazama Y, S. Kurokawa M, Chiba S, Tadokoro M, Imai T, Kondo Y, Nakatsuji N, Suzuki T, Hashimoto T, Suzuki N. SDF1/CXCR4 contributes to neural regeneration in hemiplegic mice with a monkey ES-cell-derived neural graft. Inflamm Regen 2010. [DOI: 10.2492/inflammregen.30.193] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
328
|
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
|
329
|
Bentz K, Molcanyi M, Schneider A, Riess P, Maegele M, Bosche B, Hampl JA, Hescheler J, Patz S, Schäfer U. Extract Derived from Rat Brains in the Acute Phase Following Traumatic Brain Injury Impairs Survival of Undifferentiated Stem Cells and Induces Rapid Differentiation of Surviving Cells. Cell Physiol Biochem 2010; 26:821-30. [DOI: 10.1159/000323991] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2010] [Indexed: 01/19/2023] Open
|
330
|
Exploitation of Genetically Modified Neural Stem Cells for Neurological Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 671:74-92. [DOI: 10.1007/978-1-4419-5819-8_7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
331
|
Two-component protein-engineered physical hydrogels for cell encapsulation. Proc Natl Acad Sci U S A 2009; 106:22067-72. [PMID: 20007785 DOI: 10.1073/pnas.0904851106] [Citation(s) in RCA: 236] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Current protocols to encapsulate cells within physical hydrogels require substantial changes in environmental conditions (pH, temperature, or ionic strength) to initiate gelation. These conditions can be detrimental to cells and are often difficult to reproduce, therefore complicating their use in clinical settings. We report the development of a two-component, molecular-recognition gelation strategy that enables cell encapsulation without environmental triggers. Instead, the two components, which contain multiple repeats of WW and proline-rich peptide domains, undergo a sol-gel phase transition upon simple mixing and hetero-assembly of the peptide domains. We term these materials mixing-induced, two-component hydrogels. Our results demonstrate use of the WW and proline-rich domains in protein-engineered materials and expand the library of peptides successfully designed into engineered proteins. Because both of these association domains are normally found intracellularly, their molecular recognition is not disrupted by the presence of additional biomolecules in the extracellular milieu, thereby enabling reproducible encapsulation of multiple cell types, including PC-12 neuronal-like cells, human umbilical vein endothelial cells, and murine adult neural stem cells. Precise variations in the molecular-level design of the two components including (i) the frequency of repeated association domains per chain and (ii) the association energy between domains enable tailoring of the hydrogel viscoelasticity to achieve plateau shear moduli ranging from approximately 9 to 50 Pa. Because of the transient physical crosslinks that form between association domains, these hydrogels are shear-thinning, injectable, and self-healing. Neural stem cells encapsulated in the hydrogels form stable three-dimensional cultures that continue to self-renew, differentiate, and sprout extended neurites.
Collapse
|
332
|
Das S, Ghosh D, Basu A. Japanese encephalitis virus induce immuno-competency in neural stem/progenitor cells. PLoS One 2009; 4:e8134. [PMID: 19956550 PMCID: PMC2780913 DOI: 10.1371/journal.pone.0008134] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Accepted: 11/09/2009] [Indexed: 01/19/2023] Open
Abstract
Background The low immunogenicity of neural stem/progenitor cells (NSPCs) coupled with negligible expression of MHC antigens has popularized their use in transplantation medicine. However, in an inflammatory environment, the NSPCs express costimulatory molecules and MHC antigens, and also exhibit certain immunomodulatory functions. Since NSPCs are the cellular targets in a number of virus infections both during postnatal and adult stages, we wanted to investigate the immunological properties of these stem cells in response to viral pathogen. Methodology/Principal Findings We utilized both in vivo mouse model and in vitro neurosphere model of Japanese encephalitis virus (JEV) infection for the study. The NSPCs residing in the subventricular zone of the infected brains showed prominent expression of MHC-I and costimulatory molecules CD40, CD80, and CD86. Using Flow cytometry and fluorescence microscopy, we observed increased surface expression of co-stimulatory molecule and MHC class I antigen in NSPCs upon progressive JEV infection in vitro. Moreover, significant production of pro-inflammatory cyto/chemokines was detected in JEV infected NSPCs by Cytokine Bead Array analysis. Interestingly, NSPCs were capable of providing functional costimulation to allogenic T cells and JEV infection resulted in increased proliferation of allogenic T cells, as detected by Mixed Lymphocyte reaction and CFSE experiments. We also report IL-2 production by NSPCs upon JEV infection, which possibly provides mitogenic signals to T cells and trigger their proliferation. Conclusion/Significance The in vivo and in vitro findings clearly indicate the development of immunogenicity in NSPCs following progressive JEV infection, in our case, JEV infection. Following a neurotropic virus infection, NSPCs possibly behave as immunogenic cells and contribute to both the innate and adaptive immune axes. The newly discovered immunological properties of NSPCs may have implications in assigning a new role of these cells as non-professional antigen presenting cells in the central nervous system.
Collapse
Affiliation(s)
- Sulagna Das
- National Brain Research Centre, Manesar, Haryana, India
| | | | - Anirban Basu
- National Brain Research Centre, Manesar, Haryana, India
- * E-mail:
| |
Collapse
|
333
|
Abstract
The B05 transgenic SCA1 mice, expressing human ataxin-1 with an expanded polyglutamine tract in cerebellar Purkinje cells (PCs), recapitulate many pathological and behavioral characteristics of the neurodegenerative disease spinocerebellar ataxia type 1 (SCA1), including progressive ataxia and PC loss. We transplanted neural precursor cells (NPCs) derived from the subventricular zone of GFP-expressing adult mice into the cerebellar white matter of SCA1 mice when they showed absent (5 weeks), initial (13 weeks), and significant (24 weeks) PC loss. Only in mice with significant cell loss, grafted NPCs migrated into the cerebellar cortex. These animals showed improved motor skills compared with sham-treated controls. No grafted cell adopted the morphological and immunohistochemical characteristics of PCs, but the cerebellar cortex in NPC-grafted SCA1 mice had a significantly thicker molecular layer and more surviving PCs. Perforated patch-clamp recordings revealed a normalization of the PC basal membrane potential, which was abnormally depolarized in sham-treated animals. No significant increase in levels of several neurotrophic factors was observed, suggesting, along with morphological observation, that the neuroprotective effect of grafted NPCs was mediated by direct contact with the host PCs. We postulate that a similar neuroprotective effect of NPCs may be applicable to other cerebellar degenerative diseases.
Collapse
|
334
|
Tamaki SJ, Jacobs Y, Dohse M, Capela A, Cooper JD, Reitsma M, He D, Tushinski R, Belichenko PV, Salehi A, Mobley W, Gage FH, Huhn S, Tsukamoto AS, Weissman IL, Uchida N. Neuroprotection of host cells by human central nervous system stem cells in a mouse model of infantile neuronal ceroid lipofuscinosis. Cell Stem Cell 2009; 5:310-9. [PMID: 19733542 DOI: 10.1016/j.stem.2009.05.022] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Revised: 09/23/2008] [Accepted: 05/20/2009] [Indexed: 12/16/2022]
Abstract
Infantile neuronal ceroid lipofuscinosis (INCL) is a fatal neurodegenerative disease caused by a deficiency in the lysosomal enzyme palmitoyl protein thioesterase-1 (PPT1). Ppt1 knockout mice display hallmarks of INCL and mimic the human pathology: accumulation of lipofuscin, degeneration of CNS neurons, and a shortened life span. Purified non-genetically modified human CNS stem cells, grown as neurospheres (hCNS-SCns), were transplanted into the brains of immunodeficient Ppt1(-/)(-) mice where they engrafted robustly, migrated extensively, and produced sufficient levels of PPT1 to alter host neuropathology. Grafted mice displayed reduced autofluorescent lipofuscin, significant neuroprotection of host hippocampal and cortical neurons, and delayed loss of motor coordination. Early intervention with cellular transplants of hCNS-SCns into the brains of INCL patients may supply a continuous and long-lasting source of the missing PPT1 and provide some therapeutic benefit through protection of endogenous neurons. These data provide the experimental basis for human clinical trials with these banked hCNS-SCns.
Collapse
|
335
|
Hosking MP, Lane TE. The Biology of Persistent Infection: Inflammation and Demyelination following Murine Coronavirus Infection of the Central Nervous System. ACTA ACUST UNITED AC 2009; 5:267-276. [PMID: 19946572 DOI: 10.2174/157339509789504005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Multiple Sclerosis (MS) is an immune-mediated demyelinating disease of humans. Although causes of MS are enigmatic, underlying elements contributing to disease development include both genetic and environmental factors. Recent epidemiological evidence has pointed to viral infection as a trigger to initiating white matter damage in humans. Mouse hepatitis virus (MHV) is a positive strand RNA virus that, following intracranial infection of susceptible mice, induces an acute encephalomyelitis that later resolves into a chronic fulminating demyelinating disease. Immune cell infiltration into the central nervous system is critical both to quell viral replication and instigate demyelination. Recent efforts by our laboratory and others have focused upon strategies capable of enhancing remyelination in response to viral-induced demyelination, both by dampening chronic inflammation and by surgical engraftment of remyelination - competent neural precursor cells.
Collapse
Affiliation(s)
- Martin P Hosking
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697-3900 USA
| | | |
Collapse
|
336
|
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
|
337
|
Horiuchi M, Mogi M, Iwai M. The angiotensin II type 2 receptor in the brain. J Renin Angiotensin Aldosterone Syst 2009; 11:1-6. [PMID: 19861353 DOI: 10.1177/1470320309347793] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Recent clinical studies indicate that blockade of the renin-angiotensin system is important to prevent stroke, and accumulating results of basic research also indicate the possible involvement of the central renin-angiotensin system in ischaemic brain damage and cognition. When the angiotensin II type 1 receptor is blocked by an angiotensin type 1 receptor blocker, unbound angiotensin II acts preferentially on the angiotensin II type 2 (AT(2)) receptor. These results suggest the pathophysiological importance of the AT(2) receptor in the clinical use of angiotensin type 1 receptor blockers, which are widely used in patients with hypertension with the expectation of a decrease in the onset of cardiovascular and cerebrovascular disease. We review here the possible roles of AT(2) receptor activation in the brain, focusing on ischaemic stroke, cognitive function and neurogenesis, and potential effects of specific AT(2) receptor agonists.
Collapse
Affiliation(s)
- Masatsugu Horiuchi
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Shitsukawa, Tohon, Ehime 791-0295, Japan.
| | | | | |
Collapse
|
338
|
Madhavan L, Collier TJ. A synergistic approach for neural repair: cell transplantation and induction of endogenous precursor cell activity. Neuropharmacology 2009; 58:835-44. [PMID: 19853620 DOI: 10.1016/j.neuropharm.2009.10.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Revised: 10/12/2009] [Accepted: 10/15/2009] [Indexed: 12/11/2022]
Abstract
Stem cell research offers enormous potential for treating many diseases of the nervous system. At present, therapeutic strategies in stem cell research segregate into two approaches: cell transplantation or endogenous cell stimulation. Realistically, future cell therapies will most likely involve a combination of these two approaches, a theme of our current research. Here, we propose that there exists a 'synergy' between exogenous (transplanted) and endogenous stem cell actions that can be utilized to achieve therapeutic ends. Elucidating mechanisms underlying this exogenous-endogenous stem cell synergism may lead to the development of optimal cell therapies for neural disorders.
Collapse
Affiliation(s)
- Lalitha Madhavan
- Department of Neurology, University of Cincinnati, Cincinnati, OH 45267, USA.
| | | |
Collapse
|
339
|
Optimizing the success of cell transplantation therapy for stroke. Neurobiol Dis 2009; 37:275-83. [PMID: 19822211 DOI: 10.1016/j.nbd.2009.10.003] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Revised: 09/30/2009] [Accepted: 10/02/2009] [Indexed: 12/22/2022] Open
Abstract
Stem cell transplantation has evolved as a promising experimental treatment approach for stroke. In this review, we address the major hurdles for successful translation from basic research into clinical applications and discuss possible strategies to overcome these issues. We summarize the results from present pre-clinical and clinical studies and focus on specific areas of current controversy and research: (i) the therapeutic time window for cell transplantation; (ii) the selection of patients likely to benefit from such a therapy; (iii) the optimal route of cell delivery to the ischemic brain; (iv) the most suitable cell types and sources; (v) the potential mechanisms of functional recovery after cell transplantation; and (vi) the development of imaging techniques to monitor cell therapy.
Collapse
|
340
|
Kögler G, Critser P, Trapp T, Yoder M. Future of cord blood for non-oncology uses. Bone Marrow Transplant 2009; 44:683-97. [PMID: 19802027 DOI: 10.1038/bmt.2009.287] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
For the last 5 years cord blood (CB) has been under intense experimental investigation in in vitro differentiation models and in preclinical animal models ranging from bone to muscle regeneration, cardiovascular diseases including myocardial and peripheral arterial disease, stroke and Parkinson's disease. On the basis of its biological advantages, CB can be an ideal source for tissue regeneration. However, in the hype of the so-called 'plasticity', many cell types have been characterized either on cell surface Ag expression alone or by RNA expression only, and without detailed characterization of genetic pathways; frequently, cells are defined without analysis of cellular function in vitro and in vivo, and the definition of the lineage of origin and cells have not been defined in preclinical studies. Here, we explore not only the most consistent data with regard to differentiation of CB cells in vitro and in vivo, but also show technical limitations, such as why in contrast to cell populations isolated from fresh CB, cryopreserved CB is not the ideal source for tissue regeneration. By taking advantage of numerous CB units discarded due to lack of sufficient hematopoietic cells for clinical transplantation, new concepts to produce off-the-shelf products are presented as well.
Collapse
Affiliation(s)
- G Kögler
- Institute for Transplantation Diagnostics and Cell Therapeutics, University of Duesseldorf Medical School, Duesseldorf, Germany.
| | | | | | | |
Collapse
|
341
|
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
|
342
|
Maciaczyk J, Singec I, Maciaczyk D, Klein A, Nikkhah G. Restricted Spontaneous In Vitro Differentiation and Region-Specific Migration of Long-Term Expanded Fetal Human Neural Precursor Cells After Transplantation Into the Adult Rat Brain. Stem Cells Dev 2009; 18:1043-58. [DOI: 10.1089/scd.2008.0346] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Affiliation(s)
- Jaroslaw Maciaczyk
- Laboratory of Molecular Neurosurgery, Department of Stereotactic and Functional Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany
- Department of General Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany
| | - Ilyas Singec
- Burnham Institute for Medical Research, Stem Cell and Regeneration Program, La Jolla, California
| | - Donata Maciaczyk
- Laboratory of Molecular Neurosurgery, Department of Stereotactic and Functional Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany
| | - Alexander Klein
- The Brain Research Group, School of Biosciences, University of Cardiff, Cardiff, United Kingdom
| | - Guido Nikkhah
- Laboratory of Molecular Neurosurgery, Department of Stereotactic and Functional Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany
| |
Collapse
|
343
|
Morphofunctional study of the therapeutic efficacy of human mesenchymal and neural stem cells in rats with diffuse brain injury. Bull Exp Biol Med 2009; 147:132-46. [PMID: 19526149 DOI: 10.1007/s10517-009-0432-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
We studied the effect of transplantation of human stem cells from various tissues on reparative processes in the brain of rats with closed craniocerebral injury. Combined treatment with standard drugs and systemic administration of xenogeneic stem cells had a neuroprotective effect. The morphology of neurons rapidly returned to normal after administration of fetal neural stem cells. Fetal mesenchymal stem cells produced a prolonged effect on proliferative activity of progenitor cells in the subventricular zone of neurogenesis. Adult mesenchymal stem cells had a strong effect on recovery of the vascular bed in ischemic regions.
Collapse
|
344
|
Cheshier SH, Kalani MYS, Lim M, Ailles L, Huhn SL, Weissman IL. A NEUROSURGEON'S GUIDE TO STEM CELLS, CANCER STEM CELLS, AND BRAIN TUMOR STEM CELLS. Neurosurgery 2009; 65:237-49; discussion 249-50; quiz N6. [DOI: 10.1227/01.neu.0000349921.14519.2a] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Samuel H. Cheshier
- Stanford Institute of Stem Cell Biology and Regenerative Medicine, Departments of Neurosurgery and Developmental Biology, Stanford University School of Medicine, Stanford, California
| | - M. Yashar S. Kalani
- Stanford Institute of Stem Cell Biology and Regenerative Medicine, Departments of Neurosurgery and Developmental Biology, Stanford University School of Medicine, Stanford, California
| | - Michael Lim
- Department of Neurosurgery, The Johns Hopkins Hospital, Baltimore, Maryland
| | - Laurie Ailles
- Stanford Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Steven L. Huhn
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, Stem Cells, Inc., Palo Alto, California
| | - Irving L. Weissman
- Stanford Institute of Stem Cell Biology and Regenerative Medicine, Department of Developmental Biology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
345
|
Burns TC, Verfaillie CM, Low WC. Stem cells for ischemic brain injury: a critical review. J Comp Neurol 2009; 515:125-44. [PMID: 19399885 DOI: 10.1002/cne.22038] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
No effective therapy is currently available to promote recovery following ischemic stroke. Stem cells have been proposed as a potential source of new cells to replace those lost due to central nervous system injury, as well as a source of trophic molecules to minimize damage and promote recovery. We undertook a detailed review of data from recent basic science and preclinical studies to investigate the potential application of endogenous and exogenous stem cell therapies for treatment of cerebral ischemia. To date, spontaneous endogenous neurogenesis has been observed in response to ischemic injury, and can be enhanced via infusion of appropriate cytokines. Exogenous stem cells from multiple sources can generate neural cells that survive and form synaptic connections after transplantation in the stroke-injured brain. Stem cells from multiple sources cells also exhibit neuroprotective properties that may ameliorate stroke deficits. In many cases, functional benefits observed are likely independent of neural differentiation, although the exact mechanisms remain poorly understood. Future studies of neuroregeneration will require the demonstration of function in endogenously born neurons following focal ischemia. Further, methods are currently lacking to demonstrate definitively the therapeutic effect of newly introduced neural cells. Increased plasticity following stroke may facilitate the functional integration of new neurons, but the loss of appropriate guidance cues and supporting architecture in the infarct cavity will likely impede the restoration of lost circuitry. Thus careful investigation of the mechanisms underlying trophic benefits will be essential. Evidence to date suggests that continued development of stem cell therapies may ultimately lead to viable treatment options for ischemic brain injury.
Collapse
Affiliation(s)
- Terry C Burns
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | | | | |
Collapse
|
346
|
Varghese M, Olstorn H, Berg-Johnsen J, Moe MC, Murrell W, Langmoen IA. Isolation of human multipotent neural progenitors from adult filum terminale. Stem Cells Dev 2009; 18:603-13. [PMID: 18652547 DOI: 10.1089/scd.2008.0144] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Stem cells have been isolated from several CNS regions, including the spinal cord. However, the terminal end of the spinal cord, filum terminale, has been referred to as a fibrovascular tag without neurogenic potential and of no clinical significance. Recently, we were fortunate to acquire some samples of this tissue. We show for the first time that progenitor cells exhibiting the hallmarks of stem cells can be isolated from adult human filum terminale (FTNPs). More specifically, FTNPs self-renew and proliferate to form neurospheres, and exhibit tripotent differentiation into neurons, astrocytes, and oligodendrocytes. Equally important, FTNPs develop the electrophysiological profile of neurons and glia. Whole-cell patch-clamp recordings show beta-III-tubulin(+) neurons exhibiting overshooting action potentials, displaying both the fast inactivating TTX-sensitive sodium current as well as 4-AP and TEA sensitive potassium currents. To assess potency in vivo, FTNPs were transplanted into the posterior periventricular region of control or ischemic rat brains. Despite a vigorous immune response against the xenograft, FTNPs survived and were found not only in the graft area but had also migrated to the lesioned CA1 region. Notwithstanding the immune response, FTNPs differentiated into astrocytes, but no neuronal differentiation was observed in the transplant milieu tested. However, neuronal differentiation in vivo cannot be ruled out and assessment of the conditions necessary to promote neurogenesis in vivo requires more research. Significantly, no tumor formation or aberrant cell morphology was seen in or adjacent to the graft area. Thus, filum terminale provides a novel source of adult human neural progenitor cells that develop into functional neurons with possible clinical applications.
Collapse
Affiliation(s)
- Mercy Varghese
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research, University of Oslo, Oslo, Norway.
| | | | | | | | | | | |
Collapse
|
347
|
van der Meulen AAE, Biber K, Lukovac S, Balasubramaniyan V, den Dunnen WFA, Boddeke HWGM, Mooij JJA. The role of CXC chemokine ligand (CXCL)12-CXC chemokine receptor (CXCR)4 signalling in the migration of neural stem cells towards a brain tumour. Neuropathol Appl Neurobiol 2009; 35:579-91. [PMID: 19627512 DOI: 10.1111/j.1365-2990.2009.01036.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
AIMS It has been shown that neural stem cells (NSCs) migrate towards areas of brain injury or brain tumours and that NSCs have the capacity to track infiltrating tumour cells. The possible mechanism behind the migratory behaviour of NSCs is not yet completely understood. As chemokines are involved in the migration of immune cells in the injured brain, they may also be involved in chemoattraction of NSCs towards a brain tumour. METHODS The expression profile of various chemokine receptors in NSCs, harvested from the subventricular zone of adult mice, was investigated by reverse transcriptase- polymerase chain reaction analysis. Furthermore, the functionality of the chemokine receptors was assessed in in vitro chemotaxis assays and calcium signalling experiments. To test the in vivo migration of NSCs, a syngeneic mouse model was developed, whereby a B16F10 melanoma cell line was grafted into one hemisphere and later NSCs were grafted in the contralateral hemisphere. Furthermore, the expression of chemokines in this melanoma cell line was investigated. RESULTS AND CONCLUSIONS Adult mouse NSCs functionally express various chemokine receptors of which CXC chemokine receptor (CXCR)4 shows the highest mRNA levels and most pronounced functional responses in vitro. CXC chemokine ligand (CXCL)12, the ligand for CXCR4, is expressed by the melanoma cell line. In this mouse model for metastatic brain tumours, it is shown that NSCs express CXCR4 at their cell membranes while they migrate towards the tumour, which produces CXCL12. It is therefore suggested that the CXCR4/CXCL12 pathway plays a role in the mechanism underlying tumour-mediated attraction of NSCs.
Collapse
Affiliation(s)
- A A E van der Meulen
- Departments of Neuroscience, University Medical Center Groningen, the Netherlands.
| | | | | | | | | | | | | |
Collapse
|
348
|
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
|
349
|
Human neural stem cells overexpressing glial cell line-derived neurotrophic factor in experimental cerebral hemorrhage. Gene Ther 2009; 16:1066-76. [PMID: 19554035 DOI: 10.1038/gt.2009.51] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Recent studies have reported that glial cell line-derived growth factor (GDNF) has neurotrophic effects on the central nervous system, and the neural stem cells (NSCs) engrafted in animal models of stroke survive and ameliorate the neurological deficits. In this study, a stable human NSC line overexpressing GDNF (F3.GDNF) was transplanted next to the intracerebral hemorrhage (ICH) lesion site and a possible therapeutic effect was investigated. F3.GDNF human NSC line was transplanted into the cortex overlying the striatal ICH lesion. ICH was induced in adult mice by the unilateral injection of bacterial collagenase into the striatum. The animals were evaluated for 8 weeks with rotarod and limb placement tests. Transplanted NSCs were detected by beta-gal immunostaining with double labeling of neurofilament, microtubule associated protein-2, glial fibrillary acidic protein or human nuclear matrix antigen (HuNuMA). F3.GDNF human NSCs produced a four times higher amount of GDNF over parental F3 cells in vitro, induced behavioral improvement in ICH mice after brain transplantation and two- to threefold increase in cell survival of transplanted NSCs at 2 and 8 weeks post-transplantation. In F3.GDNF-grafted ICH brain, a significant increase in the antiapoptotic protein and cell survival signal molecules, and a marked reduction in proapoptotic proteins were found as compared with control group. Brain transplantation of human NSCs overexpressing GDNF in ICH animals provided functional recovery in ICH animals, and survival and differentiation of grafted human NSCs. These results indicate that the F3.GDNF human NSCs should be of a great value as a cellular source for the cellular therapy in animal models of human neurological disorders including ICH.
Collapse
|
350
|
Wakeman DR, Hofmann MR, Redmond DE, Teng YD, Snyder EY. Long-term multilayer adherent network (MAN) expansion, maintenance, and characterization, chemical and genetic manipulation, and transplantation of human fetal forebrain neural stem cells. ACTA ACUST UNITED AC 2009; Chapter 2:Unit2D.3. [PMID: 19455542 DOI: 10.1002/9780470151808.sc02d03s9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Human neural stem/precursor cells (hNSC/hNPC) have been targeted for application in a variety of research models and as prospective candidates for cell-based therapeutic modalities in central nervous system (CNS) disorders. To this end, the successful derivation, expansion, and sustained maintenance of undifferentiated hNSC/hNPC in vitro, as artificial expandable neurogenic micro-niches, promises a diversity of applications as well as future potential for a variety of experimental paradigms modeling early human neurogenesis, neuronal migration, and neurogenetic disorders, and could also serve as a platform for small-molecule drug screening in the CNS. Furthermore, hNPC transplants provide an alternative substrate for cellular regeneration and restoration of damaged tissue in neurodegenerative disorders such as Parkinson's disease and Alzheimer's disease. Human somatic neural stem/progenitor cells (NSC/NPC) have been derived from a variety of cadaveric sources and proven engraftable in a cytoarchitecturally appropriate manner into the developing and adult rodent and monkey brain while maintaining both functional and migratory capabilities in pathological models of disease. In the following unit, we describe a new procedure that we have successfully employed to maintain operationally defined human somatic NSC/NPC from developing fetal, pre-term post-natal, and adult cadaveric forebrain. Specifically, we outline the detailed methodology for in vitro expansion, long-term maintenance, manipulation, and transplantation of these multipotent precursors.
Collapse
Affiliation(s)
- Dustin R Wakeman
- University of California at San Diego, La Jolla, California, USA
| | | | | | | | | |
Collapse
|