351
|
Sasaki M, Honmou O, Kocsis JD. A rat middle cerebral artery occlusion model and intravenous cellular delivery. Methods Mol Biol 2009; 549:187-195. [PMID: 19378204 DOI: 10.1007/978-1-60327-931-4_13] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
A useful experimental model to study the pathophysiology of cerebral ischemia without craniectomy is the middle cerebral artery occlusion (MCAO) model. In this model, an intraluminal suture is advanced from the internal carotid artery to occlude the base of the MCA. Standardized procedures in terms of suture size, animal weight, and the details of intraluminal suture insertion are well established. This procedure can produce reversible occlusion after insertion of the intraluminal suture for a specified period of time, or a permanent occlusion by leaving the suture in place. This model has been useful in the study of both the normal pathophysiology of cerebral ischemia and in assessing interventional therapeutic approaches for stroke therapy. One approach has been the intravenous delivery of bone marrow-derived mescenchymal stem cells at various times after MCAO. Histological and magnetic resonance imaging have been used to quantify infarction volume in this model system.
Collapse
Affiliation(s)
- Masanori Sasaki
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
| | | | | |
Collapse
|
352
|
Horn AP, Frozza RL, Grudzinski PB, Gerhardt D, Hoppe JB, Bruno AN, Chagastelles P, Nardi NB, Lenz G, Salbego C. Conditioned medium from mesenchymal stem cells induces cell death in organotypic cultures of rat hippocampus and aggravates lesion in a model of oxygen and glucose deprivation. Neurosci Res 2009; 63:35-41. [DOI: 10.1016/j.neures.2008.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2008] [Revised: 09/15/2008] [Accepted: 10/01/2008] [Indexed: 11/16/2022]
|
353
|
Muraro PA, Uccelli A. Immuno-therapeutic potential of haematopoietic and mesenchymal stem cell transplantation in MS. Results Probl Cell Differ 2009; 51:237-57. [PMID: 19513637 DOI: 10.1007/400_2008_14] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In the last few years there has been extraordinary progress in the field of stem cell research. Two types of stem cells populate the bone marrow: haematopoietic stem/progenitor cells (HSC) and mesenchymal stem cells (MSC). The capacity of HSC to repopulate the blood has been known and exploited therapeutically for at least four decades. Today, haematopoietic stem cell transplantation (HSCT) holds a firm place in the therapy of some haematological malignancies, and a potential role of HSCT for treatment of severe autoimmune diseases has been explored in small-scale clinical studies. Multiple sclerosis (MS) is the noncancerous immune mediated disease for which the greatest number of transplants has been performed to date. The results of clinical studies are double-faced: on the one hand, HSCT has demonstrated powerful effects on acute inflammation, arresting the development of focal CNS lesions and clinical relapses; on the other hand, the treatment did not arrest chronic worsening of disability in most patients with secondary progressive MS, suggesting limited or no beneficial effects on the chronic processes causing progressive disability. MSC are a more recent addition to the range of experimental therapies being developed to treat MS. While interest in MSC usage was originally raised by their potential capacity to differentiate into different cell lineages, recent work showing their interesting immunological properties has led to a revised concept, envisioning their utilization for immuno-modulatory purposes. In this review we will summarize the current clinical and experimental evidence on HSC and MSC and outline some key questions warranting further investigation in this exciting research area.
Collapse
Affiliation(s)
- Paolo A Muraro
- Division of Neuroscience and Mental Health, Faculty of Medicine, Imperial College Building 560/Burlington Danes, Room E415, Imperial College London, 160 Du Cane Road, London, UK.
| | | |
Collapse
|
354
|
Abstract
The characteristic CNS responses to injury including increased cell production and attempts at regenerative repair - implicitly predicted where not directly demonstrated by Cajal, but only now more fully confirmed - have important implications for regenerative therapies. Spontaneous CNS cell replacement compares poorly with the regenerative functional repair seen elsewhere, but harnessing, stimulating or supplementing this process represents a new and attractive therapeutic concept.Stem cells, traditionally defined as clone-forming, self-renewing, pluripotent progenitor cells, have already proved themselves to be an invaluable source of transplantation material in several clinical settings, most notably haematological malignancy, and attention is now turning to a wider variety of diseases in which there may be potential for therapeutic intervention with stem cell transplantation. Neurological diseases, with their reputation for relentless progression and incurability are particularly tantalising targets. The optimal source of stem cells remains to be determined but bone marrow stem cells may themselves be included amongst the contenders.Any development of therapies using stem cells must depend on an underlying knowledge of their basic biology. The haemopoietic system has long been known to maintain circulating populations of cells with short life spans, and this system has greatly informed our knowledge of stem cell biology. In particular, it has helped yield the traditional stem cell model - a hierarchical paradigm of progressive lineage restriction. As cells differentiate, their fate choices become progressively more limited, and their capacity for proliferation reduced, until fully differentiated, mitotically quiescent cells are generated. Even this, however, is now under challenge.
Collapse
Affiliation(s)
- C M Rice
- University of Bristol Institute of Clinical Neurosciences, Frenchay Hospital, Bristol, UK
| | | |
Collapse
|
355
|
Someya Y, Koda M, Dezawa M, Kadota T, Hashimoto M, Kamada T, Nishio Y, Kadota R, Mannoji C, Miyashita T, Okawa A, Yoshinaga K, Yamazaki M. Reduction of cystic cavity, promotion of axonal regeneration and sparing, and functional recovery with transplanted bone marrow stromal cell–derived Schwann cells after contusion injury to the adult rat spinal cord. J Neurosurg Spine 2008; 9:600-10. [DOI: 10.3171/spi.2008.9.08135] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Object
The authors previously reported that Schwann cells (SCs) could be derived from bone marrow stromal cells (BMSCs) in vitro and that they promoted axonal regeneration of completely transected rat spinal cords in vivo. The aim of the present study is to evaluate the efficacy of transplanted BMSC-derived SCs (BMSC-SCs) in a rat model of spinal cord contusion, which is relevant to clinical spinal cord injury.
Methods
Bone marrow stromal cells were cultured as plastic-adherent cells from the bone marrow of GFPtransgenic rats. The BMSC-SCs were derived from BMSCs in vitro with sequential treatment using beta-mercaptoethanol, all-trans-retinoic acid, forskolin, basic fibroblast growth factor, platelet derived–growth factor, and heregulin. Schwann cells were cultured from the sciatic nerve of neonatal, GFP-transgenic rats. Immunocytochemical analysis and the reverse transcriptase–polymerase chain reaction were performed to characterize the BMSC-SCs. For transplantation, contusions with the New York University impactor were delivered at T-9 in 10- to 11-week-old male Wistar rats. Four groups of rats received injections at the injury site 7 days postinjury: the first received BMSCSCs and matrigel, a second received peripheral SCs and matrigel, a third group received BMSCs and matrigel, and a fourth group received matrigel alone. Histological and immunohistochemical studies, electron microscopy, and functional assessments were performed to evaluate the therapeutic effects of BMSC-SC transplantation.
Results
Immunohistochemical analysis and reverse transcriptase–polymerase chain reaction revealed that BMSC-SCs have characteristics similar to SCs not only in their morphological characteristics but also in their immunocytochemical phenotype and genotype. Histological examination revealed that the area of the cystic cavity was significantly reduced in the BMSC-SC and SC groups compared with the control rats. Immunohistochemical analysis showed that transplanted BMSCs, BMSC-SCs, and SCs all maintained their original phenotypes. The BMSC-SC and SC groups had a larger number of tyrosine hydroxilase–positive fibers than the control group, and the BMSC-SC group had more serotonin-positive fibers than the BMSC or control group. The BMSC-SC group showed significantly better hindlimb functional recovery than in the BMSC and control group. Electron microscopy revealed that transplanted BMSC-SCs existed in association with the host axons.
Conclusions
Based on their findings, the authors concluded that BMSC-SC transplantation reduces the size of the cystic cavity, promotes axonal regeneration and sparing, results in hindlimb functional recovery, and can be a useful tool for spinal cord injury as a substitute for SCs.
Collapse
Affiliation(s)
| | - Masao Koda
- 3Department of Orthopaedic Surgery, Prefectural Togane Hospital, Chiba
| | - Mari Dezawa
- 4Department of Anatomy and Neurobiology, Kyoto University Graduate School of Medicine, Kyoto; and
| | - Tomoko Kadota
- 2Bioenvironmental Medicine, Chiba University Graduate School of Medicine, Chiba
| | | | | | | | | | | | | | | | | | | |
Collapse
|
356
|
Shen LH, Gao Q, Li Y, Savant-Bhonsale S, Chopp M. Down-regulation of neurocan expression in reactive astrocytes promotes axonal regeneration and facilitates the neurorestorative effects of bone marrow stromal cells in the ischemic rat brain. Glia 2008; 56:1747-54. [PMID: 18618668 PMCID: PMC2575136 DOI: 10.1002/glia.20722] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The glial scar, a primarily astrocytic structure bordering the infarct tissue inhibits axonal regeneration after stroke. Neurocan, an axonal extension inhibitory molecule, is up-regulated in the scar region after stroke. Bone marrow stromal cells (BMSCs) reduce the thickness of glial scar wall and facilitate axonal remodeling in the ischemic boundary zone. To further clarify the role of BMSCs in axonal regeneration and its underlying mechanism, the current study focused on the effect of BMSCs on neurocan expression in the ischemic brain. Thirty-one adult male Wistar rats were subjected to 2 h of middle cerebral artery occlusion followed by an injection of 3 x 10(6) rat BMSCs (n = 16) or phosphate-buffered saline (n = 15) into the tail vein 24 h later. Animals were sacrificed at 8 days after stroke. Immunostaining analysis showed that reactive astrocytes were the primary source of neurocan, and BMSC-treated animals had significantly lower neurocan and higher growth associated protein 43 expression in the penumbral region compared with control rats, which was confirmed by Western blot analysis of the brain tissue. To further investigate the effects of BMSCs on astrocyte neurocan expression, single reactive astrocytes were collected from the ischemic boundary zone using laser capture microdissection. Neurocan gene expression was significantly down-regulated in rats receiving BMSC transplantation (n = 4/group). Primary cultured astrocytes showed similar alterations; BMSC coculture during reoxygenation abolished the up-regulation of neurocan gene in astrocytes undergoing oxygen-glucose deprivation (n = 3/group). Our data suggest that BMSCs promote axonal regeneration by reducing neurocan expression in peri-infarct astrocytes.
Collapse
Affiliation(s)
- Li Hong Shen
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, U.S.A
| | - Qi Gao
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, U.S.A
| | - Yi Li
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, U.S.A
| | | | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, U.S.A
- Department of Physics, Oakland University, Rochester, Michigan, U.S.A
| |
Collapse
|
357
|
Dharmasaroja P. Bone marrow-derived mesenchymal stem cells for the treatment of ischemic stroke. J Clin Neurosci 2008; 16:12-20. [PMID: 19017556 DOI: 10.1016/j.jocn.2008.05.006] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2008] [Revised: 05/12/2008] [Accepted: 05/17/2008] [Indexed: 02/01/2023]
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs) have great potential as therapeutic agents in stroke management, since they are easily accessible and can be rapidly expanded ex vivo for autologous transplantation. Increasing evidence suggests that bone marrow cells migrate throughout the brain and differentiate into neurons and glial cells. Both non-human and human MSCs have been used to treat stroke in murine models with satisfactory results. Several factors, such as transdifferentiation, induction of neurogenesis and angiogenesis, neuroprotection, and activation of endogenous neurorestorative processes, contribute to the benefits of MSCs in the ischemic brain. Many variables, including types of MSCs, cell dose, timing of treatment, route of cell delivery, and characteristics of stroke patients, influence the efficacy of MSC treatment of stroke. Although the first trials of autologous MSC therapy in stroke patients showed promising results, the optimal approach for different clinical settings has yet to be determined. The fundamental properties of MSCs and their potential short-term and long-term toxicities also need to be determined before moving forward to use of these cells in clinical practice.
Collapse
Affiliation(s)
- Permphan Dharmasaroja
- Department of Anatomy and Center for Neuroscience, Faculty of Science, Mahidol University, Rama VI Road, Rajthevi, Bangkok 10400, Thailand.
| |
Collapse
|
358
|
Functional recovery of chronic paraplegic pigs after autologous transplantation of bone marrow stromal cells. Transplantation 2008; 86:845-53. [PMID: 18813110 DOI: 10.1097/tp.0b013e318186198f] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Bone marrow stromal cells (BMSC) transplantation offers promise in the treatment of chronic paraplegia in rodents. Here, we report the effect of this cell therapy in adult pigs suffering chronic paraplegia. METHODS Three months after spinal cord injury, autologous BMSC in autologous plasma was injected into lesion zone and adjacent subarachnoid space in seven paraplegic pigs. On the contrary, three paraplegic pigs only received autologous plasma. Functional outcome was measured weekly until the end of the follow-up, 3 months later. RESULTS Our present study showed progressive functional recovery in transplanted pigs. At this time, intramedullary posttraumatic cavities were filled by a neoformed tissue containing several axons, together with BMSC that expressed neuronal or glial markers. Furthermore, in the treated animals, electrophysiological studies showed recovery of the previously abolished somatosensory-evoked potentials. CONCLUSIONS These findings confirm previous observations in rodents and support the possible utility of BMSC transplantation in humans suffering chronic paraplegia.
Collapse
|
359
|
Kwon DS, Gao X, Liu YB, Dulchavsky DS, Danyluk AL, Bansal M, Chopp M, McIntosh K, Arbab AS, Dulchavsky SA, Gautam SC. Treatment with bone marrow-derived stromal cells accelerates wound healing in diabetic rats. Int Wound J 2008; 5:453-63. [PMID: 18593394 DOI: 10.1111/j.1742-481x.2007.00408.x] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Bone marrow stem cells participate in tissue repair processes and may have a role in wound healing. Diabetes is characterised by delayed and poor wound healing. We investigated the potential of bone marrow-derived mesenchymal stromal cells (BMSCs) to promote healing of fascial wounds in diabetic rats. After manifestation of streptozotocin (STZ)-induced diabetic state for 5 weeks in male adult Sprague-Dawley rats, healing of fascial wounds was severely compromised. Compromised wound healing in diabetic rats was characterised by excessive polymorphonuclear cell infiltration, lack of granulation tissue formation, deficit of collagen and growth factor [transforming growth factor (TGF-beta), epidermal growth factor (EGF), vascular endothelial growth factor (VEGF), platelet-derived growth factor PDGF-BB and keratinocyte growth factor (KGF)] expression in the wound tissue and significant decrease in biomechanical strength of wounds. Treatment with BMSC systemically or locally at the wound site improved the wound-breaking strength (WBS) of fascial wounds. The improvement in WBS was associated with an immediate and significant increase in collagen levels (types I-V) in the wound bed. In addition, treatment with BMSCs increased the expression of growth factors critical to proper repair and regeneration of the damaged tissue moderately (TGF-beta, KGF) to markedly (EGF, VEGF, PDGF-BB). These data suggest that cell therapy with BMSCs has the potential to augment healing of the diabetic wounds.
Collapse
Affiliation(s)
- David S Kwon
- Department of General Surgery, Henry Ford Health System, Detroit, MI, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
360
|
Abstract
This paper is concerned about dental-derived stem cells and their characterization in vitro and in vivo. Additionally, since conventional periodontal techniques remain insufficient to attain complete and reliable periodontal regeneration, the potential of dental-derived stem cells in promoting periodontal tissue regeneration is also reviewed.
Collapse
|
361
|
Intra-arterial transplantation of bone marrow mononuclear cells immediately after reperfusion decreases brain injury after focal ischemia in rats. Life Sci 2008; 83:433-7. [DOI: 10.1016/j.lfs.2008.07.018] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Revised: 07/10/2008] [Accepted: 07/15/2008] [Indexed: 12/14/2022]
|
362
|
Combined therapeutic strategy using erythropoietin and mesenchymal stem cells potentiates neurogenesis after transient focal cerebral ischemia in rats. J Cereb Blood Flow Metab 2008; 28:1552-63. [PMID: 18478023 DOI: 10.1038/jcbfm.2008.40] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Many studies showed beneficial effects of either erythropoietin (EPO) or mesenchymal stem cells (MSCs) treatment in cerebral ischemia. In addition to a neuroprotective role, not only EPO but also MSC favors neurogenesis and functional recovery. In an attempt to further improve postischemic tissue repair, we investigated the effect of a systemic administration of MSC, in the presence or not of EPO, on neurogenesis and functional recovery in a transient focal cerebral ischemia model in the adult rat. Twenty-four hours after ischemia, the rats were divided into four groups, namely vehicle, MSC, EPO, and MSC+EPO, and received a single intravenous injection of MSC (2 x 10(6) cells) and/or a repeated intraperitoneal administration of EPO (1,000 UI/kg) for 3 days. The lesion volume, the MSC outcome, neurogenesis, and functional recovery were assessed 51 days after ischemia. The results showed that cellular proliferation and neurogenesis were increased along the lateral ventricle wall in the MSC+EPO group, whereas no significant effect was observed in groups receiving MSC or EPO alone. This effect was accompanied by an improvement of mnesic performances. Mesenchymal stem cells expressing neuronal or glial markers were detected in the ischemic hemisphere. These results suggest that EPO could act in a synergistic way with MSC to potentiate the postischemic neurogenesis.
Collapse
|
363
|
Borlongan CV, Chopp M, Steinberg GK, Bliss TM, Li Y, Lu M, Hess DC, Kondziolka D. Potential of stem/progenitor cells in treating stroke: the missing steps in translating cell therapy from laboratory to clinic. Regen Med 2008; 3:249-50. [PMID: 18462048 DOI: 10.2217/17460751.3.3.249] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
364
|
Dai LJ, Li HY, Guan LX, Ritchie G, Zhou JX. The therapeutic potential of bone marrow-derived mesenchymal stem cells on hepatic cirrhosis. Stem Cell Res 2008; 2:16-25. [PMID: 19383405 DOI: 10.1016/j.scr.2008.07.005] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Revised: 07/11/2008] [Accepted: 07/31/2008] [Indexed: 12/13/2022] Open
Abstract
Hepatic cirrhosis is the end-stage of chronic liver diseases. The majority of patients with hepatic cirrhosis die from life-threatening complications occurring at their earlier ages. Liver transplantation has been the most effective treatment for these patients. Since liver transplantation is critically limited by the shortage of available donor livers, searching for an effective alternative therapy has attracted great interest in preclinical studies. The transplantation of autologous bone marrow-derived mesenchymal stem cells holds great potential for treating hepatic cirrhosis. Mesenchymal stem cells can differentiate to hepatocytes, stimulate the regeneration of endogenous parenchymal cells, and enhance fibrous matrix degradation. Experimental and clinical studies have shown promising beneficial effects. This review is intended to translate the bench study results to the patients' bedside. The potential interventions of mesenchymal stem cells on cirrhosis are illustrated in terms of the cellular and molecular mechanisms of hepatic fibrogenesis.
Collapse
Affiliation(s)
- Long-Jun Dai
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada.
| | | | | | | | | |
Collapse
|
365
|
Abstract
Cell-based therapy for stroke represents a third wave of therapeutics for stroke and one focused on restorative processes with a longer time window of opportunity than neuroprotective therapies. An early time window, within the first week after stroke, is an opportunity for intravenously delivered bone marrow and perinatally derived cells that can home to areas of tissue injury and target brain remodeling. Allogeneic cells will likely be the most scalable and commercially viable product. Later time windows, months after stroke, may be opportunities for intracerebral transplantation of neuronally differentiated cell types. An integrated approach of cell-based therapy with early-phase clinical trials and continued preclinical work with focus on mechanisms of action is needed.
Collapse
Affiliation(s)
- David C Hess
- Department of Neurology, Medical College of Georgia, Augusta, GA 30912, USA.
| | | |
Collapse
|
366
|
Treatment of stroke with (Z)-1-[N-(2-aminoethyl)-N-(2-ammonioethyl) amino] diazen-1-ium-1, 2-diolate and bone marrow stromal cells upregulates angiopoietin-1/Tie2 and enhances neovascularization. Neuroscience 2008; 156:155-64. [PMID: 18691637 DOI: 10.1016/j.neuroscience.2008.07.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2008] [Revised: 07/07/2008] [Accepted: 07/08/2008] [Indexed: 01/12/2023]
Abstract
Neovascularization may contribute to functional recovery after neural injury. Combination treatment of stroke with a nitric oxide donor, (Z)-1-[N-(2-aminoethyl)-N-(2-ammonioethyl) amino] diazen-1-ium-1, 2-diolate (DETA-NONOate) and bone marrow stromal cells promotes functional recovery. However, the mechanisms underlying functional improvement have not been elucidated. In this study, we tested the hypothesis that combination treatment upregulates angiopoietin-1 and its receptor Tie2 in the ischemic brain and bone marrow stromal cells, thereby enhancing cerebral neovascularization after stroke. Adult wild type male C57BL/6 mice were i.v. administered PBS, bone marrow stromal cells 5x10(5), DETA-NONOate 0.4 mg/kg or combination DETA-NONOate with bone marrow stromal cells (n=12/group) after middle cerebral artery occlusion. Combination treatment significantly upregulated angiopoietin-1/Tie2 and tight junction protein (occludin) expression, and increased the number, diameter and perimeter of blood vessels in the ischemic brain compared with vehicle control (mean+ or -S.E., P<0.05). In vitro, DETA-NONOate significantly increased angiopoietin-1/Tie2 protein (n=6/group) and Tie2 mRNA (n=3/group) expression in bone marrow stromal cells. DETA-NONOate also significantly increased angiopoietin-1 protein (n=6/group) and mRNA (n=3/group) expression in mouse brain endothelial cells (P<0.05). Angiopoietin-1 mRNA (n=3/group) was significantly increased in mouse brain endothelial cells treated with DETA-NONOate in combination with bone marrow stromal cell-conditioned medium compared with cells treated with bone marrow stromal cell-conditioned medium or DETA-NONOate alone. Mouse brain endothelial cell capillary tube-like formation assays (n=6/group) showed that angiopoietin-1 peptide, the supernatant of bone marrow stromal cells and DETA-NONOate significantly increased capillary tube formation compared with vehicle control. Combination treatment significantly increased capillary tube formation compared with DETA-NONOate treatment alone. Inhibition of angiopoietin-1 significantly attenuated combination treatment-induced tube formation. Our data indicated that combination treatment of stroke with DETA-NONOate and bone marrow stromal cells promotes neovascularization, which is at least partially mediated by upregulation of the angiopoietin-1/Tie2 axis.
Collapse
|
367
|
Liu Z, Li Y, Zhang X, Savant-Bhonsale S, Chopp M. Contralesional axonal remodeling of the corticospinal system in adult rats after stroke and bone marrow stromal cell treatment. Stroke 2008; 39:2571-7. [PMID: 18617661 DOI: 10.1161/strokeaha.107.511659] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Motor recovery after stroke is associated with neuronal reorganization in bilateral hemispheres. We investigated contralesional corticospinal tract remodeling in the brain and spinal cord in rats after stroke and treatment of bone marrow stromal cells. METHODS Adult male Wistar rats were subjected to permanent right middle cerebral artery occlusion. Phosphate-buffered saline or bone marrow stromal cells were injected into a tail vein 1 day postischemia. An adhesive removal test was performed weekly to monitor functional recovery. Threshold currents of intracortical microstimulation on the left motor cortex for evoking bilateral forelimb movements were measured 6 weeks after stroke. When intracortical microstimulation was completed, biotinylated dextran amine was injected into the left motor cortex to anterogradely label the corticospinal tract. At 4 days before euthanization, pseudorabies virus-152-EGFP and 614-mRFP were injected into left or right forelimb extensor muscles, respectively. All animals were euthanized 8 weeks after stroke. RESULTS In normal rats (n=5), the corticospinal tract showed a unilateral innervation pattern. In middle cerebral artery occlusion rats (n=8), our data demonstrated that: 1) stroke reduced the stimulation threshold evoking ipsilateral forelimb movement; 2) EGFP-positive pyramidal neurons were increased in the left intact cortex, which were labeled from the left stroke-impaired forelimb; and 3) biotinylated dextran amine-labeled contralesional axons sprouted into the denervated spinal cord. Bone marrow stromal cells significantly enhanced all 3 responses (n=8, P<0.05). CONCLUSIONS Our data demonstrated that corticospinal tract fibers originating from the contralesional motor cortex sprout into the denervated spinal cord after stroke and bone marrow stromal cells treatment, which may contribute to functional recovery.
Collapse
Affiliation(s)
- Zhongwu Liu
- Neurology Research, E&R Building, Room 3056, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI 48202, USA
| | | | | | | | | |
Collapse
|
368
|
Zhang J, Li Y, Zheng X, Gao Q, Liu Z, Qu R, Borneman J, Elias SB, Chopp M. Bone marrow stromal cells protect oligodendrocytes from oxygen-glucose deprivation injury. J Neurosci Res 2008; 86:1501-10. [PMID: 18214988 DOI: 10.1002/jnr.21617] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Oligodendrocyte (OLG) damage leads to demyelination, which is frequently observed in ischemic cerebrovascular diseases. In this study, we investigated the effect of bone marrow stromal cells (BMSCs) on OLGs subjected to oxygen-glucose deprivation (OGD). N20.1 cells (mouse OLG cell line) were transferred into an anaerobic chamber for 3 hr in glucose-free and serum-free medium. After OGD incubation, OLG cultures were divided into the following groups: 1) OGD alone, 2) OLG cocultured with BMSCs, 3) treatment with the phosphoinostide 3-kinase (PI3k) inhibitor LY294002, 4) LY294002-treated OLGs with BMSC cocultured, and 5) anti-p75 antibody-treated OLGs. After an additional 3 hr of reoxygenation incubation, OLG viability and apoptosis were measured. The mRNA expression in the BMSCs and OLGs was analyzed using quantitative real-time PCR (RT-PCR). Serine/threonine-specific protein kinase (Akt), phosphorylated Akt (p-Akt), p75, and caspase 3 protein expressions in OLGs were measured by Western blot. Our results suggest that BMSCs produce growth factors, activate the Akt pathway, and increase the survival of OLGs. BMSCs also reduce p75 and caspase 3 expressions in the OGD-OLGs, which leads to decreased OLG apoptosis. BMSCs participate in OLG protection that may occur with promoting growth factors/PI3K/Akt and inhibiting the p75/caspase pathways. Our study provides insight into white matter damage and the therapeutic benefits of BMSC-based remyelinating therapy after stroke and demyelinating diseases.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan 48202, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
369
|
Zhang RL, Zhang ZG, Chopp M. Ischemic stroke and neurogenesis in the subventricular zone. Neuropharmacology 2008; 55:345-52. [PMID: 18632119 DOI: 10.1016/j.neuropharm.2008.05.027] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Revised: 04/10/2008] [Accepted: 05/20/2008] [Indexed: 01/18/2023]
Abstract
The subventricular zone (SVZ) of the lateral ventricle contains neural stem and progenitor cells that generate neuroblasts, which migrate to the olfactory bulb where they differentiate into interneurons. Ischemic stroke induces neurogenesis in the SVZ and these cells migrate to the boundary of the ischemic lesion. This article reviews current data on cytokinetics, signaling pathways and vascular niche that are involved in processes of proliferation, differentiation, and migration of neural progenitor cells after stroke.
Collapse
Affiliation(s)
- Rui Lan Zhang
- Department of Neurology, Henry Ford Health Sciences Center, Detroit, MI 48202, USA
| | | | | |
Collapse
|
370
|
Neuhuber B, Swanger SA, Howard L, Mackay A, Fischer I. Effects of plating density and culture time on bone marrow stromal cell characteristics. Exp Hematol 2008; 36:1176-85. [PMID: 18495329 DOI: 10.1016/j.exphem.2008.03.019] [Citation(s) in RCA: 156] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2007] [Revised: 02/29/2008] [Accepted: 03/24/2008] [Indexed: 01/14/2023]
Abstract
OBJECTIVE Bone marrow stromal cells (MSC) are multipotent adult stem cells that have emerged as promising candidates for cell therapy in disorders including cardiac infarction, stroke, and spinal cord injury. While harvesting methods used by different laboratories are relatively standard, MSC culturing protocols vary widely. This study is aimed at evaluating the effects of initial plating density and total time in culture on proliferation, cell morphology, and differentiation potential of heterogeneous MSC cultures and more homogeneous cloned subpopulations. MATERIALS AND METHODS Rat MSC were plated at 20, 200, and 2000 cells/cm(2) and grown to 50% confluency. The numbers of population doublings and doubling times were determined within and across multiple passages. Changes in cell morphology and differentiation potential to adipogenic, chondrogenic, and osteogenic lineages were evaluated and compared among early, intermediate, and late passages, as well as between heterogeneous and cloned MSC populations. RESULTS We found optimal cell growth at a plating density of 200 cells/cm(2). Cultures derived from all plating densities developed increased proportions of flat cells over time. Assays for chondrogenesis, osteogenesis, and adipogenesis showed that heterogeneous MSC plated at all densities sustained the potential for all three mesenchymal phenotypes through at least passage 5; the flat subpopulation lost adipogenic and chondrogenic potential. CONCLUSION Our findings suggest that the initial plating density is not critical for maintaining a well-defined, multipotent MSC population. Time in culture, however, affects cell characteristics, suggesting that cell expansion should be limited, especially until the specific characteristics of different MSC subpopulations are better understood.
Collapse
Affiliation(s)
- Birgit Neuhuber
- Drexel University College of Medicine, Department of Neurobiology and Anatomy, Philadelphia, PA, USA.
| | | | | | | | | |
Collapse
|
371
|
Zhang ZX, Guan LX, Zhang K, Zhang Q, Dai LJ. A combined procedure to deliver autologous mesenchymal stromal cells to patients with traumatic brain injury. Cytotherapy 2008; 10:134-9. [PMID: 18368592 DOI: 10.1080/14653240701883061] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND There is increasing evidence of therapeutic benefits from bone marrow (BM)-derived mesenchymal stromal cells (MSC) in various animal models with neurologic disorders. It is of great interest to apply the approach to clinical patients, i.e. to take the investigations from laboratory bench to the patient's bedside. This clinical trial was performed to assess the safety and feasibility of a combined procedure to deliver autologous MSC to patients with traumatic brain injury. METHODS MSC were isolated by BM aspiration and expanded in culture. Seven patients received autologous cell transplantation. A primary administration of 10(7)-10(9) cells was applied directly to the injured area during the cranial operation; a second dose of 10(8)-10(10) cells was infused intravenously. All patients were followed up regularly for 6 months. RESULTS There was no immediate or delayed toxicity related to the cell administration within the 6-month follow-up period. Neurologic function was significantly improved at 6 months after cell therapy. DISCUSSION The procedure used is safe and feasible at ordinary medical facilities without additional invasive procedures for the patient. The combined cell delivery procedure is expected to enhance the engraftment efficacy of transplanted cells at injured brain tissue, thereby promoting neurologic recover.
Collapse
Affiliation(s)
- Z-X Zhang
- Department of Neurosurgery, Weifang People's Hospital, Weifang Medical College, Weifang, PR China
| | | | | | | | | |
Collapse
|
372
|
Systematic neuronal and muscle induction systems in bone marrow stromal cells: the potential for tissue reconstruction in neurodegenerative and muscle degenerative diseases. Med Mol Morphol 2008; 41:14-9. [PMID: 18470676 DOI: 10.1007/s00795-007-0389-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2007] [Accepted: 10/30/2007] [Indexed: 12/21/2022]
Abstract
Because bone marrow stromal cells (MSCs) are easily accessible from both healthy donors and patients and can be expanded on a therapeutic scale, they have attracted attention for cell-based therapy. Benefits of MSCs have been discussed mainly from two aspects: one is their tissue protective and immunomodulatory effects, and the other is their capability under specific manipulations to differentiate into various cell types. In this review, their differentiation into functional neural and muscle cell lineages is the focus, and their potential to the application for tissue reconstruction in neurodegenerative and muscle degenerative diseases is discussed.
Collapse
|
373
|
Abstract
Cells of the central nervous system were once thought to be incapable of regeneration. This dogma has been challenged in the last decade with studies showing new, migrating stem cells in the brain in many rodent injury models and findings of new neurones in the human hippocampus in adults. Moreover, there are reports of bone marrow-derived cells developing neuronal and vascular phenotypes and aiding in repair of injured brain. These findings have fuelled excitement and interest in regenerative medicine for neurological diseases, arguably the most difficult diseases to treat. There are numerous proposed regenerative approaches to neurological diseases. These include cell therapy approaches in which cells are delivered intracerebrally or are infused by an intravenous or intra-arterial route; stem cell mobilization approaches in which endogenous stem and progenitor cells are mobilized by cytokines such as granulocyte colony stimulatory factor (GCSF) or chemokines such as SDF-1; trophic and growth factor support, such as delivering brain-derived neurotrophic factor (BDNF) or glial-derived neurotrophic factor (GDNF) into the brain to support injured neurones; these approaches may be used together to maximize recovery. While initially, it was thought that cell therapy might work by a 'cell replacement' mechanism, a large body of evidence is emerging that cell therapy works by providing trophic or 'chaperone' support to the injured tissue and brain. Angiogenesis and neurogenesis are coupled in the brain. Increasing angiogenesis with adult stem cell approaches in rodent models of stroke leads to preservation of neurones and improved functional outcome. A number of stem and progenitor cell types has been proposed as therapy for neurological disease ranging from neural stem cells to bone marrow derived stem cells to embryonic stem cells. Any cell therapy approach to neurological disease will have to be scalable and easily commercialized if it will have the necessary impact on public health. Currently, bone marrow-derived cell populations such as the marrow stromal cell, multipotential progenitor cells, umbilical cord stem cells and neural stem cells meet these criteria the best. Of great clinical significance, initial evidence suggests these cell types may be delivered by an allogeneic approach, so strict tissue matching may not be necessary. The most immediate impact on patients will be achieved by making use of the trophic support capability of cell therapy and not by a cell replacement mechanism.
Collapse
Affiliation(s)
- D C Hess
- Department of Neurology, Medical College of Georgia, Augusta, GA 30912, USA.
| | | |
Collapse
|
374
|
Marcoli M, Candiani S, Tonachini L, Monticone M, Mastrogiacomo M, Ottonello A, Cervetto C, Paluzzi P, Maura G, Pestarino M, Cancedda R, Castagnola P. In vitro modulation of gamma amino butyric acid (GABA) receptor expression by bone marrow stromal cells. Pharmacol Res 2008; 57:374-82. [PMID: 18467116 DOI: 10.1016/j.phrs.2008.03.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2007] [Revised: 03/26/2008] [Accepted: 03/27/2008] [Indexed: 01/14/2023]
Abstract
Bone marrow stromal cells (BMSC) have the capability to undergo a change of morphology, reminiscent of neuronal cells, after exposure to an inductive medium. These induced BMSC-derived neuron-like (BDNL) cells express several neuronal markers, including Microtubule-Associated Protein Tau, Neurofilament M, and Nestin as revealed by immunocytochemistry analysis. To assess whether the induction process has possible functional relevance, we have focused our attention on the expression of neurotransmitter receptors. In particular, we show that the expression of GABA(A) subunits alpha1, beta2/3, epsilon and GABA(B1) mRNAs is greatly enhanced in BMSC by the induction treatment. Similar results were obtained from rat skin fibroblasts subjected to the same induction protocol, with the exception for the GABA(B2) transcript that was expressed only by BMSC and BDNL. The presence of both GABA(B1) and GABA(B2) subunits in BDNL cells suggests that functional GABA(B) receptors might be assembled: we indeed found that a functional GABA(B) receptor, negatively linked to cyclic AMP production, is expressed in BDNL. Therefore, we suggest that BMSC can be converted into cells equipped with appropriate receptors coupled to transduction mechanisms, potentially responding to a specific neurotransmitter.
Collapse
Affiliation(s)
- Manuela Marcoli
- Dip. Medicina Sperimentale, Sezione di Farmacologia e Tossicologia, Università degli Studi di, Genova, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
375
|
Sheth RN, Manzano G, Li X, Levi AD. Transplantation of human bone marrow-derived stromal cells into the contused spinal cord of nude rats. J Neurosurg Spine 2008; 8:153-62. [PMID: 18248287 DOI: 10.3171/spi/2008/8/2/153] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECT Human bone marrow stromal cells (hMSCs) constitute a potential source of pluripotent stem cells. In the present study, hMSCs were transplanted into an area of spinal cord contusion in nude rats to determine their survival, differentiation, potential for neuroprotection, and influence on axonal growth and functional recovery. METHODS Twenty-nine animals received 6 x 10(5) hMSCs in 6 microl medium 1 week after a contusion, while 14 control animals received an injection of 6 microl medium alone. Basso-Beattie-Bresnahan (BBB) tests were performed weekly. The spinal cords were collected at 6 weeks posttransplantation for histological analysis and assessment of tissue injury. RESULTS Immunostaining with anti-human mitochondria antibody and pretransplantation labeling with green fluorescent protein demonstrated that the grafted hMSCs survived and were capable of achieving a flattened appearance in the grafted area; however, none of the transplanted cells stained positively for human-specific neuronal, anti-neurofilament H or glial fibrillary acidic protein within the sites of engraftment. While neuronal or astrocytic differentiation was not seen, cells lining blood vessels in the vicinity of the transplant stained positively for anti-human endothelium CD105 antibody. Staining for anti-neurofilament H antibody demonstrated abundant axonlike structures around the transplanted area in the hMSC group. Tissue sparing analysis showed that animals with grafted hMSCs had a smaller area of contusion cyst compared with controls, but there was no significant difference between the two groups in BBB scores. CONCLUSIONS The grafted hMSCs survived for > or = 6 weeks posttransplantation, although they did not differentiate into neural or glial cells. Cells with human endothelial characteristics were observed. Spinal cord-injured rats grafted with hMSCs had smaller contusion cavities, which did not have a significant influence on functional recovery.
Collapse
Affiliation(s)
- Rishi N Sheth
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | | | | |
Collapse
|
376
|
Ross JJ, Verfaillie CM. Evaluation of neural plasticity in adult stem cells. Philos Trans R Soc Lond B Biol Sci 2008; 363:199-205. [PMID: 17282993 PMCID: PMC2605495 DOI: 10.1098/rstb.2006.2021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The role of stem cells has long been known in reproductive organs and various tissues including the haematopoietic system and skin. During the last decade, stem cells have also been identified in other organs, including the nervous system, both during development and in post-natal life. More recently, evidence has been presented that stem cells thought to be responsible for the generation of mature differentiated cells of one organ, such as haematopoietic stem cells, may have the ability to also differentiate across lineages and contribute to tissues other than haematopoietic cells, including neuronal tissue, suggesting that easily accessible stem cells sources may one day be useful in the therapy of ischaemic (stroke) and also degenerative diseases of the nervous system. Here, we will evaluate the validity of such claims based on a number of criteria we believe need to be fulfilled to definitively conclude that certain stem cells can give rise to functional neural cells that might be suitable for therapy of neural disorders.
Collapse
Affiliation(s)
- Jeffrey J Ross
- Stem Cell Institute, Cell Biology and Development, University of Minnesota Medical SchoolMN 55455, USA
- Department of Genetics, Cell Biology and Development, University of Minnesota Medical SchoolMN 55455, USA
| | - Catherine M Verfaillie
- Stem Cell Institute, Cell Biology and Development, University of Minnesota Medical SchoolMN 55455, USA
- Division of Hematology, Oncology, and Transplantation, Cell Biology and Development, University of Minnesota Medical SchoolMN 55455, USA
- Author for correspondence ()
| |
Collapse
|
377
|
Andrews EM, Tsai SY, Johnson SC, Farrer JR, Wagner JP, Kopen GC, Kartje GL. Human adult bone marrow-derived somatic cell therapy results in functional recovery and axonal plasticity following stroke in the rat. Exp Neurol 2008; 211:588-92. [PMID: 18440506 DOI: 10.1016/j.expneurol.2008.02.027] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Revised: 01/18/2008] [Accepted: 02/22/2008] [Indexed: 10/22/2022]
Abstract
Stroke is the leading cause of adult disability in the United States. To date there is no satisfactory treatment for stroke once neuronal damage has occurred. Human adult bone marrow-derived somatic cells (hABM-SC) represent a homogenous population of CD49c/CD90 co-positive, non-hematopoietic cells that have been shown to secrete therapeutically relevant trophic factors and to support axonal growth in a rodent model of spinal cord injury. Here we demonstrate that treatment with hABM-SC after ischemic stroke in adult rats results in recovery of forelimb function on a skilled motor test, and that this recovery is positively correlated with increased axonal outgrowth of the intact, uninjured corticorubral tract. While the complete mechanism of repair is still unclear, we conclude that enhancement of structural neuroplasticity from uninjured brain areas is one mechanism by which hABM-SC treatment after stroke leads to functional recovery.
Collapse
Affiliation(s)
- E M Andrews
- Department of Cell Biology, Neurobiology, and Anatomy, Loyola University Chicago, 2160 S. First Avenue, Maywood, IL 60153, USA.
| | | | | | | | | | | | | |
Collapse
|
378
|
Bouchez G, Sensebé L, Vourc'h P, Garreau L, Bodard S, Rico A, Guilloteau D, Charbord P, Besnard JC, Chalon S. Partial recovery of dopaminergic pathway after graft of adult mesenchymal stem cells in a rat model of Parkinson's disease. Neurochem Int 2008; 52:1332-42. [PMID: 18372079 DOI: 10.1016/j.neuint.2008.02.003] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Accepted: 02/06/2008] [Indexed: 11/29/2022]
Abstract
Cellular therapy with adult stem cells appears as an opportunity for treatment of Parkinson's disease. To validate this approach, we studied the effects of transplantation of rat adult bone-marrow mesenchymal stem cells in a rat model of Parkinson's disease. Animals were unilaterally lesioned in the striatum with 6-hydroxydopamine. Two weeks later, group I did not undergo grafting, group II underwent sham grafting, group III was intra-striatal grafted with cells cultured in an enriched medium and group IV was intra-striatal grafted with cells cultured in a standard medium. Rotational amphetamine-induced behavior was measured weekly until animals were killed 6 weeks later. One week after graft, the number of rotations/min was stably decreased by 50% in groups III and IV as compared with groups I and II. At 8 weeks post-lesion, the density of dopaminergic markers in the nerve terminals and cell bodies, i.e. immunoreactive tyrosine hydroxylase, membrane dopamine transporter and vesicular monoamine transporter-2 was significantly higher in group III as compared with group I. Moreover, using microdialysis studies, we observed that while the rate of pharmacologically induced release of dopamine was significantly reduced in lesioned versus intact striatum in no grafted rats, it was similar in both sides in animals transplanted with mesemchymal stem cells. These data demonstrate that graft of adult mesemchymal stem cells reduces behavioral effects induced by 6-hydroxydopamine lesion and partially restores the dopaminergic markers and vesicular striatal pool of dopamine. This cellular approach might be a restorative therapy in Parkinson's disease.
Collapse
Affiliation(s)
- Gaëlle Bouchez
- INSERM U930, Laboratory Biophysique médicale & pharmaceutique, UFR Pharmacie, 31 avenue Monge, Tours, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
379
|
Yaghoobi MM, Mahani MT. NGF and BDNF expression drop off in neurally differentiated bone marrow stromal stem cells. Brain Res 2008; 1203:26-31. [PMID: 18313646 DOI: 10.1016/j.brainres.2008.01.086] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2007] [Revised: 12/30/2007] [Accepted: 01/23/2008] [Indexed: 01/01/2023]
Abstract
Bone marrow stromal stem cells (BMSC) express two neurotrophins nerve growth factor (NGF) and brain derived growth factor (BDNF) constitutively and can be differentiated into neuronal-like cells and used to treat neural injuries and diseases. The neurotrophins are required for repair of neural tissues. However, it is not evident whether these cells supply the sufficient amounts of the functional growth factors following neuronal differentiation. This study investigates the expression of NGF, BDNF and their processing enzymes Prohormone convertases (PC) Furin, PC5 and PC6 by Real-time RT-PCR during neural differentiation of rat BMSC. The results showed that all inspected processing enzymes are expressed in the cells. The expression of NGF, BDNF and PC5 decreases following differentiation. In addition, BMSCs express Survivin, an anti-apoptotic gene; however, the differentiated cells reduce its expression similar to two neurotrophins, which could make them susceptible to apoptotic death.
Collapse
Affiliation(s)
- Mohammad Mehdi Yaghoobi
- Department of Biotechnology, Research Institute of Environmental Sciences, International Centre for Science, High Technology & Environmental Sciences, Kerman, Iran.
| | | |
Collapse
|
380
|
Volk SW, Radu A, Zhang L, Liechty KW. Stromal progenitor cell therapy corrects the wound-healing defect in the ischemic rabbit ear model of chronic wound repair. Wound Repair Regen 2008; 15:736-47. [PMID: 17971020 DOI: 10.1111/j.1524-475x.2007.00277.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Chronic wounds create a formidable clinical problem resulting in considerable morbidity and healthcare expenditure. The etiology for wound healing impairment appears to be multifactorial; however, ischemia is a common factor in most types of chronic wounds. Ideal therapy for such wounds would be to correct deficiencies in growth factors and matrix components and provide cellular precursors required for timely wound closure. We hypothesized that stromal progenitor cell (SPC) therapy could correct the ischemic wound-healing defect through both direct and indirect mechanisms. To test this hypothesis, we used the ischemic rabbit ear model of chronic wound healing. We found that treatment of the wounds with SPCs was able to reverse the ischemic wound-healing impairment, with improved granulation tissue formation and reepithelialization compared with vehicle or bone marrow mononuclear cell controls. In vitro, SPCs were found to produce factors involved in angiogenesis and reepithelialization, and extracellular matrix components, providing evidence for both direct and indirect mechanisms for the observed correction of the healing impairment in these wounds. Treatment of ischemic wounds with SPCs can dramatically improve wound healing and provides a rationale for further studies focused on SPCs as a potential cellular therapy in impaired wound healing.
Collapse
Affiliation(s)
- Susan W Volk
- Department of Clinical Studies, School of Veterinary Medicine, The University of Pennsylvania, Philadelphia, Pennyslvania, USA
| | | | | | | |
Collapse
|
381
|
Neurotrophic Schwann-cell factors induce neural differentiation of bone marrow stromal cells. Neuroreport 2008; 18:1713-7. [PMID: 17921874 DOI: 10.1097/wnr.0b013e3282f0d3b0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Neural transdifferentiation of bone marrow stromal cells has been questioned, because cell fusion could explain the development of new cell types, misinterpreted as transdifferentiated cells. We performed here cocultures of bone marrow stromal cells and Schwann cells, without possibility that both cell types can establish contact. In these conditions, bone marrow stromal cells expressed nestin 4 h after beginning cocultures, and strong expression of neuronal markers was disclosed at 72 h, increasing at 1 and 2 weeks. Our results support that neural transdifferentiation of bone marrow stromal cells is induced by soluble factors provided by glial cells, and suggest that cell fusion should not be significant when local bone marrow stromal cells administration for neural repair is considered.
Collapse
|
382
|
Miyashita K, Itoh H, Nakao K. The anti-inflammatory and vasculo-neuro-regenerative roles of adrenomedullin in ischemic brain. Inflamm Regen 2008. [DOI: 10.2492/inflammregen.28.105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
383
|
Gordon D, Glover CP, Merrison AM, Uney JB, Scolding NJ. Enhanced green fluorescent protein-expressing human mesenchymal stem cells retain neural marker expression. J Neuroimmunol 2008; 193:59-67. [DOI: 10.1016/j.jneuroim.2007.10.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2007] [Revised: 09/17/2007] [Accepted: 10/12/2007] [Indexed: 12/17/2022]
|
384
|
Chopp M, Li Y. Transplantation of Bone Marrow Stromal Cells for Treatment of Central Nervous System Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 585:49-64. [PMID: 17120776 DOI: 10.1007/978-0-387-34133-0_4] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Michael Chopp
- Department of Neurology, Henry Ford Health Sciences Center, Detroit, Michigan, USA
| | | |
Collapse
|
385
|
Neural transdifferentiation of bone marrow stromal cells obtained by chemical agents is a short-time reversible phenomenon. Neurosci Res 2007; 60:275-80. [PMID: 18164086 DOI: 10.1016/j.neures.2007.11.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2007] [Revised: 10/17/2007] [Accepted: 11/13/2007] [Indexed: 11/21/2022]
Abstract
Bone marrow stromal cells (BMSC) can acquire morphological and immunohistochemical features of neural cells when they are treated with diverse chemical agents, a finding interpreted as result of cell transdifferentiation. With the purpose of a better knowledge of the possible utility of BMSC for strategies of Nervous System (NS) repair, we have studied the morphological and immunohistochemical changes induced in BMSC by chemical agents, in comparison with those that happen when BMSC are co-cultured with Schwann cells. While chemical BMSC transdifferentiation is a short-time reversible phenomenon, BMSC transdifferentiation obtained by Schwann cell-derived neurotrophic factors remains stable after it has been reached. These findings question the possible clinical utility of BMSC trandifferentiation using chemical agents, and support that neural transdifferentiation of BMSC is a biological phenomenon that can be obtained in vivo because of the presence of environmental factors.
Collapse
|
386
|
Spees JL, Whitney MJ, Sullivan DE, Lasky JA, Laboy M, Ylostalo J, Prockop DJ. Bone marrow progenitor cells contribute to repair and remodeling of the lung and heart in a rat model of progressive pulmonary hypertension. FASEB J 2007; 22:1226-36. [PMID: 18032636 DOI: 10.1096/fj.07-8076com] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Infusion of bone marrow stem or progenitor cells may provide powerful therapies for injured tissues such as the lung and heart. We examined the potential of bone marrow-derived (BMD) progenitor cells to contribute to repair and remodeling of lung and heart in a rat monocrotaline (MCT) model of pulmonary hypertension. Bone marrow from green fluorescent protein (GFP)-transgenic male rats was transplanted into GFP-negative female rats. The chimeric animals were injected with MCT to produce pulmonary hypertension. Significant numbers of male GFP-positive BMD cells engrafted in the lungs of MCT-treated rats. Microarray analyses and double-immunohistochemistry demonstrated that many of the cells were interstitial fibroblasts or myofibroblasts, some of the cells were hematopoietic cells, and some were pulmonary epithelial cells (Clara cells), vascular endothelial cells, and smooth muscle cells. A few BMD cells fused with pulmonary cells from the host, but the frequency was low. In the hypertrophied hearts of MCT-treated rats, we found a significant increase in the relative numbers of BMD cells in the right ventricle wall as compared with the left ventricle. Some of the BMD cells in the right ventricle were vascular cells and cardiomyocytes. We report BMD cardiomyocytes with a normal chromosome number, fusion of BMD cells with host cardiomyocytes, and, in some cases, nuclear fusion.
Collapse
Affiliation(s)
- Jeffrey L Spees
- Department of Medicine, Center for Gene Therapy, Tulane University Health Sciences Center, New Orleans, Louisiana, USA
| | | | | | | | | | | | | |
Collapse
|
387
|
Rodríguez-González R, Hurtado O, Sobrino T, Castillo J. Neuroplasticity and cellular therapy in cerebral infarction. Cerebrovasc Dis 2007; 24 Suppl 1:167-80. [PMID: 17971653 DOI: 10.1159/000107393] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Stroke is the second to third most common cause of death in adults, and more than a third of people who survive a stroke will have severe disability. Therapeutic options currently centre on fibrinolytic treatment, but its limitations restrict use to a small proportion of patients. Although a wide range of neuroprotective substances has been effective in experimental models, they have repeatedly failed in clinical trials because of toxicity or loss of effectiveness. Recent strategies based on neuroplasticity and cellular therapy have shown significant efficacy in improving functional recovery in experimental models, although further study is still necessary to clarify how the brain responds to ischaemic damage and is able to reorganize itself in the long term. Although steps must still be taken to ensure the safety and feasibility of treatments based on neuroplasticity and cellular therapy, neurorepair strategies provide promising future therapeutic options for stroke.
Collapse
Affiliation(s)
- Raquel Rodríguez-González
- Clinical Neuroscience Research Laboratory, Division of Vascular Neurology, Department of Neurology, Hospital Clínico Universitario, University of Santiago de Compostela, Santiago de Compostela, Spain
| | | | | | | |
Collapse
|
388
|
Koda M, Kamada T, Hashimoto M, Murakami M, Shirasawa H, Sakao S, Ino H, Yoshinaga K, Koshizuka S, Moriya H, Yamazaki M. Adenovirus vector-mediated ex vivo gene transfer of brain-derived neurotrophic factor to bone marrow stromal cells promotes axonal regeneration after transplantation in completely transected adult rat spinal cord. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2007; 16:2206-14. [PMID: 17885772 PMCID: PMC2140138 DOI: 10.1007/s00586-007-0499-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2007] [Revised: 06/01/2007] [Accepted: 08/28/2007] [Indexed: 01/09/2023]
Abstract
The aim of this study was to evaluate the efficacy in adult rat completely transected spinal cord of adenovirus vector-mediated brain-derived neurotrophic factor (BDNF) ex vivo gene transfer to bone marrow stromal cells (BMSC). BMSC were infected with adenovirus vectors carrying beta-galactosidase (AxCALacZ) or BDNF (AxCABDNF) genes. The T8 segment of spinal cord was removed and replaced by graft containing Matrigel alone (MG group) or Matrigel and BMSC infected by AxCALacZ (BMSC-LacZ group) or AxCABDNF (BMSC-BDNF group). Axons in the graft were evaluated by immunohistochemistry and functional recovery was assessed with BBB locomotor scale. In the BMSC-BDNF group, the number of fibers positive for growth associated protein-43, tyrosine hydroxylase, and calcitonin gene-related peptide was significantly larger than numbers found for the MG and BMSC-LacZ groups. Rats from BMSC-BDNF and BMSC-LacZ groups showed significant recovery of hind limb function compared with MG rats; however, there was no significant difference between groups in degree of functional recovery. These findings demonstrate that adenovirus vector-mediated ex vivo gene transfer of BDNF enhances the capacity of BMSC to promote axonal regeneration in this completely transected spinal cord model; however, BDNF failed to enhance hind limb functional recovery. Further investigation is needed to establish an optimal combination of cell therapy and neurotrophin gene transfer for cases of spinal cord injury.
Collapse
Affiliation(s)
- Masao Koda
- Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
389
|
Lapergue B, Mohammad A, Shuaib A. Endothelial progenitor cells and cerebrovascular diseases. Prog Neurobiol 2007; 83:349-62. [PMID: 17884277 DOI: 10.1016/j.pneurobio.2007.08.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2006] [Revised: 06/26/2007] [Accepted: 08/03/2007] [Indexed: 01/09/2023]
Abstract
Identifying factors that may increase the risk of stroke and assessing if treatment of such conditions may lower that risk are important in the management of cerebrovascular disease. Tobacco smoking, poor diet, hypertension and hyperlipidemia remain the major risk factors, and treatment of these conditions has been shown to significantly reduce stroke. In recent years, research has shown that stem cells from a variety of sources can be used as a tool to study and prevent the events that lead to stroke. In this regard, a population of adult stem cells, called endothelial progenitor cells (EPCs), have been identified in peripheral blood and may play an important role in tissue vascularization and endothelium homeostasis in the adult. Most of the studies on EPCs have been carried out on patients with cardiovascular diseases; however, there is emerging evidence which suggests that the introduction or mobilization of EPCs can restore tissue vascularization even after cerebrovascular diseases (CVD), such as ischemic stroke or intracerebral haemorrhage. In this review, we discuss the present level of knowledge about the characteristics of EPCs, their possible therapeutic role in CVD and how they could alter clinical practice in the future.
Collapse
Affiliation(s)
- Bertrand Lapergue
- Stroke Research Unit, Division of Neurology, Faculty of Medicine and Dentistry, University of Alberta, Alberta, Canada
| | | | | |
Collapse
|
390
|
Qu R, Li Y, Gao Q, Shen L, Zhang J, Liu Z, Chen X, Chopp M. Neurotrophic and growth factor gene expression profiling of mouse bone marrow stromal cells induced by ischemic brain extracts. Neuropathology 2007; 27:355-63. [PMID: 17899689 PMCID: PMC2593420 DOI: 10.1111/j.1440-1789.2007.00792.x] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Treatment of rodents after stroke with bone marrow stromal cells (BMSCs) improves functional outcome. However, the mechanisms underlying this benefit have not been ascertained. This study focused on the contribution of neurotrophic and growth factors produced by BMSCs to therapeutic benefit. Rats were subjected to middle cerebral artery occlusion and the ischemic brain extract supernatant was collected to prepare the conditioned medium. The counterpart normal brain extract from non-ischemic rats was employed as the experimental control. Using microarray assay, we measured the changes of the neurotrophin associated gene expression profile in BMSCs cultured in different media. Furthermore, real-time RT-PCR and fluorescent immunocytochemistry were utilized to validate the gene changes. The morphology of BMSCs, cultured in the ischemic brain-conditioned medium for 12 h, was dramatically altered from a polygonal and flat appearance to a fibroblast-like long and thin cell appearance, compared to those in the normal brain-conditioned medium and the serum replacement medium. Forty-four neurotrophin-associated genes in BMSCs were identified by microarray assay under all three culture media. Twelve out of the 44 genes (7 neurotrophic and growth factor genes, 5 receptor genes) increased in BMSCs cultured in the ischemic brain-conditioned medium compared to the normal brain-conditioned medium. Real time RT-PCR and immunocytochemistry validated that the ischemic brain-conditioned medium significantly increased 6/7 neurotrophic and growth factor genes, compared with the normal brain-conditioned medium. These six genes consisted of fibroblast growth factor 2, insulin-like growth factor 1, vascular endothelial growth factor A, nerve growth factor beta, brain-derived neurotrophic factor and epidermal growth factor. Our results indicate that transplanted BMSCs may work as 'small molecular factories' by secreting neurotrophins, growth factors and other supportive substances after stroke, which may produce therapeutic benefits in the ischemic brain.
Collapse
Affiliation(s)
- Runjiang Qu
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Li
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
| | - Qi Gao
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
| | - Lihong Shen
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
| | - Jing Zhang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
| | - Zhongwu Liu
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
| | - Xiaoguang Chen
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
- Department of Physics, Oakland University, Rochester, Michigan, USA
| |
Collapse
|
391
|
Rice CM, Scolding NJ. Autologous bone marrow stem cells--properties and advantages. J Neurol Sci 2007; 265:59-62. [PMID: 17669432 DOI: 10.1016/j.jns.2007.06.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2007] [Revised: 05/10/2007] [Accepted: 06/14/2007] [Indexed: 10/23/2022]
Abstract
The properties of self-renewal and multi-lineage differentiation make stem cells attractive candidates for use in cellular reparative therapy, particularly in neurological diseases where there is a paucity of treatment options. However, clinical trials using foetal material in Parkinson's disease have been disappointing and highlighted problems associated with the use of embryonic stem cells, including ethical issues and practical concerns regarding teratoma formation. Understandably, this has led investigators to explore alternative sources of stem cells for transplantation. The expression of neuroectodermal markers by cells of bone marrow origin focused attention on these adult stem cells. Although early enthusiasm has been tempered by dispute regarding the validity of reports of in vitro (trans)differentiation, the demonstration of functional benefit in animal models of neurological disease is encouraging. Here we will review some of the required properties of stem cells for use in transplantation therapy with specific reference to the development of bone marrow-derived cells as a source of cells for repair in demyelination.
Collapse
Affiliation(s)
- Claire M Rice
- University of Bristol Institute of Clinical Neurosciences, Department of Neurology, Frenchay Hospital, Bristol BS16 1LE, UK
| | | |
Collapse
|
392
|
Abstract
Recent insights into the function and dysfunction of microglia may inform future therapies to combat neurodegeneration. We hypothesise how different aspects of microglial activity including migration, activation, oxidative response, phagocytosis, proteolysis, and replenishment could be targeted by novel therapeutic approaches. A combined approach is suggested, encompassing opsonization and anti-inflammatory strategies in conjunction with an engineering of microglial precursors. Xenoproteases for bioremediation could be used to enhance intracellular and extracellular proteolytic capacity. The capacity of microglial precursors to cross the blood-brain barrier and to home in on sites of neural damage and inflammation might prove to be particularly useful for future therapeutic strategies.
Collapse
Affiliation(s)
- John Schloendorn
- Biodesign Institute, Arizona State University, Tempe, Arizona 85287, USA.
| | | | | |
Collapse
|
393
|
Chopp M, Li Y, Zhang J. Plasticity and remodeling of brain. J Neurol Sci 2007; 265:97-101. [PMID: 17610903 DOI: 10.1016/j.jns.2007.06.013] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2007] [Revised: 05/15/2007] [Accepted: 06/14/2007] [Indexed: 01/01/2023]
Abstract
The injured brain can be stimulated to amplify its intrinsic restorative processes to improve neurological function. Thus, after stroke, both cell and pharmacological neurorestorative treatments, amplify the induction of brain neurogenesis and angiogenesis, and thereby reduce neurological deficits. In this manuscript, we describe the use of bone marrow mesenchymal cells (MSCs) and erythropoietin (EPO) as examples of cell-based and pharmacological neurorestorative treatments, respectively, for both stroke and a mouse model of experimental autoimmune encephalomyelitis (EAE). We demonstrate that these therapies significantly improve neurological function with treatment initiated after the onset of injury and concomitantly promote brain plasticity. The application of MRI to monitor changes in the injured brain associated with reduction of neurological deficit is also described.
Collapse
Affiliation(s)
- Michael Chopp
- Department of Neurology, Henry Ford Health System, 2799 West Grand Bouleverd, Detroit, MI 48202, USA.
| | | | | |
Collapse
|
394
|
Parr AM, Tator CH, Keating A. Bone marrow-derived mesenchymal stromal cells for the repair of central nervous system injury. Bone Marrow Transplant 2007; 40:609-19. [PMID: 17603514 DOI: 10.1038/sj.bmt.1705757] [Citation(s) in RCA: 331] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Transplantation of bone marrow-derived mesenchymal stromal cells (MSCs) into the injured brain or spinal cord may provide therapeutic benefit. Several models of central nervous system (CNS) injury have been examined, including that of ischemic stroke, traumatic brain injury and traumatic spinal cord injury in rodent, primate and, more recently, human trials. Although it has been suggested that differentiation of MSCs into cells of neural lineage may occur both in vitro and in vivo, this is unlikely to be a major factor in functional recovery after brain or spinal cord injury. Other mechanisms of recovery that may play a role include neuroprotection, creation of a favorable environment for regeneration, expression of growth factors or cytokines, vascular effects or remyelination. These mechanisms are not mutually exclusive, and it is likely that more than one contribute to functional recovery. In light of the uncertainty surrounding the fate and mechanism of action of MSCs transplanted into the CNS, further preclinical studies with appropriate animal models are urgently needed to better inform the design of new clinical trials.
Collapse
Affiliation(s)
- A M Parr
- Department of Surgery, University Health Network and Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | | | | |
Collapse
|
395
|
Mazzini L, Mareschi K, Ferrero I, Vassallo E, Oliveri G, Nasuelli N, Oggioni GD, Testa L, Fagioli F. Stem cell treatment in Amyotrophic Lateral Sclerosis. J Neurol Sci 2007; 265:78-83. [PMID: 17582439 DOI: 10.1016/j.jns.2007.05.016] [Citation(s) in RCA: 164] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2007] [Revised: 04/08/2007] [Indexed: 12/15/2022]
Abstract
Amyotrophic Lateral Sclerosis is a progressive fatal neurodegenerative disease that targets motor neurons. Its origin is unknown but a main role of reactive astrogliosis and microglia activation in the pathogenesis has been recently demonstrated. Surrounding neurons with healthy adjoining cells completely stops motor neuron death in some cases. Hence stem cell transplantation might represent a promising therapeutic strategy. In this study MSCs were isolated from bone marrow of 9 patients with definite ALS. Growth kinetics, immunophenotype, telomere length and karyotype were evaluated during in vitro expansion. No significant differences between donors or patients were observed. The patients received intraspinal injections of autologous MSCs at the thoracic level and monitored for 4 years. No significant acute or late side effects were evidenced. No modification of the spinal cord volume or other signs of abnormal cell proliferation were observed. Four patients show a significant slowing down of the linear decline of the forced vital capacity and of the ALS-FRS score. Our results seem to demonstrate that MSCs represent a good chance for stem cell cell-based therapy in ALS and that intraspinal injection of MSCs is safe also in the long term. A new phase 1 study is carried out to verify these data in a larger number of patients.
Collapse
Affiliation(s)
- Letizia Mazzini
- Department of Neurology, Azienda Ospedaliera, Eastern Piedmont University of Novara, Novara, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
396
|
Syková E, Homola A, Mazanec R, Lachmann H, Konrádová SL, Kobylka P, Pádr R, Neuwirth J, Komrska V, Vávra V, Stulík J, Bojar M. Autologous bone marrow transplantation in patients with subacute and chronic spinal cord injury. Cell Transplant 2007; 15:675-87. [PMID: 17269439 DOI: 10.3727/000000006783464381] [Citation(s) in RCA: 240] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Stem cell transplants into spinal cord lesions may help to improve regeneration and spinal cord function. Clinical studies are necessary for transferring preclinical findings from animal experiments to humans. We investigated the transplantation of unmanipulated autologous bone marrow in patients with transversal spinal cord injury (SCI) with respect to safety, therapeutic time window, implantation strategy, method of administration, and functional improvement. We report data from 20 patients with complete SCI who received transplants 10 to 467 days postinjury. The follow-up examinations were done at 3, 6, and 12 months after implantation by two independent neurologists using standard neurological classification of SCI, including the ASIA protocol, the Frankel score, the recording of motor and somatosensory evoked potentials, and MRI evaluation of lesion size. We compared intra-arterial (via catheterization of a. vertebralis) versus intravenous administration of all mononuclear cells in groups of acute (10-30 days post-SCI, n=7) and chronic patients (2-17 months postinjury, n=13). Improvement in motor and/or sensory functions was observed within 3 months in 5 of 6 patients with intra-arterial application, in 5 of 7 acute, and in 1 of 13 chronic patients. Our case study shows that the implantation of autologous bone marrow cells appears to be safe, as there have been no complications following implantation to date (11 patients followed up for more than 2 years), but longer follow-ups are required to determine that implantation is definitively safe. Also, we cannot yet confirm that the observed beneficial effects were due to the cell therapy. However, the outcomes following transplantation in acute patients, and in one chronic patient who was in stable condition for several months prior to cell implantation, are promising. It is evident that transplantation within a therapeutic window of 3-4 weeks following injury will play an important role in any type of stem cell SCI treatment. Trials involving a larger population of patients and different cell types are needed before further conclusions can be drawn.
Collapse
Affiliation(s)
- Eva Syková
- Center for Cell Therapy and Tissue Repair, Charles University, Prague, Czech Republic.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
397
|
Khalatbary AR, Tiraihi T. Localization of bone marrow stromal cells in injured spinal cord treated by intravenous route depends on the hemorrhagic lesions in traumatized spinal tissues. Neurol Res 2007; 29:21-6. [PMID: 17427270 DOI: 10.1179/016164107x165642] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVES Bone marrow stromal cells (BMSCs) have been reported to improve movement deficit in adult rats with spinal cord injury (SCI). The purpose of this study is to determine the distribution of BMSCs in the spinal cord lesion of the contusion model of SCI. METHODS Laminectomy was carried out at L1 vertebra level and SCI was carried out using the weight drop method. BMSCs were isolated from adult rats, labeled with bromodeoxyuridine (BrdU) and administered intravenously to the rats 1 week after SCI, which were killed after 4 weeks. The non-treated animals were used as negative control, which showed cavitations of the spinal cord after 5 weeks of SCI. Rats in another group were killed immediately and used to study the hemorrhagic lesions. The volume densities (Vv) of the hemorrhage and cavitation were the highest at the site of direct trauma. RESULTS The numerical densities of the transmigrated cells per area (Nat) were as follows: 0.3 +/- 0.2, 3.9 +/- 0.4, 5.4 +/- 0.4, 8.4 +/- 0.5, 5.5 +/- 0.3, 3.6 +/- 0.3 and 0.4 +/- 0.2 at the end and the middle of the thoracic vertebra 13 (T13), the region between T13 and the first lumbar vertebra, the middle of L1, the region between L1 and L2, and the middle and the end of L2 vertebra, respectively. The distribution of Nat at the above regions was a Gaussian model. The volume densities of hemorrhage in the spinal cord taken from the above regions showed that hemorrhage with the highest volume density occurred at the impact site and the volume density declined as the samples taken were more distant from the impact site. DISCUSSION The migration of BMSCs in the injured region depends on the amount of the hemorrhage and damage to blood vessels of the spinal cord.
Collapse
Affiliation(s)
- Ali Reza Khalatbary
- Department of Anatomical Sciences, School of Medical Sciences, Tarbiat Modarres University, Tehran, Iran
| | | |
Collapse
|
398
|
Tsai KJ, Tsai YC, Shen CKJ. G-CSF rescues the memory impairment of animal models of Alzheimer's disease. ACTA ACUST UNITED AC 2007; 204:1273-80. [PMID: 17517969 PMCID: PMC2118601 DOI: 10.1084/jem.20062481] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Most of the current clinical treatments for Alzheimer's disease (AD) are largely symptomatic and can have serious side effects. We have tested the feasibility of using the granulocyte colony-stimulating factor (G-CSF), which is known to mobilize hematopoietic stem cells (HSCs) from the bone marrow into the peripheral blood, as a therapeutic agent for AD. Subcutaneous administration of G-CSF into two different β-amyloid (Aβ)–induced AD mouse models substantially rescued their cognitive/memory functions. The rescue was accompanied by the accumulation of 5-bromo-2′deoxyuridine–positive HSCs, as well as local neurogenesis surrounding the Aβ aggregates. Furthermore, the level of acetylcholine in the brains of Tg2576 mice was considerably enhanced upon G-CSF treatment. We suggest that G-CSF, a drug already extensively used for treating chemotherapy-induced neutropenia, should be pursued as a novel, noninvasive therapeutic agent for the treatment of AD.
Collapse
Affiliation(s)
- Kuen-Jer Tsai
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan, Republic of China
| | | | | |
Collapse
|
399
|
Abstract
Regenerative medicine focuses on new therapies to replace or restore lost, damaged, or aging cells in the human body to restore function. This goal is being realized by collaborative efforts in nonmammalian and human development, stem cell biology, genetics, materials science, bioengineering, and tissue engineering. At present, understanding existing reparative processes in humans and exploring the latent ability to regenerate tissue remains the focus in this field. This review covers recent work in limb regeneration, fetal wound healing, stem cell biology, somatic nuclear transfer, and tissue engineering as a foundation for developing new clinical therapies to augment and stimulate human regeneration.
Collapse
Affiliation(s)
- Geoffrey C Gurtner
- Children's Surgical Research Program, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California 94305, USA.
| | | | | |
Collapse
|
400
|
Uccelli A, Pistoia V, Moretta L. Mesenchymal stem cells: a new strategy for immunosuppression? Trends Immunol 2007; 28:219-26. [PMID: 17400510 DOI: 10.1016/j.it.2007.03.001] [Citation(s) in RCA: 326] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2007] [Revised: 02/26/2007] [Accepted: 03/16/2007] [Indexed: 12/18/2022]
Abstract
In vitro-generated mesenchymal stem cells (MSCs) initially attracted interest for their ability to undergo differentiation toward cells of different lineages. More recently, a major breakthrough was the discovery that MSCs exert a profound inhibitory effect on T cell proliferation in vitro and in vivo. Subsequently, MSCs were shown also to exert similar effects on B cells, dendritic cells and natural killer cells. These results suggested that MSCs could be used to dampen immune-mediated diseases and transplant rejection. It is possible that some of the beneficial effects of MSCs might reflect, in part, the trophic and protective activities they exert on injured cells and tissues, rather than resulting from a true transdifferentiation. In immune-mediated diseases, the protective effects might function in concert with the immunosuppressive and anti-inflammatory activities.
Collapse
Affiliation(s)
- Antonio Uccelli
- Department of Neurosciences, Ophthalmology and Genetics, University of Genova, Genova, Italy
| | | | | |
Collapse
|