101
|
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
|
102
|
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
|
103
|
Li L, Jiang Q, Ding G, Zhang L, Zhang ZG, Li Q, Panda S, Lu M, Ewing JR, Chopp M. Effects of administration route on migration and distribution of neural progenitor cells transplanted into rats with focal cerebral ischemia, an MRI study. J Cereb Blood Flow Metab 2010; 30:653-62. [PMID: 19888287 PMCID: PMC2844252 DOI: 10.1038/jcbfm.2009.238] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We tested the hypotheses that administration routes affect the migration and distribution of grafted neural progenitor cells (NPCs) in the ischemic brain and that the ischemic lesion plays a role in mediating the grafting process. Male Wistar rats (n=41) were subjected to 2-h middle cerebral artery occlusion (MCAo), followed 1 day later by administration of magnetically labeled NPCs. Rats with MCAo were assigned to one of three treatment groups targeted for cell transplantation intra-arterially (IA), intracisternally (IC), or intravenously (IV). MRI measurements consisting of T2-weighted imaging and three-dimensional (3D) gradient echo imaging were performed 24 h after MCAo, 4 h after cell injection, and once a day for 4 days. Prussian blue staining was used to identify the labeled cells, 3D MRI to detect cell migration and distribution, and T2 map to assess lesion volumes. Intra-arterial (IA) administration showed significantly increased migration, a far more diffuse distribution pattern, and a larger number of transplanted NPCs in the target brain than IC or IV administration. However, high mortality with IA delivery (IA: 41%; IC: 17%; IV: 8%) poses a serious concern for using this route of administration. Animals with smaller lesions at the time of transplantation have fewer grafted cells in the parenchyma.
Collapse
Affiliation(s)
- Lian Li
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan 48202, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
104
|
Naegele JR, Maisano X, Yang J, Royston S, Ribeiro E. Recent advancements in stem cell and gene therapies for neurological disorders and intractable epilepsy. Neuropharmacology 2010; 58:855-64. [PMID: 20146928 DOI: 10.1016/j.neuropharm.2010.01.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2009] [Revised: 01/22/2010] [Accepted: 01/26/2010] [Indexed: 12/11/2022]
Abstract
The potential applications of stem cell therapies for treating neurological disorders are enormous. Many laboratories are focusing on stem cell treatments for CNS diseases, including spinal cord injury, Amyotrophic lateral sclerosis, Parkinson's disease, Huntington's disease, multiple sclerosis, stroke, traumatic brain injury, and epilepsy. Among the many stem cell types under testing for neurological treatments, the most common are fetal and adult brain stem cells, embryonic stem cells, induced pluripotent stem cells, and mesenchymal stem cells. An expanding toolbox of molecular probes is now available to allow analyses of neural stem cell fates prior to and after transplantation. Concomitantly, protocols are being developed to direct the fates of stem cell-derived neural progenitors, and also to screen stem cells for tumorigenicity and aneuploidy. The rapid progress in the field suggests that novel stem cell and gene therapies for neurological disorders are in the pipeline.
Collapse
Affiliation(s)
- Janice R Naegele
- Department of Biology and Program in Neuroscience and Behavior, Hall Atwater Laboratory, 52 Lawn Avenue, Wesleyan University, Middletown, CT 06459, USA.
| | | | | | | | | |
Collapse
|
105
|
Buddensiek J, Dressel A, Kowalski M, Storch A, Sabolek M. Adult cerebrospinal fluid inhibits neurogenesis but facilitates gliogenesis from fetal rat neural stem cells. J Neurosci Res 2009; 87:3054-66. [PMID: 19530161 DOI: 10.1002/jnr.22150] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neural stem cells (NSCs) are a promising source for cell replacement therapies for neurological diseases. Administration of NSCs into the cerebrospinal fluid (CSF) offers a nontraumatic transplantation method into the brain. However, cell survival and intraparenchymal migration of the transplants are limited. Furthermore, CSF was recently reported to be an important milieu for controlling stem cell processes in the brain. We studied the effects of adult human leptomeningeal CSF on the behavior of fetal rat NSCs. CSF increased survival of NSCs compared with standard culture media during stem cell maintenance and differentiation. The presence of CSF enhanced NSC differentiation, leading to a faster loss of self-renewal capacity and faster and stronger neurite outgrowth. Some of these effects (mainly cell survival, neurite brancing) were blocked by addition of the bone morphogenic protein (BMP) inhibitor noggin. After differentiation in CSF, significantly fewer MAP2ab(+) neurons were found, but there were more GFAP(+) astroglia compared with standard media. By RT-PCR analysis, we determined a decrease of mRNA of the NSC marker gene Nestin but an increase of Gfap mRNA during differentiation up to 72 hr in CSF compared with standard media. Our data demonstrate that adult human leptomeningeal CSF enhances cell survival of fetal rat NSCs during proliferation and differentiation. Furthermore, CSF provides a stimulus for gliogenesis but inhibits neurogenesis from fetal NSCs. Our data suggest that CSF contains factors such as BMPs regulating NSC behavior, and we hypothesize that fast differentiation of NSCs in CSF leads to a rapid loss of migration capacity of intrathecally transplanted NSCs.
Collapse
Affiliation(s)
- Judith Buddensiek
- Department of Neurology, EMA University of Greifswald, Greifswald, Germany
| | | | | | | | | |
Collapse
|
106
|
Wu W, Chen X, Hu C, Li J, Yu Z, Cai W. Transplantation of neural stem cells expressing hypoxia-inducible factor-1alpha (HIF-1alpha) improves behavioral recovery in a rat stroke model. J Clin Neurosci 2009; 17:92-5. [PMID: 19913430 DOI: 10.1016/j.jocn.2009.03.039] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2008] [Revised: 03/28/2009] [Accepted: 03/30/2009] [Indexed: 11/18/2022]
Abstract
We explored the possibility that hypoxia-inducible factor-1alpha (HIF-1alpha) might contribute to the therapeutic effect of neural stem cell (NSC) transplantation in cerebral ischemia. The relative efficacy of modified NSC to promote behavioral recovery was investigated in a rat model of stroke induced by a transient middle cerebral artery occlusion (MCAO). A recombinant adenovirus (Ad-HIF-1alpha) was engineered to express HIF-1alpha. Control NSC infected with control adenovirus (NSC-Ad), recombinant adenovirus Ad-HIF-1alpha, or NSC infected by Ad-HIF-1alpha (NSC-Ad-HIF-1alpha), were used for intraventricular transplantion into rat brain 24 hours after MCAO. Neurological deficits were assessed over 4 weeks using the modified neurological severity scale (NSS) score. Long-term in vivo expression of HIF-1alpha was demonstrated by Western blotting and immunocytochemistry, and derivatives of nestin-positive transplanted cells contributed to both neuronal (neurofilament-positive) and astroglial (glial fibrillary acidic protein-positive) lineages. All animals showed functional improvement. Improvement was accelerated in animals receiving either NSC-Ad or Ad-HIF-1alpha, while improvement at all times between 7 days and 28 days post MCAO was significantly greater in animals transplanted with NSC-Ad-HIF-1alpha than for other treated animals. NSC-Ad-HIF-1alpha cells also increased the number of factor VIII-positive cells in the region of ischemic injury, indicating that HIF-1alpha expression can promote angiogenesis. Gene-modified NSC expressing HIF-1alpha have therapeutic potential in ischemic stroke.
Collapse
Affiliation(s)
- Wanfu Wu
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | | | | | | | | | | |
Collapse
|
107
|
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
|
108
|
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: 152] [Impact Index Per Article: 9.5] [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
|
109
|
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
|
110
|
Jung SH, Lee ST, Chu K, Park JE, Lee SU, Han TR, Kim M. Cell proliferation and synaptogenesis in the cerebellum after focal cerebral ischemia. Brain Res 2009; 1284:180-90. [DOI: 10.1016/j.brainres.2009.05.051] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Revised: 05/18/2009] [Accepted: 05/20/2009] [Indexed: 10/20/2022]
|
111
|
Abstract
Systemic neurotransplantation (SNT) was introduced in the laboratory in 2000 and currently it is being widely examined in animal models of neurological disorders. The aim of this systematic review was to evaluate the current state of knowledge in the field of experimental SNT and the premise for the introduction of clinical trials. PubMed was searched and 60 articles utilizing an SNT approach were found and subjected to analysis. The time window for cell transplantation was addressed in only two studies, with contradictory results. Immunosuppression was applied in 25% of studies. No study addressed the justification for immunosuppression. Bone marrow was the most frequent source of grafted cells, followed by cord blood and then by cells of embryonic origin. Studies investigating dose-dependency revealed no satisfactory results with transplantation of less than 10(6) cells/animal; the efficient dose most frequently ranged from 10(6)-10(7) cells/animal (mice and rats). The behavioral effects of cell transplantation were assessed in 75% of all studies; significant improvement was achieved in 95% of them. Morphological effect was evaluated in half of the studies; significant positive effect was achieved in 73% of them. Experimental attempts to elucidate the mechanisms mediating cell-dependent effect were not undertaken in half of the studies. In the other half, the most frequent mechanisms were growth factors, neurogenesis and immunomodulation. SNT still seems to be at the very initial stage of development. Many critical factors have not been sufficiently addressed in laboratory studies and they must be clarified before the introduction of clinical trials.
Collapse
Affiliation(s)
- Miroslaw Janowski
- Department of NeuroRepair, Medical Research Center, Polish Academy of Science, Warsaw, Poland.
| | | |
Collapse
|
112
|
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: 157] [Impact Index Per Article: 9.8] [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
|
113
|
Abstract
The potential application for stem cell therapy is vast, and development for use in ischaemic stroke is still in its infancy. Access to stem cells for research is contentious; however, stem cells are obtainable from both animal and human. Despite a limited understanding of their mechanisms of action, clinical trials assessing stem cells in human stroke have been performed. Trials are also underway evaluating haematopoietic precursors mobilised with granulocyte-colony stimulating factor, an approach offering an autologous means of administrating stem cells for therapeutic purposes. This review summarises current knowledge in regard to stem cells and their potential for helping improve recovery after stroke.
Collapse
Affiliation(s)
- Tim England
- Stroke Trials Unit, Institute of Neuroscience, University of Nottingham, Nottingham, UK
| | | | | |
Collapse
|
114
|
Boucherie C, Hermans E. Adult stem cell therapies for neurological disorders: benefits beyond neuronal replacement? J Neurosci Res 2009; 87:1509-21. [PMID: 19115417 DOI: 10.1002/jnr.21970] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The modest capacity of endogenous repair processes in the central nervous system (CNS) justifies the broad interest in the development of effective stem cell based therapies for neurodegenerative disorders and other acute or chronic lesions. Motivated by the ambitious expectation to achieve functional neuronal replacement, several studies have already evidenced a potential benefit of stem cell grafts in animal models of human disorders. Nevertheless, growing evidence suggests that the effects orchestrated by stem cells, in most experimental cases, are not necessarily associated with the generation of new neurons. This hypothesis correlates with the versatile properties of adult and embryonic stem cells. When introduced into the lesioned CNS, nondifferentiated stem cells can have a positive influence through intrinsic neuroprotective capacities related to the production of neurotrophic factors, stimulation of endogenous neurogenesis, and modulation of neuroinflammation. Stem cells are also endowed with a multipotent differentiation profile, suggesting that a positive outcome could result from the replacement of nonneuronal cell types, in particular astrocytes and oligodendrocytes. Focusing on adult stem cells, this Review aims at summarizing experimental observations supporting the concept that, in cell-based therapies, stem cells operate not through a unidirectional mechanism (e.g., generating neurons) but rather as cellular mediators of a multitude of biological activities that could provide a favorable outcome for diverse nervous disorders.
Collapse
Affiliation(s)
- Cédric Boucherie
- Laboratory of Experimental Pharmacology, Institute of Neurosciences (INES), Université catholique de Louvain, Brussels, Belgium
| | | |
Collapse
|
115
|
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
|
116
|
Pluchino S, Zanotti L, Brambilla E, Rovere-Querini P, Capobianco A, Alfaro-Cervello C, Salani G, Cossetti C, Borsellino G, Battistini L, Ponzoni M, Doglioni C, Garcia-Verdugo JM, Comi G, Manfredi AA, Martino G. Immune regulatory neural stem/precursor cells protect from central nervous system autoimmunity by restraining dendritic cell function. PLoS One 2009; 4:e5959. [PMID: 19543526 PMCID: PMC2694997 DOI: 10.1371/journal.pone.0005959] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Accepted: 05/22/2009] [Indexed: 12/18/2022] Open
Abstract
Background The systemic injection of neural stem/precursor cells (NPCs) provides remarkable amelioration of the clinico-pathological features of experimental autoimmune encephalomyelitis (EAE). This is dependent on the capacity of transplanted NPCs to engage concurrent mechanisms of action within specific microenvironments in vivo. Among a wide range of therapeutic actions alternative to cell replacement, neuroprotective and immune modulatory capacities of transplanted NPCs have been described. However, lacking is a detailed understanding of the mechanisms by which NPCs exert their therapeutic plasticity. This study was designed to identify the first candidate that exemplifies and sustains the immune modulatory capacity of transplanted NPCs. Methodology/Principal Findings To achieve the exclusive targeting of the peripheral immune system, SJL mice with PLP-induced EAE were injected subcutaneously with NPCs and the treatment commenced prior to disease onset. NPC-injected EAE mice showed significant clinical improvement, as compared to controls. Exogenous NPCs lacking the expression of major neural antigens were reliably (and for long-term) found at the level of draining lymph nodes, while establishing sophisticated anatomical interactions with lymph node cells. Importantly, injected NPCs were never found in organs other than lymph nodes, including the brain and the spinal cord. Draining lymph nodes from transplanted mice showed focal up-regulation of major developmental stem cell regulators, such as BMP-4, Noggin and Sonic hedgehog. In lymph nodes, injected NPCs hampered the activation of myeloid dendritic cells (DCs) and steadily restrained the expansion of antigen-specific encephalitogenic T cells. Both ex vivo and in vitro experiments identified a novel highly NPC-specific–BMP-4-dependent–mechanism hindering the DC maturation. Conclusion/Significance The study described herein, identifies the first member of the TGF β/BMP family of stem cell regulators as a novel tolerogenic factor released by NPCs. Full exploitation of this pathway as an efficient tool for vaccination therapy in autoimmune inflammatory conditions is underway.
Collapse
Affiliation(s)
- Stefano Pluchino
- Neuroimmunology Unit, San Raffaele Scientific Institute and Università Vita–Salute, Milan, Italy
- DIBIT II and Institute of Experimental Neurology (InSpe), San Raffaele Scientific Institute and Università Vita–Salute, Milan, Italy
- * E-mail: (SP); (GM)
| | - Lucia Zanotti
- Neuroimmunology Unit, San Raffaele Scientific Institute and Università Vita–Salute, Milan, Italy
- DIBIT II and Institute of Experimental Neurology (InSpe), San Raffaele Scientific Institute and Università Vita–Salute, Milan, Italy
| | - Elena Brambilla
- Neuroimmunology Unit, San Raffaele Scientific Institute and Università Vita–Salute, Milan, Italy
- DIBIT II and Institute of Experimental Neurology (InSpe), San Raffaele Scientific Institute and Università Vita–Salute, Milan, Italy
| | - Patrizia Rovere-Querini
- Clinical Immunology Unit, San Raffaele Scientific Institute and Università Vita–Salute, Milan, Italy
| | - Annalisa Capobianco
- Clinical Immunology Unit, San Raffaele Scientific Institute and Università Vita–Salute, Milan, Italy
| | - Clara Alfaro-Cervello
- Department Comparative Neurobiology, Instituto Cavanilles, University of Valencia, Valencia, Spain
| | - Giuliana Salani
- Neuroimmunology Unit, San Raffaele Scientific Institute and Università Vita–Salute, Milan, Italy
- DIBIT II and Institute of Experimental Neurology (InSpe), San Raffaele Scientific Institute and Università Vita–Salute, Milan, Italy
| | - Chiara Cossetti
- Neuroimmunology Unit, San Raffaele Scientific Institute and Università Vita–Salute, Milan, Italy
- Instituto de Ciências Biomedicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Giovanna Borsellino
- Neuroimmunology Unit, European Brain Research Institute, Santa Lucia Foundation, Rome, Italy
| | - Luca Battistini
- Neuroimmunology Unit, European Brain Research Institute, Santa Lucia Foundation, Rome, Italy
| | - Maurilio Ponzoni
- Pathology Unit, San Raffaele Scientific Institute and Università Vita–Salute, Milan, Italy
| | - Claudio Doglioni
- Pathology Unit, San Raffaele Scientific Institute and Università Vita–Salute, Milan, Italy
| | - Jose Manuel Garcia-Verdugo
- Department Comparative Neurobiology, Instituto Cavanilles, University of Valencia, Valencia, Spain
- Department of Cellular Therapy, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Giancarlo Comi
- DIBIT II and Institute of Experimental Neurology (InSpe), San Raffaele Scientific Institute and Università Vita–Salute, Milan, Italy
- Department of Neurology and Neurophysiology, San Raffaele Scientific Institute and Università Vita–Salute, Milan, Italy
| | - Angelo A. Manfredi
- Clinical Immunology Unit, San Raffaele Scientific Institute and Università Vita–Salute, Milan, Italy
| | - Gianvito Martino
- Neuroimmunology Unit, San Raffaele Scientific Institute and Università Vita–Salute, Milan, Italy
- DIBIT II and Institute of Experimental Neurology (InSpe), San Raffaele Scientific Institute and Università Vita–Salute, Milan, Italy
- Department of Neurology and Neurophysiology, San Raffaele Scientific Institute and Università Vita–Salute, Milan, Italy
- * E-mail: (SP); (GM)
| |
Collapse
|
117
|
Chen B, Gao XQ, Yang CX, Tan SK, Sun ZL, Yan NH, Pang YG, Yuan M, Chen GJ, Xu GT, Zhang K, Yuan QL. Neuroprotective effect of grafting GDNF gene-modified neural stem cells on cerebral ischemia in rats. Brain Res 2009; 1284:1-11. [PMID: 19520066 DOI: 10.1016/j.brainres.2009.05.100] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Revised: 05/24/2009] [Accepted: 05/28/2009] [Indexed: 10/20/2022]
Abstract
Previous studies indicated the beneficial effects of glial cell line-derived neurotrophic factor (GDNF) and transplanted neural stem cells (NSCs) on stroke. Here, we explored whether transplantation of neural stem cells (NSCs) modified by GDNF gene provides a better therapeutic effect than native NSCs after stroke. Primary rat NSCs were transfected with GDNF plasmid (GDNF/NSCs, labeled by green fluorescent protein from AdEasy-1, GFP). Adult rats were subjected to two-hour middle cerebral artery occlusion and reperfusion, followed by infusion of NSCs (labeled with5-bromo-2'-deoxyuridine before infusion, BrdU), GDNF/NSCs and saline at 3 days after reperfusion (NSCs group, GDNF/NSCs group, control group), respectively. All rats were sacrificed at 1, 2, 3, 5, and 7 weeks after reperfusion. Modified Neurological Severity Scores (mNSS) test and H and E staining were respectively performed to evaluate neurological function and lesion volume. Immunohistochemistry was used to identify implanted cells and observe the expressions of Synaptophysin (Syp) and postsynaptic density-95 (PSD-95) and caspase-3. TdT-mediated dUTP-biotin nick-end labeling (TUNEL) was employed to observe apoptotic cells. Western blotting was used to detect brain-derived neurotrophic factor (BDNF) and NT-3 protein expression. Significant recovery of mNSS was found in GDNF/NSCs rats at 2 and 3 weeks after reperfusion compared with NSCs rats. Lesion volume in the NSCs and GDNF/NSCs groups was reduced significantly compared with control group. The number of NSCs in the GDNF/NSCs group was significantly increased in comparison with NSCs group. Moreover, Syp-immunoreactive product at 2 and 3 weeks after reperfusion and PSD-95 immunoreactive product in the GDNF/NSCs group were significantly increased compared with NSCs group. In contrast, caspase-3 positive cells and TUNEL-positive cells in the GDNF/NSCs group were significantly decreased compared with NSCs group. Significant increase of BDNF protein in the GDNF/NSCs and NSCs groups was observed compared to the control group at different time points of reperfusion, and GDNF/NSCs grafting significantly increased BDNF protein expression compared to NSCs grafting. In addition, significant increase of NT-3 protein in GDNF/NSCs and NSCs groups was detected only at 1 week of reperfusion compared to control group. The results demonstrate that grafting NSCs modified by GDNF gene provides better neuroprotection for stroke than NSCs grafting alone.
Collapse
Affiliation(s)
- B Chen
- Luzhou Medical College, Department of Neurobiology, 646000, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Lee HJ, Kim MK, Kim HJ, Kim SU. Human neural stem cells genetically modified to overexpress Akt1 provide neuroprotection and functional improvement in mouse stroke model. PLoS One 2009; 4:e5586. [PMID: 19440551 PMCID: PMC2679145 DOI: 10.1371/journal.pone.0005586] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2008] [Accepted: 04/02/2009] [Indexed: 12/22/2022] Open
Abstract
In a previous study, we have shown that human neural stem cells (hNSCs) transplanted in brain of mouse intracerebral hemorrhage (ICH) stroke model selectively migrate to the ICH lesion and induce behavioral recovery. However, low survival rate of grafted hNSCs in the brain precludes long-term therapeutic effect. We hypothesized that hNSCs overexpressing Akt1 transplanted into the lesion site could provide long-term improved survival of hNSCs, and behavioral recovery in mouse ICH model. F3 hNSC was genetically modified with a mouse Akt1 gene using a retroviral vector. F3 hNSCs expressing Akt1 were found to be highly resistant to H2O2-induced cytotoxicity in vitro. Following transplantation in ICH mouse brain, F3.Akt1 hNSCs induced behavioral improvement and significantly increased cell survival (50–100% increase) at 2 and 8 weeks post-transplantation as compared to parental F3 hNSCs. Brain transplantation of hNSCs overexpressing Akt1 in ICH animals provided functional recovery, and survival and differentiation of grafted hNSCs. These results indicate that the F3.Akt1 human NSCs should be a great value as a cellular source for the cellular therapy in animal models of human neurological disorders including ICH.
Collapse
Affiliation(s)
- Hong J. Lee
- Division of Neurology, Department of Medicine, UBC Hospital, University of British Columbia, Vancouver, Canada
- Medical Research Institute, Chungang University College of Medicine, Seoul, Korea
| | - Mi K. Kim
- Division of Neurology, Department of Medicine, UBC Hospital, University of British Columbia, Vancouver, Canada
| | - Hee J. Kim
- Division of Neurology, Department of Medicine, UBC Hospital, University of British Columbia, Vancouver, Canada
- Department of Pharmacology, Dankook University School of Medicine, Cheonan, Korea
| | - Seung U. Kim
- Division of Neurology, Department of Medicine, UBC Hospital, University of British Columbia, Vancouver, Canada
- Medical Research Institute, Chungang University College of Medicine, Seoul, Korea
- * E-mail:
| |
Collapse
|
119
|
MRI tracking of intravenously transplanted human neural stem cells in rat focal ischemia model. Neurosci Res 2009; 64:235-9. [PMID: 19428705 DOI: 10.1016/j.neures.2009.03.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2007] [Revised: 01/19/2009] [Accepted: 03/06/2009] [Indexed: 12/15/2022]
Abstract
The present study investigated the ability of 1.5 T clinical magnetic resonance imaging (MRI) to detect ferumoxides- labeled human neural stem cells (NSCs) that had been intravenously (i.v.) injected into a rat model of focal cerebral ischemia. To detect transplanted cells, hNSCs were labeled with ferumoxide then followed by bromodeoxyuridine (BrdU) prior to transplantation. In the rat ischemia-human NSC group, human NSCs (4 x 10(6)cells in 5 ml PBS) were injected via tail vein 24 h after middle cerebral artery occlusion (MCAo), and the brains of the rats were scanned using a 1.5 T MRI unit over a period of 4 weeks (1 day before MCAo, then 1 and 3 days after cell injection, and weekly thereafter). In histologic sections, transplanted cells were identified by Prussian blue and anti-BrdU fluorescence staining. Regions with hypointense signals on T2-weighted and 3D gradient echo MR images corresponded with areas stained by Prussian blue, which suggested the presence of superparamagnetic iron oxide (SPIO) nanoparticles within the engrafted cells. Hypointense areas on MR images were observed in peri-infarct areas 3 days after cell injection. The findings indicate that 1.5 T MRI has sufficient sensitivity to track engrafted stem cells in vivo.
Collapse
|
120
|
Zhang P, Li J, Liu Y, Chen X, Kang Q. Transplanted human embryonic neural stem cells survive, migrate, differentiate and increase endogenous nestin expression in adult rat cortical peri-infarction zone. Neuropathology 2009; 29:410-21. [PMID: 19170896 DOI: 10.1111/j.1440-1789.2008.00993.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Transplantation of stem cells is a potential therapeutic strategy for stroke damage. The survival, migration, and differentiation of transplanted human embryonic neural stem cells in the acute post-ischemic environment were characterized and endogenous nestin expression after transplantation was investigated. Human embryonic neural stem cells obtained from the temporal lobe cortex were cultured and labeled with fluorescent 1,1'-dioctadecy-6,6'-di (4-sulfopheyl)-3,3,3',3'-tetramethylindocarbocyanin (DiI) in vitro. Labeled cells were transplanted into cortical peri-infarction zones of adult rats 24 h after permanent middle cerebral artery occlusion. Survival, migration, and differentiation of grafted cells were quantified in immunofluorescence-stained sections from rats sacrificed at 7, 14, and 28 days after transplantation. Endogenous nestin-positive cells in the cortical peri-infarction zone were counted at serial time points. The cells transplanted into the cortical peri-infarction zone displayed the morphology of living cells and became widely located around the ischemic area. Moreover, some of the transplanted cells expressed nestin, GFAP, or NeuN in the peri-infarction zone. Furthermore, compared with the control group, endogenous nestin-positive cells in the peri-infarction zone had increased significantly 7 days after cell transplantation. These results confirm the survival, migration, and differentiation of transplanted cells in the acute post-ischemic environment and enhanced endogenous nestin expression within a brief time window. These findings indicate that transplantation of neural stem cells into the peri-infarction zone may be performed as early as 24 h after ischemia.
Collapse
Affiliation(s)
- Pengbo Zhang
- Institute of Neurobiology, National Key Academic Subject of Physiology, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | | | | | | | | |
Collapse
|
121
|
Yasuhara T, Date I. Gene therapy for Parkinson's disease. JOURNAL OF NEURAL TRANSMISSION. SUPPLEMENTUM 2009:301-309. [PMID: 20411788 DOI: 10.1007/978-3-211-92660-4_25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Parkinson's disease is characterized by the degeneration of the nigrostriatal dopaminergic neurons with the manifestation of tremor, rigidity, akinesia, and disturbances of postural reflexes. Medication using L-DOPA and surgeries including deep brain stimulation are the established therapies for Parkinson's disease. Cell therapies are also effective and have rapidly developed with the recent advancement in molecular biological technology including gene transfer. In this review, ex vivo gene therapy using genetically engineered cell transplantation for Parkinson's disease model of animals is described, including catecholamine/neurotrophic factor-secreting cell transplantation with or without encapsulation, as well as in vivo gene therapy using direct injection of viral vector to increase dopamine-production, ameliorate the survival of dopaminergic neurons, correct the deteriorated microenvironment, or normalize genetic abnormality. Furthermore, the future directions for clinical application are described together with recent clinical trials of gene therapy.
Collapse
Affiliation(s)
- Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, 2-5-1, Shikata-cho, Okayama, 700-8558, Japan.
| | | |
Collapse
|
122
|
Neural stem cells target intracranial glioma to deliver an oncolytic adenovirus in vivo. Gene Ther 2008; 16:262-78. [PMID: 19078993 DOI: 10.1038/gt.2008.165] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Adenoviral oncolytic virotherapy represents an attractive treatment modality for central nervous system (CNS) neoplasms. However, successful application of virotherapy in clinical trials has been hampered by inadequate distribution of oncolytic vectors. Neural stem cells (NSCs) have been shown as suitable vehicles for gene delivery because they track tumor foci. In this study, we evaluated the capability of NSCs to deliver a conditionally replicating adenovirus (CRAd) to glioma. We examined NSC specificity with respect to viral transduction, migration and capacity to deliver a CRAd to tumor cells. Fluorescence-activated cell sorter (FACS) analysis of NSC shows that these cells express a variety of surface receptors that make them amenable to entry by recombinant adenoviruses. Luciferase assays with replication-deficient vectors possessing a variety of transductional modifications targeted to these receptors confirm these results. Real-time PCR analysis of the replication profiles of different CRAds in NSCs and a representative glioma cell line, U87MG, identified the CRAd-Survivin (S)-pk7 virus as optimal vector for further delivery studies. Using in vitro and in vivo migration studies, we show that NSCs infected with CRAd-S-pk7 virus migrate and preferentially deliver CRAd to U87MG glioma. These results suggest that NSCs mediate an enhanced intratumoral distribution of an oncolytic vector in malignant glioma when compared with virus injection alone.
Collapse
|
123
|
Transduction of human neural progenitor cells with foamy virus vectors for differentiation-dependent gene expression. Gene Ther 2008; 16:349-58. [DOI: 10.1038/gt.2008.173] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
124
|
Park JE, Lee ST, Im WS, Chu K, Kim M. Galantamine reduces striatal degeneration in 3-nitropropionic acid model of Huntington's disease. Neurosci Lett 2008; 448:143-7. [DOI: 10.1016/j.neulet.2008.10.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2008] [Accepted: 10/01/2008] [Indexed: 01/17/2023]
|
125
|
Takahashi K, Yasuhara T, Shingo T, Muraoka K, Kameda M, Takeuchi A, Yano A, Kurozumi K, Agari T, Miyoshi Y, Kinugasa K, Date I. Embryonic neural stem cells transplanted in middle cerebral artery occlusion model of rats demonstrated potent therapeutic effects, compared to adult neural stem cells. Brain Res 2008; 1234:172-82. [DOI: 10.1016/j.brainres.2008.07.086] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2008] [Revised: 07/20/2008] [Accepted: 07/22/2008] [Indexed: 01/17/2023]
|
126
|
Structural and functional characteristic of a model for deep-seated lacunar infarct in rats. J Neurol Sci 2008; 273:40-8. [DOI: 10.1016/j.jns.2008.06.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2007] [Revised: 05/30/2008] [Accepted: 06/17/2008] [Indexed: 01/19/2023]
|
127
|
Fatar M, Stroick M, Griebe M, Marwedel I, Kern S, Bieback K, Giesel FL, Zechmann C, Kreisel S, Vollmar F, Alonso A, Back W, Meairs S, Hennerici MG. Lipoaspirate-derived adult mesenchymal stem cells improve functional outcome during intracerebral hemorrhage by proliferation of endogenous progenitor cells stem cells in intracerebral hemorrhages. Neurosci Lett 2008; 443:174-8. [PMID: 18691631 DOI: 10.1016/j.neulet.2008.07.077] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2008] [Revised: 07/14/2008] [Accepted: 07/23/2008] [Indexed: 01/14/2023]
Abstract
Stem cell therapy seems promising in reducing deficits after focal cerebral ischemia. As stroke may result from intracerebral hemorrhage (ICH) in up to 20% we investigated whether human processed lipoaspirate mesenchymal stem cells (PLA-MSC) influence the functional outcome, migration behavior and the activation of endogenous progenitor cells. Experimental ICH was induced by stereotactic administration of collagenase in rats randomly assigned to the control or treatment group. The latter received 3 x 10(6) PLA-MSC by intravenous (i.v.) injection 24h after ICH induction. The outcome was continuously monitored using the RotaRod test over a period of 4 weeks. Morphometric analysis of ICH was performed consecutively by magnetic resonance imaging (MRI) studies and immunohistochemical analysis. The RotaRod test revealed a significant 1.5-fold improvement (p<0.005) in functional outcome for the PLA-MSC treated group after 4 weeks compared to controls. Histological and MRI assessment of lesion size showed no difference between the two groups. Although i.v. injected human cells could not be detected in the post mortem brain, evaluation of the number of endogenous progenitor cells revealed a twofold increase in the treated animals compared to controls. Treatment with PLA-MSC improved the functional outcome significantly in an experimental ICH model. This effect was achieved by stimulation of endogenous progenitor cells rather than integration and differentiation of the infused PLA-MSC.
Collapse
Affiliation(s)
- Marc Fatar
- Department of Neurology, Universitätsklinikum Mannheim, University of Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Arterially perfused neurosphere-derived cells distribute outside the ischemic core in a model of transient focal ischemia and reperfusion in vitro. PLoS One 2008; 3:e2754. [PMID: 18648648 PMCID: PMC2453234 DOI: 10.1371/journal.pone.0002754] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Accepted: 06/25/2008] [Indexed: 01/19/2023] Open
Abstract
Background Treatment with neural stem cells represents a potential strategy to improve functional recovery of post-ischemic cerebral injury. The potential benefit of such treatment in acute phases of human ischemic stroke depends on the therapeutic viability of a systemic vascular delivery route. In spite of the large number of reports on the beneficial effects of intracerebral stem cells injection in experimental stroke, very few studies demonstrated the effectiveness of the systemic intravenous delivery approach. Metodology/Principal Findings We utilized a novel in vitro model of transient focal ischemia to analyze the brain distribution of neurosphere-derived cells (NCs) in the early 3 hours that follow transient occlusion of the medial cerebral artery (MCA). NCs obtained from newborn C57/BL6 mice are immature cells with self-renewal properties that could differentiate into neurons, astrocytes and oligodendrocytes. MCA occlusion for 30 minutes in the in vitro isolated guinea pig brain preparation was followed by arterial perfusion with 1×106 NCs charged with a green fluorescent dye, either immediately or 60 minutes after reperfusion onset. Changes in extracellular pH and K+ concentration during and after MCAO were measured through ion-sensitive electrodes. Conclusion/Significance It is demonstrated that NCs injected through the vascular system do not accumulate in the ischemic core and preferentially distribute in non-ischemic areas, identified by combined electrophysiological and morphological techniques. Direct measurements of extracellular brain ions during and after MCA occlusion suggest that anoxia-induced tissue changes, such as extracellular acidosis, may prevent NCs from entering the ischemic area in our in vitro model of transitory focal ischemia and reperfusion suggesting a role played by the surrounding microenviroment in driving NCs outside the ischemic core. These findings strongly suggest that the potential beneficial effect of NCs in experimental focal brain ischemia is not strictly dependent on their homing into the ischemic region, but rather through a bystander mechanism possibly mediated by the release of neuroprotective factors in the peri-infarct region.
Collapse
|
129
|
The SPECT imaging shows the accumulation of neural progenitor cells into internal organs after systemic administration in middle cerebral artery occlusion rats. Neurosci Lett 2008; 440:246-50. [PMID: 18572314 DOI: 10.1016/j.neulet.2008.05.090] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Revised: 05/12/2008] [Accepted: 05/20/2008] [Indexed: 11/21/2022]
Abstract
The regenerative potential of stem cells from various sources has been under intense investigation in the experimental models of cerebral ischemia. To end up with a restorative therapeutic treatment, it is crucial to get the cell transplants to the site of injury. Here, we evaluated the feasibility of small animal SPECT/CT in assessing the definite accumulation of (111)In-oxine-labeled human embryonic stem (ES) cell-derived neural progenitors and rat hippocampal progenitors after intravenous or intra-arterial administration (femoral vein vs. common carotid artery) in middle cerebral artery occlusion (MCAO) and sham-operated rats. Cell detection was carried out immediately and 24h after the infusion using a SPECT/CT device. The results showed that after intravenous injections both cell types accumulated primarily into internal organs, instead of brain. In contrast, after intra-arterial injection, a weak signal was detected in the ischemic hemisphere. Additional studies showed that the detection sensitivity of SPECT/CT device was approximately 1000 (111)In-oxine-labeled cells and labeling did not affect the cell viability. In conclusion, a small animal SPECT is powerful technique to study the whole body biodistribution of cell-based therapies. Our data showed that intravenous administration is not an optimal route to deliver neural progenitor cell-containing transplants into the brain after MCAO in rats.
Collapse
|
130
|
Yin L, Fu SL, Shi GY, Li Y, Jin JQ, Ma ZW, Lu PH. Expression and regulation of major histocompatibility complex on neural stem cells and their lineages. Stem Cells Dev 2008; 17:53-65. [PMID: 18230026 DOI: 10.1089/scd.2007.0063] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The expression of major histocompatibility complex (MHC) antigens on neural stem cells (NSCs) and their lineages is tightly related to the fate of these cells as grafts in allogenic transplantation. In this study, we observed that NSCs derived from embryonic rat forebrain expressed MHC class I and class II molecules at a low level, whereas the cells differentiated from NSCs, including neurons, astrocytes, and oligodendrocytes, lost their MHC expression. However, a proinflammatory factor, interferon-gamma (IFN-gamma), could induce and up-regulate the expression of MHC in both NSCs and their differentiated lineages in vitro. These results suggest that predifferentiating NSCs into lineage-limited cells prior to transplantation combined with controlling the local production of proinflammatory cytokines moderately may potentially benefit the survival of transplants.
Collapse
Affiliation(s)
- Lan Yin
- Department of Neurobiology, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | | | | | | | | | | | | |
Collapse
|
131
|
Production and characterization of immortal human neural stem cell line with multipotent differentiation property. Methods Mol Biol 2008; 438:103-21. [PMID: 18369753 DOI: 10.1007/978-1-59745-133-8_10] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
We document the protocols and methods for the production of immortalized cell lines of human neural stem cells from the human fetal central nervous system (CNS) cells by using a retroviral vector encoding v-myc oncogene. One of the human neural stem cell lines (HB1.F3) was found to express nestin and other specific markers for human neural stem cells, giving rise to three fundamental cell types of the CNS: neurons, astrocytes, and oligodendrocytes. After transplantation into the brain of mouse model of stroke, implanted human neural stem cells were observed to migrate extensively from the site of implantation into other anatomical sites and to differentiate into neurons and glial cells.
Collapse
|
132
|
Yu D, Silva GA. Stem cell sources and therapeutic approaches for central nervous system and neural retinal disorders. Neurosurg Focus 2008; 24:E11. [PMID: 18341387 DOI: 10.3171/foc/2008/24/3-4/e10] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the past decades, stem cell biology has made a profound impact on our views of mammalian development as well as opened new avenues in regenerative medicine. The potential of stem cells to differentiate into various cell types of the body is the principal reason they are being explored in treatments for diseases in which there may be dysfunctional cells and/or loss of healthy cells due to disease. In addition, other properties are unique to stem cells; their endogenous trophic support, ability to home to sites of pathological entities, and stability in culture, which allows genetic manipulation, are also being utilized to formulate stem cell-based therapy for central nervous system (CNS) disorders. In this review, the authors will review key characteristics of embryonic and somatic (adult) stem cells, consider therapeutic strategies employed in stem cell therapy, and discuss the recent advances made in stem cell-based therapy for a number of progressive neurodegenerative diseases in the CNS as well as neuronal degeneration secondary to other abnormalities and injuries. Although a great deal of progress has been made in our knowledge of stem cells and their utility in treating CNS disorders, much still needs to be elucidated regarding the biology of the stem cells and the pathogenesis of targeted CNS diseases to maximize therapeutic benefits. Nonetheless, stem cells present tremendous promise in the treatment of a variety of neurodegenerative diseases.
Collapse
Affiliation(s)
- Diana Yu
- Department of Bioengineering, University of California, San Diego, USA
| | | |
Collapse
|
133
|
Lim HC, Lee ST, Chu K, Joo KM, Kang L, Im WS, Park JE, Kim SU, Kim M, Cha CI. Neuroprotective effect of neural stem cell-conditioned media in in vitro model of Huntington's disease. Neurosci Lett 2008; 435:175-80. [DOI: 10.1016/j.neulet.2008.02.035] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2007] [Revised: 02/01/2008] [Accepted: 02/13/2008] [Indexed: 11/16/2022]
|
134
|
Chu K, Jung KH, Kim SJ, Lee ST, Kim J, Park HK, Song EC, Kim SU, Kim M, Lee SK, Roh JK. Transplantation of human neural stem cells protect against ischemia in a preventive mode via hypoxia-inducible factor-1alpha stabilization in the host brain. Brain Res 2008; 1207:182-92. [PMID: 18371939 DOI: 10.1016/j.brainres.2008.02.043] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Revised: 02/12/2008] [Accepted: 02/13/2008] [Indexed: 01/11/2023]
Abstract
Hypoxia-inducible factor-1 (HIF-1) plays important roles in the prevention of cerebral ischemia. Deferoxamine (DFX), an iron chelator stabilizes the HIF-1alpha and activates target genes involved in compensation for ischemia. In this study, we are to investigate whether HIF-1alpha can be stabilized in human neural stem cells (NSCs) by DFX, and pre-transplantation of NSCs with HIF-1alpha stabilization can induce prolonged ischemic tolerance. In the DFX-treated NSCs, the HIF-1alpha protein expression was increased about 100-fold time-dependently, and subsequent transcriptional activation (VEGF, BDNF and CXCR4) was also observed. To test an ability to induce ischemic prevention in vivo, DFX-treated NSCs or naïve NSCs were transplanted in the striatum of adult rats. Seven days following the transplantation, focal cerebral ischemia was done. Infarct volumes were reduced in both NSCs-transplanted groups, compared with ischemia-only, but more reduced in DFX-treated NSCs group. The protective effects of NSCs were ablated when HIF-1alpha was silenced. HIF-1alpha protein levels were increased in both NSCs-transplanted groups, but more increased in DFX-treated NSCs group. RT-PCR analysis manifested a downregulation of mRNA expression of TNF-alpha, IL-6 and MMP-9 in both NSCs groups, but further decrease in DFX-treated NSCs group. These findings provide evidence that HIF-1alpha stabilization in human NSCs can be achieved effectively by DFX, and HIF-1alpha-stabilized NSCs protect against ischemia in a preventive mode.
Collapse
Affiliation(s)
- Kon Chu
- Stroke and Neural Stem Cell Laboratory in Clinical Research Institute, Department of Neurology, Seoul National University Hospital, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Lee ST, Chu K, Park JE, Hong NH, Im WS, Kang L, Han Z, Jung KH, Kim MW, Kim M. Atorvastatin attenuates mitochondrial toxin-induced striatal degeneration, with decreasing iNOS/c-Jun levels and activating ERK/Akt pathways. J Neurochem 2008; 104:1190-200. [DOI: 10.1111/j.1471-4159.2007.05044.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
136
|
Guzman R, Choi R, Gera A, De Los Angeles A, Andres RH, Steinberg GK. Intravascular cell replacement therapy for stroke. Neurosurg Focus 2008; 24:E15. [DOI: 10.3171/foc/2008/24/3-4/e14] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
✓ The use of stem cell transplantation to restore neurological function after stroke is being recognized as a potential novel therapy. Before stem cell transplantation can become widely applicable, however, questions remain about the optimal site of delivery and timing of transplantation. In particular, there seems to be increasing evidence that intravascular cell delivery after stroke is a viable alternative to intracerebral transplantation. In this review, the authors focus on the intravascular delivery of stem cells for stroke treatment with an emphasis on timing, transendothelial migration and possible mechanisms leading to neuroprotection, angiogenesis, immunomodulation, and neural plasticity. They also review current concepts of in vivo imaging and tracking of stem cells after stroke.
Collapse
|
137
|
Jung KH, Chu K, Lee ST, Song EC, Sinn DI, Kim JM, Kim SJ, Kim JH, Kang KM, Park HK, Lee SH, Kim M, Lee SK, Roh JK. Identification of neuronal outgrowth cells from peripheral blood of stroke patients. Ann Neurol 2008; 63:312-22. [DOI: 10.1002/ana.21303] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
138
|
Granulocyte-colony stimulating factor attenuates striatal degeneration with activating survival pathways in 3-nitropropionic acid model of Huntington's disease. Brain Res 2008; 1194:130-7. [DOI: 10.1016/j.brainres.2007.11.058] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2007] [Revised: 11/24/2007] [Accepted: 11/27/2007] [Indexed: 12/11/2022]
|
139
|
Pluchino S, Martino G. The therapeutic plasticity of neural stem/precursor cells in multiple sclerosis. J Neurol Sci 2008; 265:105-10. [PMID: 17706971 DOI: 10.1016/j.jns.2007.07.020] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2007] [Revised: 06/08/2007] [Accepted: 07/17/2007] [Indexed: 01/19/2023]
Abstract
Adult multipotent neural stem/precursor cells (NPCs) have the capacity to self-renew and generate functional differentiated cells (e.g. neurons, astrocytes or oligodendrocytes) within discrete tissue-specific germinal niches, such as the subventricular zone of the lateral ventricles and the subgranular zone of the dentate gyrus of the hippocampus. Due to their intrinsic plasticity NPCs can be considered an essential part of the cellular mechanism(s) by which the central nervous system (CNS) tries to repair itself after an injury and, as a consequence, they also represents an attractive therapeutic tool for the treatment of neurological disorders. Here we will discuss not only the role of NPC-based transplantation therapies in multiple sclerosis (MS) but also recent data suggesting that endogenous NPCs, while contributing to CNS repair in MS, may also become the target of the disease itself.
Collapse
Affiliation(s)
- Stefano Pluchino
- Neuroimmunology Unit-DIBIT and Institute of Experimental Neurology, San Raffaele Scientific Institute, Via Olgettina 58, I-20132 Milan, Italy.
| | | |
Collapse
|
140
|
Zietlow R, Lane EL, Dunnett SB, Rosser AE. Human stem cells for CNS repair. Cell Tissue Res 2008; 331:301-22. [PMID: 17901985 DOI: 10.1007/s00441-007-0488-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2007] [Accepted: 07/25/2007] [Indexed: 12/31/2022]
Abstract
Although most peripheral tissues have at least a limited ability for self-repair, the central nervous system (CNS) has long been known to be relatively resistant to regeneration. Small numbers of stem cells have been found in the adult brain but do not appear to be able to affect any significant recovery following disease or insult. In the last few decades, the idea of being able to repair the brain by introducing new cells to repair damaged areas has become an accepted potential treatment for neurodegenerative diseases. This review focuses on the suitability of various human stem cell sources for such treatments of both slowly progressing conditions, such as Parkinson's disease, Huntington's disease and multiple sclerosis, and acute insult, such as stroke and spinal cord injury. Despite stem cell transplantation having now moved a step closer to the clinic with the first trials of autologous mesenchymal stem cells, the effects shown are moderate and are not yet at the stage of development that can fulfil the hopes that have been placed on stem cells as a means to replace degenerating cells in the CNS. Success will depend on careful investigation in experimental models to enable us to understand not just the practicalities of stem cell use, but also the underlying biological principles.
Collapse
Affiliation(s)
- Rike Zietlow
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, CF10 3US, UK.
| | | | | | | |
Collapse
|
141
|
Lee ST, Chu K, Jung KH, Kim SJ, Kim DH, Kang KM, Hong NH, Kim JH, Ban JJ, Park HK, Kim SU, Park CG, Lee SK, Kim M, Roh JK. Anti-inflammatory mechanism of intravascular neural stem cell transplantation in haemorrhagic stroke. ACTA ACUST UNITED AC 2007; 131:616-29. [PMID: 18156155 DOI: 10.1093/brain/awm306] [Citation(s) in RCA: 314] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Neural stem cell (NSC) transplantation has been investigated as a means to reconstitute the damaged brain after stroke. In this study, however, we investigated the effect on acute cerebral and peripheral inflammation after intracerebral haemorrhage (ICH). NSCs (H1 clone) from fetal human brain were injected intravenously (NSCs-iv, 5 million cells) or intracerebrally (NSCs-ic, 1 million cells) at 2 or 24 h after collagenase-induced ICH in a rat model. Only NSCs-iv-2 h resulted in fewer initial neurologic deteriorations and reduced brain oedema formation, inflammatory infiltrations (OX-42, myeloperoxidase) and apoptosis (activated caspase-3, TUNEL) compared to the vehicle-injected control animals. Rat neurosphere-iv-2 h, but not human fibroblast-iv-2 h, also reduced the brain oedema and the initial neurologic deficits. Human NSCs-iv-2 h also attenuated both cerebral and splenic activations of tumour necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), and nuclear factor-kappa B (NF-kappaB). However, we observed only a few stem cells in brain sections of the NSCs-iv-2 h group; in the main, they were detected in marginal zone of spleens. To investigate whether NSCs interact with spleen to reduce cerebral inflammation, we performed a splenectomy prior to ICH induction, which eliminated the effect of NSCs-iv-2 h transplantation on brain water content and inflammatory infiltrations. NSCs also inhibited in vitro macrophage activations after lipopolysaccharide stimulation in a cell-to-cell contact dependent manner. In summary, early intravenous NSC injection displayed anti-inflammatory functionality that promoted neuroprotection, mainly by interrupting splenic inflammatory responses after ICH.
Collapse
Affiliation(s)
- Soon-Tae Lee
- Stroke & Stem Cell Laboratory, Clinical Research Institute, Stem Cell Research Center, Department of Neurology, Seoul National University Hospital, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Abstract
Huntington disease (HD) is a devastating neurodegenerative disorder and no proven medical therapy is currently available to mitigate its clinical manifestations. Although fetal neural transplantation has been tried in both preclinical and clinical investigations, the efficacy is not satisfactory. With the recent explosive progress of stem cell biology, application of stem cell-based therapy in HD is an exciting prospect. Three kinds of stem cells, embryonic stem cells, bone marrow mesenchymal stem cells and neural stem cells, have previously been utilized in cell therapy in animal models of neurological disorders. However, neural stem cells were preferably used by investigators in experimental HD studies, since they have a clear capacity to become neurons or glial cells after intracerebral or intravenous transplantation, and they induce functional recovery. In this review, we summarize the current state of cell therapy utilizing stem cells in experimental HD animal models, and discuss the future considerations for developing new therapeutic strategies using neural stem cells.
Collapse
Affiliation(s)
- Manho Kim
- Department of Neurology, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | |
Collapse
|
143
|
Nagai A, Kim WK, Lee HJ, Jeong HS, Kim KS, Hong SH, Park IH, Kim SU. Multilineage potential of stable human mesenchymal stem cell line derived from fetal marrow. PLoS One 2007; 2:e1272. [PMID: 18060066 PMCID: PMC2092394 DOI: 10.1371/journal.pone.0001272] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Accepted: 09/04/2007] [Indexed: 12/21/2022] Open
Abstract
Human bone marrow contains two major cell types, hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs). MSCs possess self-renewal capacity and pluripotency defined by their ability to differentiate into osteoblasts, chondrocytes, adipocytes and muscle cells. MSCs are also known to differentiate into neurons and glial cells in vitro, and in vivo following transplantation into the brain of animal models of neurological disorders including ischemia and intracerebral hemorrhage (ICH) stroke. In order to obtain sufficient number and homogeneous population of human MSCs, we have clonally isolated permanent and stable human MSC lines by transfecting primary cell cultures of fetal human bone marrow MSCs with a retroviral vector encoding v-myc gene. One of the cell lines, HM3.B10 (B10), was found to differentiate into neural cell types including neural stem cells, neurons, astrocytes and oligodendrocytes in vitro as shown by expression of genetic markers for neural stem cells (nestin and Musashi1), neurons (neurofilament protein, synapsin and MAP2), astrocytes (glial fibrillary acidic protein, GFAP) and oligodendrocytes (myelin basic protein, MBP) as determined by RT-PCR assay. In addition, B10 cells were found to differentiate into neural cell types as shown by immunocytochical demonstration of nestin (for neural stem cells), neurofilament protein and β-tubulin III (neurons) GFAP (astrocytes), and galactocerebroside (oligodendrocytes). Following brain transplantation in mouse ICH stroke model, B10 human MSCs integrate into host brain, survive, differentiate into neurons and astrocytes and induce behavioral improvement in the ICH animals. B10 human MSC cell line is not only a useful tool for the studies of organogenesis and specifically for the neurogenesis, but also provides a valuable source of cells for cell therapy studies in animal models of stroke and other neurological disorders.
Collapse
Affiliation(s)
- Atsushi Nagai
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, Canada
- Department of Laboratory Medicine, Shimane University School of Medicine, Izumo, Japan
| | - Woo K. Kim
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, Canada
- Institute for Regnerative Medicine, Gachon University Gil Hospital, Inchon, Korea
| | - Hong J. Lee
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, Canada
- Institute for Regnerative Medicine, Gachon University Gil Hospital, Inchon, Korea
| | - Han S. Jeong
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, Canada
- Department of Physiology, Chonnam National University Medical School, Gwangju, Korea
| | - Kwang S. Kim
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Seok H. Hong
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, Canada
| | - In H. Park
- Institute for Regnerative Medicine, Gachon University Gil Hospital, Inchon, Korea
| | - Seung U. Kim
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, Canada
- Institute for Regnerative Medicine, Gachon University Gil Hospital, Inchon, Korea
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
144
|
Abstract
Myelination is critical for the normal functioning of the vertebrate nervous system. In the CNS, myelin is produced by oligodendrocytes, and the loss of oligodendrocytes and myelin results in severe functional impairment. Although spontaneous remyelination occurs in chronic demyelinating diseases such as multiple sclerosis, the repair process eventually fails, often resulting in long-term disability. Two distinct general approaches can be considered to promote myelin repair. In one the target is stimulation of the endogenous myelin repair process through delivery of growth factors, and in the second the target is augmentation of the repair process through the delivery of exogenous cells with myelination potential. In both cases, effective treatment of diseases such as multiple sclerosis requires modulation of the immune system, since demyelination is associated with specific immunological activation. Recent studies have shown that some populations of stem cells, including mesenchymal stem cells, have the capacity of promoting endogenous myelin repair and modulating the immune response, prompting an assessment of their use as therapy in demyelinating diseases such as MS. Other types of demyelinating disorders, such as the leukodystrophies, may require multiple repair strategies including both replacement of dysfunctional cells and delivery or supplementation of growth factors, immune modulators or metabolic enzymes. Here we discuss the use of stem cells for the treatment of demyelinating diseases. While the current number of stem cell-based clinical trials for demyelinating diseases is limited, this is likely to increase significantly in the next few years, and a clear understanding of the applicability, limitations and underlying mechanisms mediating stem cell repair is critical.
Collapse
Affiliation(s)
- Robert H Miller
- Center for Translational Neuroscience, Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | | |
Collapse
|
145
|
Darsalia V, Kallur T, Kokaia Z. Survival, migration and neuronal differentiation of human fetal striatal and cortical neural stem cells grafted in stroke-damaged rat striatum. Eur J Neurosci 2007; 26:605-14. [PMID: 17686040 DOI: 10.1111/j.1460-9568.2007.05702.x] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Stroke is a neurodegenerative disorder and the leading cause of disability in adult humans. Treatments to support efficient recovery in stroke patients are lacking. Several studies have demonstrated the ability of grafted neural stem cells (NSCs) to partly improve impaired neurological functions in stroke-subjected animals. Recently, we reported that NSCs from human fetal striatum and cortex exhibit region-specific differentiation in vitro, but survive, migrate and form neurons to a similar extent after intrastriatal transplantation in newborn rats. Here, we have transplanted the same cells into the stroke-damaged striatum of adult rats. The two types of NSCs exhibited a similar robust survival (30%) at 1 month after transplantation, and migrated throughout the damaged striatum. Striatal NSCs migrated farther and occupied a larger volume of striatum. In the transplantation core, cells were undifferentiated and expressed nestin and, to a lesser extent, also GFAP, betaIII-tubulin, DCX and calretinin, markers of immature neural lineage. Immunocytochemistry using markers of proliferation (p-H3 and Ki67) revealed a very low content of proliferating cells (<1%) in the grafts. Human cells outside the transplantation core differentiated, exhibited mature neuronal morphology and expressed mature neuronal markers such as HuD, calbindin and parvalbumin. Interestingly, striatal NSCs generated a greater number of parvalbumin+ and calbindin+ neurons. Virtually none of the grafted cells differentiated into astrocytes or oligodendrocytes. Based on these data, human fetal striatum- and cortex-derived NSCs could be considered potentially safe and viable for transplantation, with strong neurogenic potential, for further exploration in animal models of stroke.
Collapse
Affiliation(s)
- Vladimer Darsalia
- Laboratory of Neural Stem Cell Biology, Section of Restorative Neurology, Lund Strategic Research Center for Stem Cell Biology and Cell Therapy, University Hospital BMC B10, Klinikgatan 26, SE-221 84 Lund, Sweden
| | | | | |
Collapse
|
146
|
Hwang DW, Kang JH, Jeong JM, Chung JK, Lee MC, Kim S, Lee DS. Noninvasive in vivo monitoring of neuronal differentiation using reporter driven by a neuronal promoter. Eur J Nucl Med Mol Imaging 2007; 35:135-45. [PMID: 17885755 DOI: 10.1007/s00259-007-0561-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2007] [Accepted: 07/15/2007] [Indexed: 01/27/2023]
Abstract
PURPOSE We imaged neuronal differentiation in vivo using dual reporters (sodium iodide symporter [NIS] and luciferase) coupled to a neuron-specific enolase (NSE) promoter. METHODS PC12 (NSE positive) and F11 cells were transfected with a bicistronic (NIS and luciferase; pNSE-NF) or a luciferase (pNSE-Fluc) reporter coupled to the NSE promoter. Weak NSE promoter activity was overcome by a two-step transcriptional amplification (TSTA) system (pNSE-TSTA-Fluc). In vivo, NIS and luciferase expression were examined using a (99m)Tc-pertechnetate gamma camera and bioluminescence imaging, respectively. RESULTS pNSE-NF-transfected PC12 cells showed 3-fold higher radioiodine uptakes and >100-fold higher luciferase activity than parental cells. NIS or luciferase activity was not detected in pNSE-NF-transfected HeLa cells. When F11 cells were differentiated into neurons by db-cAMP, NIS and luciferase activities increased 4-fold compared to those without treatment, which was confirmed by Western blot and RT-PCR of NSE. In vivo in pNSE-NF-transfected F11 cells, db-cAMP treatment increased the luciferase activity but not the scintigraphic activity. In vitro, pNSE-TSTA-Fluc produced 130-fold higher luciferase activity than pNSE-Fluc and neuronal differentiation showed 4-fold higher activity from both pNSE-TSTA-Fluc and pNSE-Fluc than before differentiation. In vivo, in pNSE-TSTA-Fluc-transfected F11 cells, luciferase activity increased after neuronal differentiation. In vivo luciferase activity persisted up to 2 days after db-cAMP-induced neuronal differentiation. CONCLUSION NSE promoter-driven dual reporter transgenes revealed the possibility of in vivo imaging of neuronal differentiation, which was further enabled by high amplification using a TSTA system. We propose that this strategy be used to follow the transplanted stem cells during differentiation in live animals.
Collapse
Affiliation(s)
- Do Won Hwang
- Programs in Neuroscience, Seoul National University, Seoul, South Korea
| | | | | | | | | | | | | |
Collapse
|
147
|
Kim JM, Lee ST, Chu K, Jung KH, Song EC, Kim SJ, Sinn DI, Kim JH, Park DK, Kang KM, Hyung Hong N, Park HK, Won CH, Kim KH, Kim M, Kun Lee S, Roh JK. Systemic transplantation of human adipose stem cells attenuated cerebral inflammation and degeneration in a hemorrhagic stroke model. Brain Res 2007; 1183:43-50. [PMID: 17920570 DOI: 10.1016/j.brainres.2007.09.005] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2007] [Revised: 09/04/2007] [Accepted: 09/04/2007] [Indexed: 12/18/2022]
Abstract
Adipose-derived stem cells (ASCs) are readily accessible multipotent mesenchymal stem cells and are known to secrete multiple growth factors, and thereby to have cytoprotective effects in various injury models. In the present study, the authors investigated the neuroprotective effect of ASCs in an intracerebral hemorrhage (ICH) model. ICH was induced via the stereotaxic infusion of collagenase, and human ASCs (three million cells per animal) isolated from human fresh fat tissue, were intravenously administered at 24 h post-ICH induction. Acute brain inflammation markers, namely, cell numbers positively stained for terminal transferase dUTP nick end labeling (TUNEL), myeloperoxidase (MPO), or OX-42, and brain water content were checked at 3 days post-ICH. In addition, the authors quantified brain degeneration by measuring hemispheric atrophy and perihematomal glial thickness at 6 weeks post-ICH, and determined modified limb placing behavioral scores weekly over 5 weeks post-ICH. The results showed that brain water content, TUNEL+, and MPO+ cell numbers were significantly reduced in the ASC-transplanted rats. ASC transplantation attenuated neurological deficits from 4 to 5 weeks post-ICH, and reduced both the brain atrophy and the glial proliferation at 6 weeks. Transplanted ASCs were found to densely populate perihematomal areas at 6 weeks, and to express endothelial markers (von Willebrand factor and endothelial barrier antigen), but not neuronal or glial markers. In summary, ASCs transplantation in the ICH model reduced both acute cerebral inflammation and chronic brain degeneration, and promoted long-term functional recovery.
Collapse
Affiliation(s)
- Jeong-Min Kim
- Stroke and Stem Cell Laboratory in the Clinical Research Institute, Stem Cell Research Center, Department of Neurology, Seoul National University Hospital, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Bacigaluppi M, Pluchino S, Martino G, Kilic E, Hermann DM. Neural stem/precursor cells for the treatment of ischemic stroke. J Neurol Sci 2007; 265:73-7. [PMID: 17610905 DOI: 10.1016/j.jns.2007.06.012] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2007] [Revised: 06/04/2007] [Accepted: 06/14/2007] [Indexed: 12/23/2022]
Abstract
In ischemic stroke, the third most frequent cause of mortality in industrialized countries, therapeutic options have until now been limited to the first hours after disease onset. Cell transplantation has emerged in various neurological disorders, including experimental stroke, as a successful recovery-promoting approach also in the post-acute stroke phase. However, before envisaging any translation into humans of such promising cell-based approaches we still need to clarify: (i) the ideal cell source for transplantation, (ii) the most appropriate route of cell administration, and, last but not least, (iii) the best approach to achieve an appropriate and functional integration of transplanted cells into the host tissue. Here we discuss, with special emphasis on neural stem/precursor cells, potential mechanisms that may be involved in the action of cell-based therapies in stroke.
Collapse
Affiliation(s)
- Marco Bacigaluppi
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstr, 26, CH-8091 Zurich, Switzerland
| | | | | | | | | |
Collapse
|
149
|
Viktorov IV, Savchenko EA, Ukhova OV, Alekseyeva NY, Chekhonin VP. Multipotent stem and progenitor cells of the olfactory epithelium. Bull Exp Biol Med 2007; 142:495-502. [PMID: 17415447 DOI: 10.1007/s10517-006-0402-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In recent decades, a wide spectrum of fetal and embryonic stem and progenitor cells were used for cell therapy of diseases of the central nervous system, but the olfactory glial ensheathing cells exhibited certain advantages due to their biological properties and capacity to stimulate regeneratory processes in spinal injury. The therapeutic effect of a heterogeneous complex of olfactory epithelial cells is more pronounced; apart from glial ensheathing cells, this complex includes fibroblasts, Schwann cells, stem and progenitor cells of this structure. The use of minimally invasive methods for isolation of human olfactory epithelial tissue is important for clinical practice, because they provide cells for autologous transplantation and rule out graft rejection immune reaction and the risk of transmission viral infection and transfer of genetic defects, which can be associated with allotransplantation.
Collapse
Affiliation(s)
- I V Viktorov
- V. P. Serbsky State Research Center of Social and Forensic Psychiatry, Moscow.
| | | | | | | | | |
Collapse
|
150
|
Kim SU. Genetically engineered human neural stem cells for brain repair in neurological diseases. Brain Dev 2007; 29:193-201. [PMID: 17303360 DOI: 10.1016/j.braindev.2006.07.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2006] [Accepted: 07/31/2006] [Indexed: 11/23/2022]
Abstract
Neural stem cells (NSCs)of the central nervous system (CNS) have recently received a great deal of attention and interest for their therapeutic potential for neurological disorders. NSCs are defined as CNS progenitor cells that have the capacity for self-renewal and multipotent potential to become neurons or glial cells. Recent studies have shown that NSCs isolated from mammalian CNS including human can be propagated in vitro and then implanted into the brain of animal models of human neurological disorders. Recently, we have generated clonally derived immortalized human NSC cell lines via a retroviral vector encoded with v-myc oncogene. One of the human NSC lines, HB1.F3, was utilized in stem-cell based therapy in animal models of human neurological disorders. When F3 human NSCs were implanted into the brain of murine models of lysosomal storage diseases, stroke, Parkinson disease, Huntington disease or stroke, implanted F3 NSCs were found to migrate to the lesion sites, differentiate into neurons and glial cells, and restore functional deficits found in these neurological disorders. In animal models of brain tumors, F3 NSCs could deliver a bioactive therapeutically relevant molecules to effect a significant anti-tumor response intracranial tumor mass. Since these genetically engineered human NSCs are immortalized and continuously multiplying, there would be limitless supply of human neurons for treatment for patients suffering from neurological disorders including stroke, Parkinson disease, Huntington disease, ALS, multiple sclerosis and spinal cord injury. The promising field of stem cell research as it applies to regenerative medicine is still in infancy, but its potential appears limitless, and we are blessed to be involved in this exciting realm of research.
Collapse
Affiliation(s)
- Seung U Kim
- Brain Disease Research Center, Ajou University School of Medicine, Suwon 442-721, Republic of Korea.
| |
Collapse
|