151
|
Cai H, Mu Z, Jiang Z, Wang Y, Yang GY, Zhang Z. Hypoxia-controlled matrix metalloproteinase-9 hyperexpression promotes behavioral recovery after ischemia. Neurosci Bull 2015; 31:550-60. [PMID: 25975730 DOI: 10.1007/s12264-015-1533-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 04/17/2015] [Indexed: 01/03/2023] Open
Abstract
Matrix metalloproteinase-9 (MMP-9) plays a beneficial role in the sub-acute phase after ischemic stroke. However, unrestrained MMP-9 may disrupt the blood-brain barrier (BBB), which has limited its use for the treatment of brain ischemia. In the present study, we constructed lentivirus mediated hypoxia-controlled MMP-9 expression and explored its role after stroke. Hypoxia response element (HRE) was used to confine MMP-9 expression only to the hypoxic region of mouse brain after 120-min transient middle cerebral artery occlusion. Lentiviruses were injected into the peri-infarct area on day 7 after transient ischemia. We found hyperexpression of exogenous HRE-MMP-9 under the control of hypoxia, and its expression was mainly located in neurons and astrocytes without aggravation of BBB damage compared to the CMV group. Furthermore, mice in the HRE-MMP-9 group showed the best behavioral recovery compared with the normal saline, GFP, and SB-3CT groups. Therefore, hypoxia-controlled MMP-9 hyperexpression during the sub-acute phase of ischemia may provide a novel promising approach of gene therapy for stroke.
Collapse
Affiliation(s)
- Hongxia Cai
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Neuroscience and Neuroengineering Center, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Zhihao Mu
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Neuroscience and Neuroengineering Center, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Zhen Jiang
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Neuroscience and Neuroengineering Center, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Yongting Wang
- Neuroscience and Neuroengineering Center, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Guo-Yuan Yang
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Neuroscience and Neuroengineering Center, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Zhijun Zhang
- Neuroscience and Neuroengineering Center, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China.
| |
Collapse
|
152
|
Wildburger NC, Wood PL, Gumin J, Lichti CF, Emmett MR, Lang FF, Nilsson CL. ESI-MS/MS and MALDI-IMS Localization Reveal Alterations in Phosphatidic Acid, Diacylglycerol, and DHA in Glioma Stem Cell Xenografts. J Proteome Res 2015; 14:2511-9. [PMID: 25880480 DOI: 10.1021/acs.jproteome.5b00076] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Glioblastoma (GBM) is the most common adult primary brain tumor. Despite aggressive multimodal therapy, the survival of patients with GBM remains dismal. However, recent evidence has demonstrated the promise of bone marrow-derived mesenchymal stem cells (BM-hMSCs) as a therapeutic delivery vehicle for anti-glioma agents due to their ability to migrate or home to human gliomas. While several studies have demonstrated the feasibility of harnessing the homing capacity of BM-hMSCs for targeted delivery of cancer therapeutics, it is now also evident, based on clinically relevant glioma stem cell (GSC) models of GBMs, that BM-hMSCs demonstrate variable tropism toward these tumors. In this study, we compared the lipid environment of GSC xenografts that attract BM-hMSCs (N = 9) with those that do not attract (N = 9) to identify lipid modalities that are conducive to homing of BM-hMSC to GBMs. We identified lipids directly from tissue by matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry (IMS) and electrospray ionization-tandem mass spectrometry (ESI-MS/MS) of lipid extracts. Several species of signaling lipids, including phosphatidic acid (PA 36:2, PA 40:5, PA 42:5, and PA 42:7) and diacylglycerol (DAG 34:0, DAG 34:1, DAG 36:1, DAG 38:4, DAG 38:6, and DAG 40:6), were lower in attracting xenografts. Molecular lipid images showed that PA (36:2), DAG (40:6), and docosahexaenoic acid (DHA) were decreased within tumor regions of attracting xenografts. Our results provide the first evidence for lipid signaling pathways and lipid-mediated tumor inflammatory responses in the homing of BM-hMSCs to GSC xenografts. Our studies provide new fundamental knowledge on the molecular correlates of the differential homing capacity of BM-hMSCs toward GSC xenografts.
Collapse
Affiliation(s)
| | - Paul L Wood
- ∥Department of Physiology and Pharmacology, Lincoln Memorial University, 6965 Cumberland Gap Parkway, Harrogate, Tennessee 37752, United States
| | | | - Cheryl F Lichti
- §UTMB Cancer Center, University of Texas Medical Branch, 301 University Boulevard, Galveston, Texas 77555-1074, United States
| | - Mark R Emmett
- §UTMB Cancer Center, University of Texas Medical Branch, 301 University Boulevard, Galveston, Texas 77555-1074, United States
| | | | - Carol L Nilsson
- §UTMB Cancer Center, University of Texas Medical Branch, 301 University Boulevard, Galveston, Texas 77555-1074, United States
| |
Collapse
|
153
|
Wu Y, Wu J, Ju R, Chen Z, Xu Q. Comparison of intracerebral transplantation effects of different stem cells on rodent stroke models. Cell Biochem Funct 2015; 33:174-82. [PMID: 25914321 PMCID: PMC4687466 DOI: 10.1002/cbf.3083] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/24/2014] [Accepted: 11/07/2014] [Indexed: 12/28/2022]
Abstract
In the present study, induced pluripotent stem cells (iPSCs), induced neural stem cells (iNSCs), mesenchymal stem cells (MSCs) and an immortalized cell line (RMNE6), representing different characteristics of stem cells, were transplanted into normal and/or injured brain areas of rodent stroke models, and their effects were compared to select suitable stem cells for cell replacement stroke therapy. The rat and mice ischaemic models were constructed using the middle cerebral artery occlusion technique. Both electrocoagulation of the artery and the intraluminal filament technique were used. The behaviour changes and fates of grafted stem cells were determined mainly by behaviour testing and immunocytochemistry. Following iPSC transplantation into the corpora striata of normal mice, a tumour developed in the brain. The iNSCs survived well and migrated towards the injured area without differentiation. Although there was no tumourigenesis in the brain of normal or ischaemic mice after the iNSCs were transplanted in the cortices, the behaviour in ischaemic mice was not improved. Upon transplanting MSC and RMNE6 cells into ischaemic rat brains, results similar to iNSCs in mice were seen. However, transplantation of RMNE6 caused a brain tumour. Thus, tumourigenesis and indeterminate improvement of behaviour are challenging problems encountered in stem cell therapy for stroke, and the intrinsic characteristics of stem cells should be remodelled before transplantation. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Yun Wu
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.,Beijing Center of Neural Regeneration and Repair, Capital Medical University, Beijing, China.,Department of Neurobiology, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Major Brain Disorders, Capital Medical University, Beijing, China.,Beijing Municipal Key Laboratory for Neural Regeneration and Repairing, Capital Medical University, Beijing, China
| | - Jianyu Wu
- Department of Cell Biology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Rongkai Ju
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.,Beijing Center of Neural Regeneration and Repair, Capital Medical University, Beijing, China.,Department of Neurobiology, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Major Brain Disorders, Capital Medical University, Beijing, China.,Beijing Municipal Key Laboratory for Neural Regeneration and Repairing, Capital Medical University, Beijing, China
| | - Zhiguo Chen
- Department of Cell Biology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Qunyuan Xu
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.,Beijing Center of Neural Regeneration and Repair, Capital Medical University, Beijing, China.,Department of Neurobiology, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Major Brain Disorders, Capital Medical University, Beijing, China.,Beijing Municipal Key Laboratory for Neural Regeneration and Repairing, Capital Medical University, Beijing, China
| |
Collapse
|
154
|
Cameron SH, Alwakeel AJ, Goddard L, Hobbs CE, Gowing EK, Barnett ER, Kohe SE, Sizemore RJ, Oorschot DE. Delayed post-treatment with bone marrow-derived mesenchymal stem cells is neurorestorative of striatal medium-spiny projection neurons and improves motor function after neonatal rat hypoxia-ischemia. Mol Cell Neurosci 2015; 68:56-72. [PMID: 25828540 DOI: 10.1016/j.mcn.2015.03.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 03/19/2015] [Accepted: 03/27/2015] [Indexed: 02/07/2023] Open
Abstract
Perinatal hypoxia-ischemia is a major cause of striatal injury and may lead to cerebral palsy. This study investigated whether delayed administration of bone marrow-derived mesenchymal stem cells (MSCs), at one week after neonatal rat hypoxia-ischemia, was neurorestorative of striatal medium-spiny projection neurons and improved motor function. The effect of a subcutaneous injection of a high-dose, or a low-dose, of MSCs was investigated in stereological studies. Postnatal day (PN) 7 pups were subjected to hypoxia-ischemia. At PN14, pups received treatment with either MSCs or diluent. A subset of high-dose pups, and their diluent control pups, were also injected intraperitoneally with bromodeoxyuridine (BrdU), every 24h, on PN15, PN16 and PN17. This permitted tracking of the migration and survival of neuroblasts originating from the subventricular zone into the adjacent injured striatum. Pups were euthanized on PN21 and the absolute number of striatal medium-spiny projection neurons was measured after immunostaining for DARPP-32 (dopamine- and cAMP-regulated phosphoprotein-32), double immunostaining for BrdU and DARPP-32, and after cresyl violet staining alone. The absolute number of striatal immunostained calretinin interneurons was also measured. There was a statistically significant increase in the absolute number of DARPP-32-positive, BrdU/DARPP-32-positive, and cresyl violet-stained striatal medium-spiny projection neurons, and fewer striatal calretinin interneurons, in the high-dose mesenchymal stem cell (MSC) group compared to their diluent counterparts. A high-dose of MSCs restored the absolute number of these neurons to normal uninjured levels, when compared with previous stereological data on the absolute number of cresyl violet-stained striatal medium-spiny projection neurons in the normal uninjured brain. For the low-dose experiment, in which cresyl violet-stained striatal medium-spiny neurons alone were measured, there was a lower statistically significant increase in their absolute number in the MSC group compared to their diluent controls. Investigation of behavior in another cohort of animals showed that delayed administration of a high-dose of bone marrow-derived MSCs, at one week after neonatal rat hypoxia-ischemia, improved motor function on the cylinder test. Thus, delayed therapy with a high- or low-dose of adult MSCs, at one week after injury, is effective in restoring the loss of striatal medium-spiny projection neurons after neonatal rat hypoxia-ischemia and a high-dose of MSCs improved motor function.
Collapse
Affiliation(s)
- Stella H Cameron
- Department of Anatomy, Otago School of Medical Sciences, and the Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Amr J Alwakeel
- Department of Anatomy, Otago School of Medical Sciences, and the Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Liping Goddard
- Department of Anatomy, Otago School of Medical Sciences, and the Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Catherine E Hobbs
- Department of Anatomy, Otago School of Medical Sciences, and the Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Emma K Gowing
- Department of Anatomy, Otago School of Medical Sciences, and the Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Elizabeth R Barnett
- Department of Anatomy, Otago School of Medical Sciences, and the Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Sarah E Kohe
- Department of Anatomy, Otago School of Medical Sciences, and the Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Rachel J Sizemore
- Department of Anatomy, Otago School of Medical Sciences, and the Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Dorothy E Oorschot
- Department of Anatomy, Otago School of Medical Sciences, and the Brain Health Research Centre, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
155
|
Dulamea AO. The potential use of mesenchymal stem cells in stroke therapy--From bench to bedside. J Neurol Sci 2015; 352:1-11. [PMID: 25818674 DOI: 10.1016/j.jns.2015.03.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 03/09/2015] [Accepted: 03/10/2015] [Indexed: 12/11/2022]
Abstract
Stroke is the second main cause of morbidity and mortality worldwide. The rationale for the use of mesenchymal stem cells (MSCs) in stroke is based on the capacity of MSCs to secrete a large variety of bioactive molecules such as growth factors, cytokines and chemokines leading to reduction of inflammation, increased neurogenesis from the germinative niches of central nervous system, increased angiogenesis, effects on astrocytes, oligodendrocytes and axons. This review presents the data derived from experimental studies and the evidence available from clinical trials about the use of MSCs in stroke therapy.
Collapse
Affiliation(s)
- Adriana Octaviana Dulamea
- U.M.F. "Carol Davila", Fundeni Clinical Institute, Department of Neurology, 258 Sos. Fundeni, Sector 2, Bucharest, Romania.
| |
Collapse
|
156
|
Yamauchi T, Kuroda Y, Morita T, Shichinohe H, Houkin K, Dezawa M, Kuroda S. Therapeutic effects of human multilineage-differentiating stress enduring (MUSE) cell transplantation into infarct brain of mice. PLoS One 2015; 10:e0116009. [PMID: 25747577 PMCID: PMC4351985 DOI: 10.1371/journal.pone.0116009] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 12/03/2014] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVE Bone marrow stromal cells (BMSCs) are heterogeneous and their therapeutic effect is pleiotropic. Multilineage-differentiating stress enduring (Muse) cells are recently identified to comprise several percentages of BMSCs, being able to differentiate into triploblastic lineages including neuronal cells and act as tissue repair cells. This study was aimed to clarify how Muse and non-Muse cells in BMSCs contribute to functional recovery after ischemic stroke. METHODS Human BMSCs were separated into stage specific embryonic antigen-3-positive Muse cells and -negative non-Muse cells. Immunodeficient mice were subjected to permanent middle cerebral artery occlusion and received transplantation of vehicle, Muse, non-Muse or BMSCs (2.5×104 cells) into the ipsilateral striatum 7 days later. RESULTS Motor function recovery in BMSC and non-Muse groups became apparent at 21 days after transplantation, but reached the plateau thereafter. In Muse group, functional recovery was not observed for up to 28 days post-transplantation, but became apparent at 35 days post-transplantation. On immunohistochemistry, only Muse cells were integrated into peri-infarct cortex and differentiate into Tuj-1- and NeuN-expressing cells, while negligible number of BMSCs and non-Muse cells remained in the peri-infarct area at 42 days post-transplantation. CONCLUSIONS These findings strongly suggest that Muse cells and non-Muse cells may contribute differently to tissue regeneration and functional recovery. Muse cells may be more responsible for replacement of the lost neurons through their integration into the peri-infarct cortex and spontaneous differentiation into neuronal marker-positive cells. Non-Muse cells do not remain in the host brain and may exhibit trophic effects rather than cell replacement.
Collapse
Affiliation(s)
- Tomohiro Yamauchi
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Yasumasa Kuroda
- Department of Stem Cell Biology and Histology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Takahiro Morita
- Department of Stem Cell Biology and Histology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Hideo Shichinohe
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Kiyohiro Houkin
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Mari Dezawa
- Department of Stem Cell Biology and Histology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Satoshi Kuroda
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
- Department of Neurosurgery, Graduate School of Medicine and Pharmacological Science, University of Toyama, Toyama, Japan
- * E-mail:
| |
Collapse
|
157
|
Duan W, Zhang YP, Hou Z, Huang C, Zhu H, Zhang CQ, Yin Q. Novel Insights into NeuN: from Neuronal Marker to Splicing Regulator. Mol Neurobiol 2015; 53:1637-1647. [PMID: 25680637 DOI: 10.1007/s12035-015-9122-5] [Citation(s) in RCA: 194] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 02/01/2015] [Indexed: 01/07/2023]
Abstract
Neuronal nuclei (NeuN) is a well-recognized "marker" that is detected exclusively in post-mitotic neurons and was initially identified through an immunological screen to produce neuron-specific antibodies. Immunostaining evidence indicates that NeuN is distributed in the nuclei of mature neurons in nearly all parts of the vertebrate nervous system. NeuN is highly conserved among species and is stably expressed during specific stages of development. Therefore, NeuN has been considered to be a reliable marker of mature neurons for the past two decades. However, this role has been challenged by recent studies indicating that NeuN staining is variable and even absent during certain diseases and specific physiological states. More importantly, despite the widespread use of the anti-NeuN antibody, the natural identity of the NeuN protein remained elusive for 17 years. NeuN was recently eventually identified as an epitope of Rbfox3, which is a novel member of the Rbfox1 family of splicing factors. This identification might provide a novel perspective on NeuN expression during both physiological and pathological conditions. This review summarizes the current progress on the biochemical identity and biological significance of NeuN and recommends caution when applying NeuN immunoreactivity as a definitive marker of mature neurons in certain diseases and specific physiological states.
Collapse
Affiliation(s)
- Wei Duan
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China
| | - Yu-Ping Zhang
- Department of Pediatrics, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China
| | - Zhi Hou
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China
| | - Chen Huang
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, 37232, USA
| | - He Zhu
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, 37232, USA.,Department of Biological Sciences, Vanderbilt University, Nashville, TN, 37232, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.,The Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN, 37232, USA
| | - Chun-Qing Zhang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China.
| | - Qing Yin
- Department of Rehabilitation and Physical Therapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China.
| |
Collapse
|
158
|
Intranasal delivery of stem cells as therapy for central nervous system disease. Exp Mol Pathol 2015; 98:145-51. [PMID: 25645932 DOI: 10.1016/j.yexmp.2015.01.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 01/29/2015] [Indexed: 12/13/2022]
Abstract
Stem cells, upon entering the CNS, can preferentially migrate into disease foci, where they exert therapeutic effects that compensate for lost tissue, reconstructing damaged neuronal circuitry and establishing in the brain a new microenvironment suitable for cell survival. However, the route of stem cell delivery into the CNS remains a challenge: with systemic administration (e.g., intravenous injection), a fraction of cells may be trapped in other organs than the CNS, while direct CNS injections, e.g., intracerebroventricular or transcranial, are invasive. Intranasal (i.n.) delivery of stem cells, in contrast, can effectively bypass the blood-brain barrier, rapidly enter the CNS, and minimize systemic distribution. I.n. delivery of stem cells may therefore be a safe and non-invasive way of targeting the CNS and would thus be a promising therapeutic option for CNS disease. In this review we discuss the i.n. route for stem cell delivery into the CNS, and the perspectives of i.n. stem cell-based therapy in CNS disease.
Collapse
|
159
|
Nik Ramli NN, Omar N, Husin A, Ismail Z, Siran R. Preconditioning effect of (S)-3,5-dihydroxyphenylglycine on ischemic injury in middle cerebral artery occluded Sprague-Dawley rats. Neurosci Lett 2015; 588:137-41. [PMID: 25562631 DOI: 10.1016/j.neulet.2014.12.062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 12/30/2014] [Accepted: 12/31/2014] [Indexed: 10/24/2022]
Abstract
Glutamate receptors are the integral cellular components associated with excitotoxicity mechanism induced by the ischemic cascade events. Therefore the glutamate receptors have become the major molecular targets of neuroprotective agents in stroke researches. Recent studies have demonstrated that a Group I metabotropic glutamate receptor agonist, (S)-3,5-dihydroxyphenylglycine ((S)-3,5-DHPG) preconditioning elicits neuroprotection in the hippocampal slice cultures exposed to toxic level of N-methyl-d-aspartate (NMDA). We further investigated the preconditioning effects of (S)-3,5-DHPG on acute ischemic stroke rats. One 10 or 100μM of (S)-3,5-DHPG was administered intrathecally to Sprague-Dawley adult male rats, 2h prior to induction of acute ischemic stroke by middle cerebral artery occlusion (MCAO). After 24h, neurological deficits were evaluated by modified stroke severity scores and grid-walking test. All rats were sacrificed and infarct volumes were determined by 2,3,5-triphenyltetrazolium chloride staining. The serum level of neuron-specific enolase (NSE) of each rat was analyzed by enzyme-linked immunosorbent assay (ELISA). One and 10μM of (S)-3,5-DHPG preconditioning in the stroke rats showed significant improvements in motor impairment (P<0.01), reduction in the infarct volume (P<0.01) and reduction in the NSE serum level (P<0.01) compared to the control stroke rats. We conclude that 1 and 10μM (S)-3,5-DHPG preconditioning induced protective effects against acute ischemic insult in vivo.
Collapse
Affiliation(s)
- Nik Nasihah Nik Ramli
- Institute of Medical Molecular Biotechnology, Universiti Teknologi MARA, 47000 Selangor, Malaysia
| | - Nursyazwani Omar
- Institute of Medical Molecular Biotechnology, Universiti Teknologi MARA, 47000 Selangor, Malaysia
| | - Andrean Husin
- Faculty of Dentistry, Universiti Teknologi MARA, 40450, Shah Alam, Selangor, Malaysia; Brain and Neuroscience Communities of Research, Universiti Teknologi MARA, 40450, Shah Alam, Selangor, Malaysia
| | - Zalina Ismail
- Brain Research and Information Network, Centre for Neurocognitive Sciences, Universiti Sains Malaysia, 16150 Kelantan, Malaysia
| | - Rosfaiizah Siran
- Brain and Neuroscience Communities of Research, Universiti Teknologi MARA, 40450, Shah Alam, Selangor, Malaysia.
| |
Collapse
|
160
|
Cell-based therapy for acute organ injury: preclinical evidence and ongoing clinical trials using mesenchymal stem cells. Anesthesiology 2014; 121:1099-121. [PMID: 25211170 DOI: 10.1097/aln.0000000000000446] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Critically ill patients often suffer from multiple organ failures involving lung, kidney, liver, or brain. Genomic, proteomic, and metabolomic approaches highlight common injury mechanisms leading to acute organ failure. This underlines the need to focus on therapeutic strategies affecting multiple injury pathways. The use of adult stem cells such as mesenchymal stem or stromal cells (MSC) may represent a promising new therapeutic approach as increasing evidence shows that MSC can exert protective effects following injury through the release of promitotic, antiapoptotic, antiinflammatory, and immunomodulatory soluble factors. Furthermore, they can mitigate metabolomic and oxidative stress imbalance. In this work, the authors review the biological capabilities of MSC and the results of clinical trials using MSC as therapy in acute organ injuries. Although preliminary results are encouraging, more studies concerning safety and efficacy of MSC therapy are needed to determine their optimal clinical use. (ANESTHESIOLOGY 2014; 121:1099-121).
Collapse
|
161
|
Tang G, Liu Y, Zhang Z, Lu Y, Wang Y, Huang J, Li Y, Chen X, Gu X, Wang Y, Yang GY. Mesenchymal Stem Cells Maintain Blood-Brain Barrier Integrity by Inhibiting Aquaporin-4 Upregulation After Cerebral Ischemia. Stem Cells 2014; 32:3150-62. [DOI: 10.1002/stem.1808] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 07/07/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Guanghui Tang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering; Shanghai Jiao Tong University; Shanghai People's Republic of China
| | - Yanqun Liu
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering; Shanghai Jiao Tong University; Shanghai People's Republic of China
- Department of Neurology, Ruijin Hospital; Shanghai Jiao Tong University School of Medicine; Shanghai People's Republic of China
| | - Zhijun Zhang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering; Shanghai Jiao Tong University; Shanghai People's Republic of China
| | - Yifan Lu
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering; Shanghai Jiao Tong University; Shanghai People's Republic of China
| | - Yang Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering; Shanghai Jiao Tong University; Shanghai People's Republic of China
- Department of Neurology, Ruijin Hospital; Shanghai Jiao Tong University School of Medicine; Shanghai People's Republic of China
| | - Jun Huang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering; Shanghai Jiao Tong University; Shanghai People's Republic of China
| | - Yaning Li
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering; Shanghai Jiao Tong University; Shanghai People's Republic of China
| | - Xiaoyan Chen
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering; Shanghai Jiao Tong University; Shanghai People's Republic of China
| | - Xiang Gu
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering; Shanghai Jiao Tong University; Shanghai People's Republic of China
| | - Yongting Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering; Shanghai Jiao Tong University; Shanghai People's Republic of China
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering; Shanghai Jiao Tong University; Shanghai People's Republic of China
- Department of Neurology, Ruijin Hospital; Shanghai Jiao Tong University School of Medicine; Shanghai People's Republic of China
| |
Collapse
|
162
|
Reparative Therapy for Acute Ischemic Stroke with Allogeneic Mesenchymal Stem Cells from Adipose Tissue: A Safety Assessment. J Stroke Cerebrovasc Dis 2014; 23:2694-2700. [DOI: 10.1016/j.jstrokecerebrovasdis.2014.06.011] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 05/12/2014] [Accepted: 06/15/2014] [Indexed: 12/16/2022] Open
|
163
|
Liu Y, Nie L, Zhao H, Zhang W, Zhang YQ, Wang SS, Cheng L. Conserved dopamine neurotrophic factor-transduced mesenchymal stem cells promote axon regeneration and functional recovery of injured sciatic nerve. PLoS One 2014; 9:e110993. [PMID: 25343619 PMCID: PMC4208796 DOI: 10.1371/journal.pone.0110993] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 09/28/2014] [Indexed: 01/09/2023] Open
Abstract
Peripheral nerve injury (PNI) is a common disease that often results in axonal degeneration and the loss of neurons, ultimately leading to limited nerve regeneration and severe functional impairment. Currently, there are no effective treatments for PNI. In the present study, we transduced conserved dopamine neurotrophic factor (CDNF) into mesenchymal stem cells (MSCs) in collagen tubes to investigate their regenerative effects on rat peripheral nerves in an in vivo transection model. Scanning electron microscopy of the collagen tubes demonstrated their ability to be resorbed in vivo. We observed notable overexpression of the CDNF protein in the distal sciatic nerve after application of CDNF-MSCs. Quantitative analysis of neurofilament 200 (NF200) and S100 immunohistochemistry showed significant enhancement of axonal and Schwann cell regeneration in the group receiving CDNF-MSCs (CDNF-MSCs group) compared with the control groups. Myelination thickness, axon diameter and the axon-to fiber diameter ratio (G-ratio) were significantly higher in the CDNF-MSCs group at 8 and 12 weeks after nerve transection surgery. After surgery, the sciatic functional index, target muscle weight, wet weight ratio of gastrocnemius muscle and horseradish peroxidase (HRP) tracing demonstrated functional recovery. Light and electron microscopy confirmed successful regeneration of the sciatic nerve. The greater numbers of HRP-labeled neuron cell bodies and increased sciatic nerve index values (SFI) in the CDNF-MSCs group suggest that CDNF exerts neuroprotective effects in vivo. We also observed higher target muscle weights and a significant improvement in muscle atrophism in the CDNF-MSCs group. Collectively, these findings indicate that CDNF gene therapy delivered by MSCs is capable of promoting nerve regeneration and functional recovery, likely because of the significant neuroprotective and neurotrophic effects of CDNF and the superior environment offered by MSCs and collagen tubes.
Collapse
Affiliation(s)
- Yi Liu
- Department of Spine Surgery, Qilu Hospital of Shandong University, Jinan, China
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, China
| | - Lin Nie
- Department of Spine Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Hua Zhao
- Department of Spine Surgery, Qilu Hospital of Shandong University, Jinan, China
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, China
| | - Wen Zhang
- Department of Spine Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Yuan-Qiang Zhang
- Department of Spine Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Shuai-Shuai Wang
- Department of Spine Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Lei Cheng
- Department of Spine Surgery, Qilu Hospital of Shandong University, Jinan, China
- * E-mail:
| |
Collapse
|
164
|
Ikegame Y, Yamashita K, Nakashima S, Nomura Y, Yonezawa S, Asano Y, Shinoda J, Hara H, Iwama T. Fate of graft cells: what should be clarified for development of mesenchymal stem cell therapy for ischemic stroke? Front Cell Neurosci 2014; 8:322. [PMID: 25374506 PMCID: PMC4204523 DOI: 10.3389/fncel.2014.00322] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 09/24/2014] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are believed to be promising for cell administration therapy after ischemic stroke. Because of their advantageous characteristics, such as ability of differentiation into neurovascular lineages, avoidance of immunological problems, and abundance of graft cells in mesodermal tissues, studies regarding MSC therapy have increased recently. However, several controversies are yet to be resolved before a worldwide consensus regarding a standard protocol is obtained. In particular, the neuroprotective effects, the rate of cell migration to the lesion, and differentiation direction differ depending on preclinical observations. Analyses of these differences and application of recent developments in stem cell biology or engineering in imaging modality may contribute to identification of criteria for optimal stem cell therapy in which reliable protocols, which control cell quality and include safe administration procedures, are defined for each recovery phase after cerebral ischemia. In this mini review, we examine controversies regarding the fate of grafts and the prospects for advanced therapy that could be obtained through recent developments in stem cell research as direct conversion to neural cells.
Collapse
Affiliation(s)
- Yuka Ikegame
- Department of Neurosurgery, Chubu Medical Center for Prolonged Traumatic Brain Dysfunction Gifu, Japan ; Department of Clinical Brain Sciences, Gifu University Graduate School of Medicine Gifu, Japan ; Department of Cell Signaling, Gifu University Graduate School of Medicine Gifu, Japan
| | - Kentaro Yamashita
- Department of Neurosurgery, Gifu University Graduate School of Medicine Gifu, Japan ; Department of Neurosurgery, Murakami Memorial Hospital, Asahi University Gifu, Japan
| | - Shigeru Nakashima
- Department of Cell Signaling, Gifu University Graduate School of Medicine Gifu, Japan
| | - Yuichi Nomura
- Department of Neurosurgery, Chubu Medical Center for Prolonged Traumatic Brain Dysfunction Gifu, Japan
| | - Shingo Yonezawa
- Department of Neurosurgery, Chubu Medical Center for Prolonged Traumatic Brain Dysfunction Gifu, Japan
| | - Yoshitaka Asano
- Department of Neurosurgery, Chubu Medical Center for Prolonged Traumatic Brain Dysfunction Gifu, Japan ; Department of Clinical Brain Sciences, Gifu University Graduate School of Medicine Gifu, Japan
| | - Jun Shinoda
- Department of Neurosurgery, Chubu Medical Center for Prolonged Traumatic Brain Dysfunction Gifu, Japan ; Department of Clinical Brain Sciences, Gifu University Graduate School of Medicine Gifu, Japan
| | - Hideaki Hara
- Department of Biofunctional Evaluation, Molecular Pharmacology, Gifu Pharmaceutical University Gifu, Japan
| | - Toru Iwama
- Department of Neurosurgery, Gifu University Graduate School of Medicine Gifu, Japan
| |
Collapse
|
165
|
Tang Y, Cai B, Yuan F, He X, Lin X, Wang J, Wang Y, Yang GY. Melatonin Pretreatment Improves the Survival and Function of Transplanted Mesenchymal Stem Cells after Focal Cerebral Ischemia. Cell Transplant 2014; 23:1279-1291. [PMID: 23635511 DOI: 10.3727/096368913x667510] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stem cell (MSC) transplantation has been shown to be beneficial in treating cerebral ischemia. However, such benefit is limited by the low survival of transplanted MSCs in an ischemic microenvironment. Previous studies showed that melatonin pretreatment can increase MSC survival in the ischemic kidney. However, whether it will improve MSC survival in cerebral ischemia is unknown. Our study examined the effect of melatonin pretreatment on MSCs under ischemia-related conditions in vitro and after transplantation into ischemic rat brain. Results showed that melatonin pretreatment greatly increased survival of MSCs in vitro and reduced their apoptosis after transplantation into ischemic brain. Melatonin-treated MSCs (MT-MSCs) further reduced brain infarction and improved neurobehavioral outcomes. Angiogenesis, neurogenesis, and the expression of vascular endothelial growth factor (VEGF) were greatly increased in the MT-MSC-treated rats. Melatonin treatment increased the level of p-ERK1/2 in MSCs, which can be blocked by the melatonin receptor antagonist luzindole. ERK phosphorylation inhibitor U0126 completely reversed the protective effects of melatonin, suggesting that melatonin improves MSC survival and function through activating the ERK1/2 signaling pathway. Thus, stem cells pretreated by melatonin may represent a feasible approach for improving the beneficial effects of stem cell therapy for cerebral ischemia.
Collapse
Affiliation(s)
- Yaohui Tang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute, Shanghai, China.,School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Beibei Cai
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute, Shanghai, China.,School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Falei Yuan
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute, Shanghai, China.,School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaosong He
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute, Shanghai, China.,School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaojie Lin
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute, Shanghai, China.,School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jixian Wang
- Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yongting Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute, Shanghai, China.,School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute, Shanghai, China.,School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
166
|
Turner RJ, Vink R. NK1 tachykinin receptor treatment is superior to capsaicin pre-treatment in improving functional outcome following acute ischemic stroke. Neuropeptides 2014; 48:267-72. [PMID: 25151181 DOI: 10.1016/j.npep.2014.07.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 07/12/2014] [Accepted: 07/22/2014] [Indexed: 12/15/2022]
Abstract
Previous results from our laboratory have shown that blockade of the substance P (SP) pathway with an NK1 tachykinin receptor antagonist significantly reduces blood brain barrier breakdown, cerebral edema and functional deficits following ischemic stroke. However, it is unclear whether removal of all neuropeptides is more efficacious than blocking SP alone. As such, the aim of the present study was to determine the effect of neuropeptide depletion with capsaicin pre-treatment on functional outcome following acute ischemic stroke in rats. Animals received 125 mg/kg of capsaicin or equal volume of saline vehicle, administered subcutaneously over a 3-day period. At 14 days following treatment animals were subject to 2h of middle cerebral artery occlusion followed by reperfusion. A subset of animals was treated with an NK1 tachykinin receptor antagonist (NAT) or vehicle at 4h after the onset of stroke only. The functional outcome of animals was assessed for a 7-day period following stroke using a rotarod device, the bilateral asymmetry test, modified neurological severity score, open field and angleboard. Although capsaicin pre-treatment improved outcome, treatment with an NK1 tachykinin receptor antagonist was superior in improving post-stroke functional outcome. This data suggests that some neuropeptides may play a beneficial role following stroke, whilst others such as SP are deleterious.
Collapse
Affiliation(s)
- Renée J Turner
- Adelaide Centre for Neuroscience Research, School of Medical Sciences, University of Adelaide, Adelaide, SA, Australia.
| | - Robert Vink
- Adelaide Centre for Neuroscience Research, School of Medical Sciences, University of Adelaide, Adelaide, SA, Australia; Divsion of Health Sciences, The University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
167
|
Shimada Y, Tanaka R, Shimura H, Yamashiro K, Urabe T, Hattori N. Phosphorylation enhances recombinant HSP27 neuroprotection against focal cerebral ischemia in mice. Neuroscience 2014; 278:113-21. [DOI: 10.1016/j.neuroscience.2014.07.073] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 07/18/2014] [Accepted: 07/31/2014] [Indexed: 11/29/2022]
|
168
|
Yu H, Chen P, Yang Z, Luo W, Pi M, Wu Y, Wang L. Electro-acupuncture at Conception and Governor vessels and transplantation of umbilical cord blood-derived mesenchymal stem cells for treating cerebral ischemia/reperfusion injury. Neural Regen Res 2014; 9:84-91. [PMID: 25206747 PMCID: PMC4146313 DOI: 10.4103/1673-5374.125334] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2013] [Indexed: 12/24/2022] Open
Abstract
Mesenchymal stem cell transplantation is a novel means of treating cerebral ischemia/reperfusion, and can promote angiogenesis and neurological functional recovery. Acupuncture at Conception and Governor vessels also has positive effects as a treatment for cerebral ischemia/reperfusion. Therefore, we hypothesized that electro-acupuncture at Conception and Governor vessels plus mesenchymal stem cell transplantation may have better therapeutic effects on the promotion of angiogenesis and recovery of neurological function than either treatment alone. In the present study, human umbilical cord blood-derived mesenchymal stem cells were isolated, cultured, identified and intracranially transplanted into the striatum and subcortex of rats at 24 hours following cerebral ischemia/reperfusion. Subsequently, rats were electro-acupunctured at Conception and Governor vessels at 24 hours after transplantation. Modified neurological severity scores and immunohistochemistry findings revealed that the combined interventions of electro-acupuncture and mesenchymal stem cell transplantation clearly improved neurological impairment and up-regulated vascular endothelial growth factor expression around the ischemic focus. The combined intervention provided a better outcome than mesenchymal stem cell transplantation alone. These findings demonstrate that electro-acupuncture at Conception and Governor vessels and mesenchymal stem cell transplantation have synergetic effects on promoting neurological function recovery and angiogenesis in rats after cerebral ischemia/reperfusion.
Collapse
Affiliation(s)
- Haibo Yu
- Affiliated Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong Province, China
| | - Pengdian Chen
- Affiliated Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong Province, China
| | - Zhuoxin Yang
- Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong Province, China
| | - Wenshu Luo
- Affiliated Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong Province, China
| | - Min Pi
- Affiliated Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong Province, China
| | - Yonggang Wu
- Affiliated Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong Province, China
| | - Ling Wang
- Affiliated Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong Province, China
| |
Collapse
|
169
|
Li N, Wang P, Ma XL, Wang J, Zhao LJ, Du L, Wang LY, Wang XR, Liu KD. Effect of bone marrow stromal cell transplantation on neurologic function and expression of VEGF in rats with focal cerebral ischemia. Mol Med Rep 2014; 10:2299-305. [PMID: 25174606 PMCID: PMC4214338 DOI: 10.3892/mmr.2014.2502] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 03/05/2014] [Indexed: 12/03/2022] Open
Abstract
There is evidence that the transplantation of mesenchymal stem cells into rat models of cerebral ischemia reduces ischemic damage; however, the mechanism remains to be elucidated. The present study aimed to assess the effect of transplantation of human bone marrow stromal cells (hBMSCs) on neurologic function and the expression of vascular endothelial growth factor (VEGF) in a rat model of focal cerebral ischemia. The left middle cerebral artery of adult Wistar rats was occluded for 90 min using a nylon thread, followed by reperfusion for 1 h. hBMSCs labeled with 5-bromo-2-deoxyuridine (BrdU) were stereotaxically injected into the ischemic boundary zone. Behavioral analysis using the Neurological Severity Score (NSS) was conducted on days 1, 3, 7 and 28, and a histologic evaluation was performed simultaneously. VEGF was detected by immunofluorescence staining and western blot analysis. Fifty rats were divided equally into five groups: Normal control, sham-operated, operated (no transplantation), Dulbecco’s medium Eagle’s medium (DMEM)-injected (received only serum-free DMEM), and hBMSC-transplanted. The hBMSC-transplanted group showed significantly improved behavioral recovery compared with the operated and DMEM-transplanted groups on days 3, 7 and 28. Histological examination showed that transplanted cells migrated from the injection site into nearby areas including the cortex. Expression of VEGF was significantly greater in the hBMSC group compared with the other four groups on each assessment day. The expression of VEGF was found to be beneficial for functional recovery following cerebral ischemic injury and hBMSC transplantation stimulated the expression of VEGF. Transplantation of BMSCs may be a promising therapeutic strategy for treating cerebral infarction.
Collapse
Affiliation(s)
- Nan Li
- Department of Neurology, The First Bethune Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ping Wang
- The Otolaryngology Research Institute, The First Bethune Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xue-Ling Ma
- Department of Neurology, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Juan Wang
- Department of Neurology, The First Bethune Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Li-Jing Zhao
- Department of Neurology, The First Bethune Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Li Du
- Department of Neurology, The First Bethune Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Li-Ya Wang
- Department of Neurology, The First Bethune Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xin-Rui Wang
- Amphixenosis Research Institute, Jilin University, Changchun, Jilin 130000, P.R. China
| | - Kang-Ding Liu
- Department of Neurology, The First Bethune Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
170
|
Banerjee S, Bentley P, Hamady M, Marley S, Davis J, Shlebak A, Nicholls J, Williamson DA, Jensen SL, Gordon M, Habib N, Chataway J. Intra-Arterial Immunoselected CD34+ Stem Cells for Acute Ischemic Stroke. Stem Cells Transl Med 2014; 3:1322-30. [PMID: 25107583 DOI: 10.5966/sctm.2013-0178] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Treatment with CD34+ hematopoietic stem/progenitor cells has been shown to improve functional recovery in nonhuman models of ischemic stroke via promotion of angiogenesis and neurogenesis. We aimed to determine the safety and feasibility of treatment with CD34+ cells delivered intra-arterially in patients with acute ischemic stroke. This was the first study in human subjects. We performed a prospective, nonrandomized, open-label, phase I study of autologous, immunoselected CD34+ stem/progenitor cell therapy in patients presenting within 7 days of onset with severe anterior circulation ischemic stroke (National Institutes of Health Stroke Scale [NIHSS] score≥8). CD34+ cells were collected from the bone marrow of the subjects before being delivered by catheter angiography into the ipsilesional middle cerebral artery. Eighty-two patients with severe anterior circulation ischemic stroke were screened, of whom five proceeded to treatment. The common reasons for exclusion were age>80 years (n=19); medical instability (n=17), and significant carotid stenosis (n=13). The procedure was well tolerated in all patients, and no significant treatment-related adverse effects occurred. All patients showed improvements in clinical functional scores (Modified Rankin Score and NIHSS score) and reductions in lesion volume during a 6-month follow-up period. Autologous CD34+ selected stem/progenitor cell therapy delivered intra-arterially into the infarct territory can be achieved safely in patients with acute ischemic stroke. Future studies that address eligibility criteria, dosage, delivery site, and timing and that use surrogate imaging markers of outcome are desirable before larger scale clinical trials.
Collapse
Affiliation(s)
- Soma Banerjee
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - Paul Bentley
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - Mohammad Hamady
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - Stephen Marley
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - John Davis
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - Abdul Shlebak
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - Joanna Nicholls
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - Deborah A Williamson
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - Steen L Jensen
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - Myrtle Gordon
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - Nagy Habib
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - Jeremy Chataway
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| |
Collapse
|
171
|
Abstract
On average, every four minutes an individual dies from a stroke, accounting for 1 out of every 18 deaths in the United States. Approximately 795,000 Americans have a new or recurrent stroke each year, with just over 600,000 of these being first attack [1]. There have been multiple animal models of stroke demonstrating that novel therapeutics can help improve the clinical outcome. However, these results have failed to show the same outcomes when tested in human clinical trials. This review will discuss the current in vivo animal models of stroke, advantages and limitations, and the rationale for employing these animal models to satisfy translational gating items for examination of neuroprotective, as well as neurorestorative strategies in stroke patients. An emphasis in the present discussion of therapeutics development is given to stem cell therapy for stroke.
Collapse
|
172
|
Wang RY, Lin XJ, Yang GY, Gao PJ, Shen GX. Effect of hirulog-like peptide on middle cerebral artery occlusion-induced brain injury in mice. Neuroscience 2014; 277:568-76. [PMID: 25065624 DOI: 10.1016/j.neuroscience.2014.07.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 07/12/2014] [Accepted: 07/16/2014] [Indexed: 01/21/2023]
Abstract
Hirulog-like peptide (HLP) and low-molecular-weight heparin (LMWH) are thrombin inhibitor peptides. Our previous study demonstrated that HLP could reduce vascular neointimal formation or restenosis in animals undergoing balloon catheter injury in the carotid artery. However, the function of HLP during ischemic stroke is largely unknown. The present study investigated the effect of HLP on brain injury, which was induced by suture of middle cerebral artery occlusion in mice. Mice were divided into four groups, which included a sham group and three treatment groups. Ischemia was induced by transient suture insertion into the middle cerebral artery for 90 min, and mice were either treated with saline, HLP or LMWH. Infarct volume, neurologic deficits and apoptotic factors were measured following 1-14 days of ischemia. We demonstrated that HLP intravenous injection alleviated brain infarct volume and improved neurologic outcomes (p<0.05). HLP decreased levels of protease-activated receptor-1 (PAR-1), caspase-3, malondialdehyde (MDA) and Bcl-2-associated X protein (Bax), increased the activities of catalase and B cell lymphoma-2 (Bcl-2), and improved the ratio of Bcl-2/Bax compared with the control (p<0.05). This study indicates that HLP and LMWH reduced infarct volume and improved neurobehavioral outcomes induced by transient middle cerebral artery occlusion (tMCAO). In addition, HLP had a beneficial effect on the regulation of the thrombin receptor and key apoptosis regulators in the mouse brain. These results suggest that HLP may be a potential alternative therapy for arterial occlusion-induced cerebral ischemia.
Collapse
Affiliation(s)
- R-Y Wang
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension and Department of Hypertension, Ruijin Hospital, Shanghai 200025, China; The Laboratory of Vascular Biology and Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, China; Shanghai Institute of Hypertension, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - X-J Lin
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China; Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200025, China
| | - G-Y Yang
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China; Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200025, China.
| | - P-J Gao
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension and Department of Hypertension, Ruijin Hospital, Shanghai 200025, China; The Laboratory of Vascular Biology and Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, China; Shanghai Institute of Hypertension, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China.
| | - G X Shen
- Departments of Internal Medicine and Physiology, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
173
|
Simone C, Nizzardo M, Rizzo F, Ruggieri M, Riboldi G, Salani S, Bucchia M, Bresolin N, Comi GP, Corti S. iPSC-Derived neural stem cells act via kinase inhibition to exert neuroprotective effects in spinal muscular atrophy with respiratory distress type 1. Stem Cell Reports 2014; 3:297-311. [PMID: 25254343 PMCID: PMC4176534 DOI: 10.1016/j.stemcr.2014.06.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Revised: 06/04/2014] [Accepted: 06/05/2014] [Indexed: 12/01/2022] Open
Abstract
Spinal muscular atrophy with respiratory distress type 1 (SMARD1) is a motor neuron disease caused by mutations in the IGHMBP2 gene, without a cure. Here, we demonstrate that neural stem cells (NSCs) from human-induced pluripotent stem cells (iPSCs) have therapeutic potential in the context of SMARD1. We show that upon transplantation NSCs can appropriately engraft and differentiate in the spinal cord of SMARD1 animals, ameliorating their phenotype, by protecting their endogenous motor neurons. To evaluate the effect of NSCs in the context of human disease, we generated human SMARD1-iPSCs motor neurons that had a significantly reduced survival and axon length. Notably, the coculture with NSCs ameliorate these disease features, an effect attributable to the production of neurotrophic factors and their dual inhibition of GSK-3 and HGK kinases. Our data support the role of iPSC as SMARD1 disease model and their translational potential for therapies in motor neuron disorders.
Collapse
Affiliation(s)
- Chiara Simone
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Monica Nizzardo
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Federica Rizzo
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Margherita Ruggieri
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Giulietta Riboldi
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Sabrina Salani
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Monica Bucchia
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Nereo Bresolin
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Giacomo P Comi
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy.
| |
Collapse
|
174
|
Tang Y, Wang J, Lin X, Wang L, Shao B, Jin K, Wang Y, Yang GY. Neural stem cell protects aged rat brain from ischemia-reperfusion injury through neurogenesis and angiogenesis. J Cereb Blood Flow Metab 2014; 34:1138-47. [PMID: 24714034 PMCID: PMC4083376 DOI: 10.1038/jcbfm.2014.61] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 03/04/2014] [Accepted: 03/05/2014] [Indexed: 12/28/2022]
Abstract
Neural stem cells (NSCs) show therapeutic potential for ischemia in young-adult animals. However, the effect of aging on NSC therapy is largely unknown. In this work, NSCs were transplanted into aged (24-month-old) and young-adult (3-month-old) rats at 1 day after stroke. Infarct volume and neurobehavioral outcomes were examined. The number of differentiated NSCs was compared in aged and young-adult ischemic rats and angiogenesis and neurogenesis were also determined. We found that aged rats developed larger infarcts than young-adult rats after ischemia (P<0.05). The neurobehavioral outcome was also worse for aged rats comparing with young-adult rats. Brain infarction and neurologic deficits were attenuated after NSC transplantation in both aged and young-adult rats. The number of survived NSCs in aged rats was similar to that of the young-adult rats (P>0.05) and most of them were differentiated into glial fibrillary acidic protein(+) (GFAP(+)) cells. More importantly, angiogenesis and neurogenesis were greatly enhanced in both aged and young-adult rats after transplantation compared with phosphate-buffered saline (PBS) control (P<0.05), accompanied by increased expression of vascular endothelial growth factor (VEGF). Our results showed that NSC therapy reduced ischemic brain injury, along with increased angiogenesis and neurogenesis in aged rats, suggesting that aging-related microenvironment does not preclude a beneficial response to NSCs transplantation during cerebral ischemia.
Collapse
Affiliation(s)
- Yaohui Tang
- Neuroscience and Neuroengineering Center, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Jixian Wang
- Department of Neurology, Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaojie Lin
- Neuroscience and Neuroengineering Center, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Liuqing Wang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research the First Affiliated Hospital, Department of Neurology, Wenzhou Medical College, Zhejiang, China
| | - Bei Shao
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research the First Affiliated Hospital, Department of Neurology, Wenzhou Medical College, Zhejiang, China
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Yongting Wang
- Neuroscience and Neuroengineering Center, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- 1] Neuroscience and Neuroengineering Center, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China [2] Department of Neurology, Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
175
|
Yang Z, Zhu L, Li F, Wang J, Wan H, Pan Y. Bone marrow stromal cells as a therapeutic treatment for ischemic stroke. Neurosci Bull 2014; 30:524-34. [PMID: 24817388 DOI: 10.1007/s12264-013-1431-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 12/12/2013] [Indexed: 12/15/2022] Open
Abstract
Cerebral ischemia remains the most frequent cause of death and quality-of-life impairments due to neurological deficits, and accounts for the majority of total healthcare costs. However, treatments for cerebral ischemia are limited. Over the last decade, bone marrow stromal cell (BMSC) therapy has emerged as a particularly appealing option, as it is possible to help patients even when initiated days or even weeks after the ischemic insult. BMSCs are a class of multipotent, self-renewing cells that give rise to differentiated progeny when implanted into appropriate tissues. Therapeutic effects of BMSC treatment for ischemic stroke, including sensory and motor recovery, have been reported in pre-clinical studies and clinical trials. In this article, we review the recent progress in BMSC-based therapy for ischemic stroke, focusing on the route of delivery and pre-processing of BMSCs. Selecting an optimal delivery route is of particular importance. The ideal approach, as well as the least risky, for translational applications still requires further identification. Appropriate preprocessing of BMSCs or combination therapy has the benefit of achieving the maximum possible restoration. Further pre-clinical studies are required to determine the time-window for transplantation and the appropriate dosage of cells.
Collapse
Affiliation(s)
- Zizhen Yang
- Department of Neurology, First Hospital and Clinical College, Harbin Medical University, Harbin, 150001, China
| | | | | | | | | | | |
Collapse
|
176
|
Jiang W, Liang G, Li X, Li Z, Gao X, Feng S, Wang X, Liu M, Liu Y. Intracarotid transplantation of autologous adipose-derived mesenchymal stem cells significantly improves neurological deficits in rats after MCAo. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2014; 25:1357-1366. [PMID: 24469290 DOI: 10.1007/s10856-014-5157-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 01/12/2014] [Indexed: 06/03/2023]
Abstract
We aimed to evaluate whether adipose-derived mesenchymal stem cells (ADMSCs) that were transplanted via internal carotid can improve the neurological function after acute ischemic stroke and explore the underlying mechanisms. Total 40 adult Sprague-Dawley rats were subjected to transient (1.5 h) middle cerebral artery occlusion (MCAo) to induce ischemia/reperfusion injury. These rats were randomly divided into two groups with 20 ones in each group, which were intracarotid-injected with autologous ADMSCs (2.0 × 10(6)) and saline (control) at day 3 after MCAo, respectively. Behavioral tests (adhesive-removal and modified neurological severity score) were performed before and after MCAo. Histology was used to evaluate the ischemia lesion volume and pathological changes. The apoptosis and astroglial reactivity were determined by TUNEL and glial fibrillary acidic protein (GFAP) staining, respectively. Besides, we applied immunofluorescence to identify the distribution of ADMSCs and the neural makers (NeuN and GFAP) expressed by them under confocal microscope. Significant improvement of neurological deficits was observed in rats transplanted with ADMSCs when compared to controls. But there was no obvious difference on ischemia lesion volume between these two groups. The injected ADMSCs migrated to the brain infarct region and mainly localized in the ischemic core and boundary zone of the lesion, which can express NeuN and GFAP in the brain. In addition, autologous transplantation of ADMSCs significantly attenuated astroglial reactivity, inhibited cellular apoptosis and promoted cellular proliferation. Our data indicated that intracarotid transplantation of autologous ADMSCs had the potential therapeutic application for ischemic stroke.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Neurosurgery, The General Hospital of Shenyang Military Region, No. 83 Wenhua Road, Shenyang, 110840, Liaoning, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
177
|
Appaix F, Nissou MF, Sanden BVD, Dreyfus M, Berger F, Issartel JP, Wion D. Brain mesenchymal stem cells: The other stem cells of the brain? World J Stem Cells 2014; 6:134-143. [PMID: 24772240 PMCID: PMC3999771 DOI: 10.4252/wjsc.v6.i2.134] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 02/20/2014] [Indexed: 02/06/2023] Open
Abstract
Multipotent mesenchymal stromal cells (MSC), have the potential to differentiate into cells of the mesenchymal lineage and have non-progenitor functions including immunomodulation. The demonstration that MSCs are perivascular cells found in almost all adult tissues raises fascinating perspectives on their role in tissue maintenance and repair. However, some controversies about the physiological role of the perivascular MSCs residing outside the bone marrow and on their therapeutic potential in regenerative medicine exist. In brain, perivascular MSCs like pericytes and adventitial cells, could constitute another stem cell population distinct to the neural stem cell pool. The demonstration of the neuronal potential of MSCs requires stringent criteria including morphological changes, the demonstration of neural biomarkers expression, electrophysiological recordings, and the absence of cell fusion. The recent finding that brain cancer stem cells can transdifferentiate into pericytes is another facet of the plasticity of these cells. It suggests that the perversion of the stem cell potential of pericytes might play an even unsuspected role in cancer formation and tumor progression.
Collapse
|
178
|
Ultrasound-Microbubble Transplantation of Bone Marrow Stromal Cells Improves Neurological Function after Forebrain Ischemia in Adult Mice. Cell Biochem Biophys 2014; 70:499-504. [DOI: 10.1007/s12013-014-9947-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
179
|
Tseng LS, Chen SH, Lin MT, Lin YC. Transplantation of human dental pulp-derived stem cells protects against heatstroke in mice. Cell Transplant 2014; 24:921-37. [PMID: 24612725 DOI: 10.3727/096368914x678580] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Stem cells from human exfoliated deciduous tooth pulp (SHED) is a promising approach for the treatment of stroke and spinal cord injury. In this study, we investigated the therapeutic effects of SHED for the treatment of multiple organ (including brain, particularly hypothalamus) injury in heatstroke mice. ICR male mice were exposed to whole body heating (WBH; 41.2°C, relative humidity 50-55%, for 1 h) and then returned to normal room temperature (26°C). We observed that intravenous administration of SHED immediately post-WBH exhibited the following therapeutic benefits for recovery after heatstroke: (a) inhibition of WBH-induced neurologic and thermoregulatory deficits; (b) reduction of WBH-induced ischemia, hypoxia, and oxidative damage to the brain (particularly the hypothalamus); (c) attenuation of WBH-induced increased plasma levels of systemic inflammatory response molecules, such as tumor necrosis factor-α and intercellular adhesion molecule-1; (d) improvement of WBH-induced hypothalamo-pituitary-adrenocortical (HPA) axis activity (as reflected by enhanced plasma levels of both adrenocorticotrophic hormone and corticosterone); and (e) attenuation of WBH-induced multiple organ apoptosis as well as lethality. In conclusion, post-WBH treatment with SHED reduced induction of proinflammatory cytokines and oxidative radicals, enhanced plasma induction of both adrenocorticotrophic hormone and corticosterone, and improved lethality in mouse heatstroke. The protective effect of SHED may be related to a decreased inflammatory response, decreased oxidative stress, and an increased HPA axis activity following the WBH injury.
Collapse
Affiliation(s)
- Ling-Shu Tseng
- School of Dentistry, Kaohsiung Medical University, Kaohsiung City, Taiwan
| | | | | | | |
Collapse
|
180
|
Mao L, Huang M, Chen SC, Li YN, Xia YP, He QW, Wang MD, Huang Y, Zheng L, Hu B. Endogenous endothelial progenitor cells participate in neovascularization via CXCR4/SDF-1 axis and improve outcome after stroke. CNS Neurosci Ther 2014; 20:460-8. [PMID: 24581269 DOI: 10.1111/cns.12238] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 01/17/2014] [Accepted: 01/18/2014] [Indexed: 11/26/2022] Open
Abstract
AIM To study whether endogenous endothelial progenitor cells (EPCs) are involved in neovascularization after stroke. MATERIALS AND METHODS Animal stroke models were established by subjecting male SD rats to permanent middle cerebral artery occlusion (pMCAO). Vessels in ischemic boundary zone (IBZ) were stained with antibody against laminin at 1 to 21 days after pMCAO. EPCs and newly formed vessels were identified by staining with special markers. After inhibiting recruitment of EPCs with AMD3100, a CXCR4 antagonist, endogenous EPCs, capillary density, cerebral blood flow (CBF) in IBZ, and neurobehavioral functions were assessed by staining, FITC-dextran, laser-Doppler perfusion monitor, and neurologic severity score. RESULTS After pMCAO, vessels were found in IBZ at day 3, reaching a peak at day 14. The change in number of laminin-positive cells showed a similar pattern with that of vessels. Apart from few endothelial cells, most of laminin-positive cells were endogenous EPCs. After treatment with AMD3100, the number of endogenous EPCs, capillary density, and CBF in IBZ were significantly reduced, and neurobehavioral functions were worse as compared with the normal saline group. CONCLUSIONS Our findings suggested that endogenous EPCs participated in the neovascularization via CXCR4/SDF-1 axis after pMCAO and mobilizing endogenous EPCs could be a treatment alternative for stroke.
Collapse
Affiliation(s)
- Ling Mao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Gold EM, Su D, López-Velázquez L, Haus DL, Perez H, Lacuesta GA, Anderson AJ, Cummings BJ. Functional assessment of long-term deficits in rodent models of traumatic brain injury. Regen Med 2014; 8:483-516. [PMID: 23826701 DOI: 10.2217/rme.13.41] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Traumatic brain injury (TBI) ranks as the leading cause of mortality and disability in the young population worldwide. The annual US incidence of TBI in the general population is estimated at 1.7 million per year, with an estimated financial burden in excess of US$75 billion a year in the USA alone. Despite the prevalence and cost of TBI to individuals and society, no treatments have passed clinical trial to clinical implementation. The rapid expansion of stem cell research and technology offers an alternative to traditional pharmacological approaches targeting acute neuroprotection. However, preclinical testing of these approaches depends on the selection and characterization of appropriate animal models. In this article we consider the underlying pathophysiology for the focal and diffuse TBI subtypes, discuss the existing preclinical TBI models and functional outcome tasks used for assessment of injury and recovery, identify criteria particular to preclinical animal models of TBI in which stem cell therapies can be tested for safety and efficacy, and review these criteria in the context of the existing TBI literature. We suggest that 2 months post-TBI is the minimum period needed to evaluate human cell transplant efficacy and safety. Comprehensive review of the published TBI literature revealed that only 32% of rodent TBI papers evaluated functional outcome ≥1 month post-TBI, and only 10% evaluated functional outcomes ≥2 months post-TBI. Not all published papers that evaluated functional deficits at a minimum of 2 months post-TBI reported deficits; hence, only 8.6% of overall TBI papers captured in this review demonstrated functional deficits at 2 months or more postinjury. A 2-month survival and assessment period would allow sufficient time for differentiation and integration of human neural stem cells with the host. Critically, while trophic effects might be observed at earlier time points, it will also be important to demonstrate the sustainability of such an effect, supporting the importance of an extended period of in vivo observation. Furthermore, regulatory bodies will likely require at least 6 months survival post-transplantation for assessment of toxicology/safety, particularly in the context of assessing cell abnormalities.
Collapse
Affiliation(s)
- Eric M Gold
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine 2030 Gross Hall, CA 92697-1705, USA
| | | | | | | | | | | | | | | |
Collapse
|
182
|
Gonzales-Portillo GS, Sanberg PR, Franzblau M, Gonzales-Portillo C, Diamandis T, Staples M, Sanberg CD, Borlongan CV. Mannitol-enhanced delivery of stem cells and their growth factors across the blood-brain barrier. Cell Transplant 2014; 23:531-9. [PMID: 24480552 PMCID: PMC4083632 DOI: 10.3727/096368914x678337] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Ischemic brain injury in adults and neonates is a significant clinical problem with limited therapeutic interventions. Currently, clinicians have only tPA available for stroke treatment and hypothermia for cerebral palsy. Owing to the lack of treatment options, there is a need for novel treatments such as stem cell therapy. Various stem cells including cells from embryo, fetus, perinatal, and adult tissues have proved effective in preclinical and small clinical trials. However, a limiting factor in the success of these treatments is the delivery of the cells and their by-products (neurotrophic factors) into the injured brain. We have demonstrated that mannitol, a drug with the potential to transiently open the blood-brain barrier and facilitate the entry of stem cells and trophic factors, as a solution to the delivery problem. The combination of stem cell therapy and mannitol may improve therapeutic outcomes in adult stroke and neonatal cerebral palsy.
Collapse
Affiliation(s)
- Gabriel S. Gonzales-Portillo
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Paul R. Sanberg
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Max Franzblau
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Chiara Gonzales-Portillo
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Theo Diamandis
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Meaghan Staples
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Cyndy D. Sanberg
- Saneron CCEL Therapeutics, Saneron CCEL Therapeutics, Inc., Tampa, FL, USA
| | - Cesar V. Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
183
|
Abstract
Quantitative measurement of blood-brain barrier (BBB) permeability using MRI and its application to cerebral ischemia are reviewed. Measurement of BBB permeability using MRI has been employed to evaluate ischemic damage during acute and subacute phases of stroke and to predict hemorrhagic transformation. There is also an emerging interest on the development and use of MRI to monitor vascular structural changes and angiogenesis during stroke recovery. In this review, we describe MRI BBB permeability and susceptibility-weighted MRI measurements and its applications to evaluate ischemic damage during the acute and subacute phases of stroke and vascular remodeling during stroke recovery.
Collapse
|
184
|
Sakar M, Korkusuz P, Demirbilek M, Cetinkaya DU, Arslan S, Denkbaş EB, Temuçin ÇM, Bilgiç E, Hazer DB, Bozkurt G. The effect of poly(3-hydroxybutyrate-co-3- hydroxyhexanoate) (PHBHHx) and human mesenchymal stem cell (hMSC) on axonal regeneration in experimental sciatic nerve damage. Int J Neurosci 2014; 124:685-96. [PMID: 24350993 DOI: 10.3109/00207454.2013.876636] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This study is designed to evaluate the treatment effect of poly(3-hydroxybutyrate-co-3-hydroxyhexanoate) (PHBHHx) and human mesenchymal stem cells (hMSC) on axonal regeneration in experimental rat sciatic nerve damage, and compare the results of this modality with autologous nerve grafting. In Spraque-Dawley albino rats, 10-mm-long experimental nerve gaps were created. Three groups were constituted, the gap was repaired with autologous nerve graft (autograft group), PHBHHx nerve graft alone (PHBHHx alone group), and PHBHHx nerve graft with hMSCs inside (PHBHHx with hMSC group), respectively. The results were evaluated with functional recovery, electrophysiological evaluation, and histological evaluation either with light microscopy and transmission electron microscopy for axonal regeneration and myelin formation. In functional evaluation, autograft and PHBHHx with hMSC groups showed functional improvement with time, whereas PHBHHx alone group did not. Electrophysiological evaluation showed better results in autograft and PHBHHx with hMSC groups when compared to PHBHHx alone group. There was no statistical difference between autograft and PHBHHx with hMSC groups. Histological evaluation showed regenerated axons in each group. Autograft group was better than the others, and PHBHHx with hMSC group was better than PHBHHx alone group both for axonal regeneration and myelin formation. This study showed that the nerve grafts which were prepared from PHBHHx with oriented nanofiber three-dimensional surfaces aided to nerve regeneration, either used alone or with hMSC. PHBHHx provided better nerve regeneration when used with hMSCs inside than alone, and reached the same statistical treatment effect in functional evaluation and electrophysiological evaluation when compared to autografting.
Collapse
|
185
|
Liu X, Ye R, Yan T, Yu SP, Wei L, Xu G, Fan X, Jiang Y, Stetler RA, Liu G, Chen J. Cell based therapies for ischemic stroke: from basic science to bedside. Prog Neurobiol 2013; 115:92-115. [PMID: 24333397 DOI: 10.1016/j.pneurobio.2013.11.007] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 11/11/2013] [Accepted: 11/26/2013] [Indexed: 12/20/2022]
Abstract
Cell therapy is emerging as a viable therapy to restore neurological function after stroke. Many types of stem/progenitor cells from different sources have been explored for their feasibility and efficacy for the treatment of stroke. Transplanted cells not only have the potential to replace the lost circuitry, but also produce growth and trophic factors, or stimulate the release of such factors from host brain cells, thereby enhancing endogenous brain repair processes. Although stem/progenitor cells have shown a promising role in ischemic stroke in experimental studies as well as initial clinical pilot studies, cellular therapy is still at an early stage in humans. Many critical issues need to be addressed including the therapeutic time window, cell type selection, delivery route, and in vivo monitoring of their migration pattern. This review attempts to provide a comprehensive synopsis of preclinical evidence and clinical experience of various donor cell types, their restorative mechanisms, delivery routes, imaging strategies, future prospects and challenges for translating cell therapies as a neurorestorative regimen in clinical applications.
Collapse
Affiliation(s)
- Xinfeng Liu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China.
| | - Ruidong Ye
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Tao Yan
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA; Department of Neurology, Tianjin General Hospital, Tianjin University School of Medicine, Tianjin, China
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Gelin Xu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Xinying Fan
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yongjun Jiang
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - R Anne Stetler
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - George Liu
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing, China
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.
| |
Collapse
|
186
|
Yang Z, Chen P, Yu H, Luo W, Pi M, Wu Y, Wang L, Yang F, Gou Y. Combinatorial effects of conception and governor vessel electroacupuncture and human umbilical cord blood-derived mesenchymal stem cells on pathomorphologic lesion and cellular apoptosis in rats with cerebral ischemia/reperfusion. J TRADIT CHIN MED 2013; 33:779-86. [DOI: 10.1016/s0254-6272(14)60012-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
187
|
Young CC, Al-Dalahmah O, Lewis NJ, Brooks KJ, Jenkins MM, Poirier F, Buchan AM, Szele FG. Blocked angiogenesis in Galectin-3 null mice does not alter cellular and behavioral recovery after middle cerebral artery occlusion stroke. Neurobiol Dis 2013; 63:155-64. [PMID: 24269916 DOI: 10.1016/j.nbd.2013.11.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 10/24/2013] [Accepted: 11/12/2013] [Indexed: 12/31/2022] Open
Abstract
Angiogenesis is thought to decrease stroke size and improve behavioral outcomes and therefore several clinical trials are seeking to augment it. Galectin-3 (Gal-3) expression increases after middle cerebral artery occlusion (MCAO) and has been proposed to limit damage 3days after stroke. We carried out mild MCAO that damages the striatum but spares the cerebral cortex and SVZ. Gal-3 gene deletion prevented vascular endothelial growth factor (VEGF) upregulation after MCAO. This inhibited post-MCAO increases in endothelial proliferation and angiogenesis in the striatum allowing us to uniquely address the function of angiogenesis in this model of stroke. Apoptosis and infarct size were unchanged in Gal-3(-/-) mice 7 and 14 days after MCAO, suggesting that angiogenesis does not affect lesion size. Microglial and astrocyte activation/proliferation after MCAO was similar in wild type and Gal-3(-/-) mice. In addition, openfield activity, motor hemiparesis, proprioception, reflex, tremors and grooming behaviors were essentially identical between WT and Gal-3(-/-) mice at 1, 3, 7, 10 and 14 days after MCAO, suggesting that penumbral angiogenesis has limited impact on behavioral recovery. In addition to angiogenesis, increased adult subventricular zone (SVZ) neurogenesis is thought to provide neuroprotection after stroke in animal models. SVZ neurogenesis and migration to lesion were overall unaffected by the loss of Gal-3, suggesting no compensation for the lack of angiogenesis in Gal-3(-/-) mice. Because angiogenesis and neurogenesis are usually coordinately regulated, identifying their individual effects on stroke has hitherto been difficult. These results show that Gal-3 is necessary for angiogenesis in stroke in a VEGF-dependant manner, but suggest that angiogenesis may be dispensable for post-stroke endogenous repair, therefore drawing into question the clinical utility of augmenting angiogenesis.
Collapse
Affiliation(s)
- Christopher C Young
- University of Oxford, Department of Physiology, Anatomy and Genetics, University of Oxford, OX1 3QX, UK
| | - Osama Al-Dalahmah
- University of Oxford, Department of Physiology, Anatomy and Genetics, University of Oxford, OX1 3QX, UK
| | - Nicola J Lewis
- University of Oxford, Department of Physiology, Anatomy and Genetics, University of Oxford, OX1 3QX, UK
| | - Keith J Brooks
- Nuffield Department of Clinical Medicine, University of Oxford, OX1 3QX, UK
| | - Micaela M Jenkins
- Nuffield Department of Clinical Medicine, University of Oxford, OX1 3QX, UK
| | - Françoise Poirier
- Institut Jacques Monod, UMR CNRS 7592, Université Paris Diderot, 75205 Paris 13, France
| | - Alastair M Buchan
- Nuffield Department of Clinical Medicine, University of Oxford, OX1 3QX, UK
| | - Francis G Szele
- University of Oxford, Department of Physiology, Anatomy and Genetics, University of Oxford, OX1 3QX, UK.
| |
Collapse
|
188
|
Ruan GP, Han YB, Wang TH, Xing ZG, Zhu XB, Yao X, Ruan GH, Wang JX, Pang RQ, Cai XM, He J, Zhao J, Pan XH. Comparative study among three different methods of bone marrow mesenchymal stem cell transplantation following cerebral infarction in rats. Neurol Res 2013; 35:212-20. [PMID: 23452580 DOI: 10.1179/1743132812y.0000000152] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Affiliation(s)
- Guang-Ping Ruan
- Stem Cell Engineering Laboratory of Yunnan Province, Kunming General Hospital of Chengdu Military Command, China
| | - Yi-Bing Han
- Shanghai First Maternity and Infants HospitalTongji University, China
| | - Ting-Hua Wang
- Institute of NeuroscienceKunming Medical University, China
| | - Zhi-Guo Xing
- Stem Cell Engineering Laboratory of Yunnan Province, Kunming General Hospital of Chengdu Military Command, China
| | - Xing-Bao Zhu
- Stem Cell Engineering Laboratory of Yunnan Province, Kunming General Hospital of Chengdu Military Command, China
| | - Xiang Yao
- Stem Cell Engineering Laboratory of Yunnan Province, Kunming General Hospital of Chengdu Military Command, China
| | - Guang-Hong Ruan
- Stem Cell Engineering Laboratory of Yunnan Province, Kunming General Hospital of Chengdu Military Command, China
| | - Jin-Xiang Wang
- Stem Cell Engineering Laboratory of Yunnan Province, Kunming General Hospital of Chengdu Military Command, China
| | - Rong-Qing Pang
- Stem Cell Engineering Laboratory of Yunnan Province, Kunming General Hospital of Chengdu Military Command, China
| | - Xue-Min Cai
- Stem Cell Engineering Laboratory of Yunnan Province, Kunming General Hospital of Chengdu Military Command, China
| | - Jie He
- Stem Cell Engineering Laboratory of Yunnan Province, Kunming General Hospital of Chengdu Military Command, China
| | - Jing Zhao
- Stem Cell Engineering Laboratory of Yunnan Province, Kunming General Hospital of Chengdu Military Command, China
| | - Xing-Hua Pan
- Stem Cell Engineering Laboratory of Yunnan Province, Kunming General Hospital of Chengdu Military Command, China
| |
Collapse
|
189
|
Li Y, Liu Z, Xin H, Chopp M. The role of astrocytes in mediating exogenous cell-based restorative therapy for stroke. Glia 2013; 62:1-16. [PMID: 24272702 DOI: 10.1002/glia.22585] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 08/08/2013] [Accepted: 09/18/2013] [Indexed: 12/19/2022]
Abstract
Astrocytes have not been a major therapeutic target for the treatment of stroke, with most research emphasis on the neuron. Given the essential role that astrocytes play in maintaining physiological function of the central nervous system and the very rapid and sensitive reaction astrocytes have in response to cerebral injury or ischemic insult, we propose to replace the neurocentric view for treatment with a more nuanced astrocytic centered approach. In addition, after decades of effort in attempting to develop neuroprotective therapies, which target reduction of the ischemic lesion, there are no effective clinical treatments for stroke, aside from thrombolysis with tissue plasminogen activator, which is used in a small minority of patients. A more promising therapeutic approach, which may affect nearly all stroke patients, may be in promoting endogenous restorative mechanisms, which enhance neurological recovery. A focus of efforts in stimulating recovery post stroke is the use of exogenously administered cells. The present review focuses on the role of the astrocyte in mediating the brain network, brain plasticity, and neurological recovery post stroke. As a model to describe the interaction of a restorative cell-based therapy with astrocytes, which drives recovery from stroke, we specifically highlight the subacute treatment of stroke with multipotent mesenchymal stromal cell therapy.
Collapse
Affiliation(s)
- Yi Li
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan
| | | | | | | |
Collapse
|
190
|
Fargen KM, Mocco J, Hoh BL. Can We Rebuild the Human Brain? The Exciting Promise and Early Evidence That Stem Cells May Provide a Real Clinical Cure for Stroke in Humans. World Neurosurg 2013; 80:e69-72. [DOI: 10.1016/j.wneu.2012.07.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 07/20/2012] [Indexed: 01/29/2023]
|
191
|
Miao J, Shen LH, Tang YH, Wang YT, Tao MX, Jin KL, Zhao YJ, Yang GY. Overexpression of adiponectin improves neurobehavioral outcomes after focal cerebral ischemia in aged mice. CNS Neurosci Ther 2013; 19:969-77. [PMID: 24164711 DOI: 10.1111/cns.12198] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 09/25/2013] [Accepted: 09/25/2013] [Indexed: 12/14/2022] Open
Abstract
AIMS To study whether adiponectin (APN) could improve neurological outcomes in aged mice after ischemic stroke. METHODS Adeno-associated virus carrying APN gene was injected into aged and young adult mice 7 days before transient middle cerebral artery occlusion (tMCAO). Atrophic volumes and neurobehavioral deficiencies were determined up to 28 days after tMCAO. Focal angiogenesis was determined based on blood vessel number in the ischemic regions. RESULTS Increased atrophic volume and more sever neurobehavioral deficits were found in the aged mice compared with young adult mice (P < 0.05). AAV-APN gene transfer attenuated atrophic volume and improved neurobehavioral outcomes, along with increased focal angiogenesis in both aged and young adult mice, compared with control animals (P < 0.05). In addition, the attenuation of atrophic volume and the improvement in neurobehavioral outcomes were much more significant in aged mice than in young adult mice after AAV-APN administration (P < 0.05). The number of microvessels in aged AAV-APN mouse ischemic brain was higher than in young adult AAV-APN treated mouse brain (P < 0.05). CONCLUSIONS Our results demonstrate that APN overexpression reduces ischemic brain injury and improves neurobehavioral function recovery in aged mice than in young mice, suggesting APN is more beneficial in aged animals after ischemic stroke.
Collapse
Affiliation(s)
- Jie Miao
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
192
|
Rosado-de-Castro PH, Schmidt FDR, Battistella V, Lopes de Souza SA, Gutfilen B, Goldenberg RCDS, Kasai-Brunswick TH, Vairo L, Silva RM, Wajnberg E, Alvarenga Americano do Brasil PE, Gasparetto EL, Maiolino A, Alves-Leon SV, Andre C, Mendez-Otero R, Rodriguez de Freitas G, Barbosa da Fonseca LM. Biodistribution of bone marrow mononuclear cells after intra-arterial or intravenous transplantation in subacute stroke patients. Regen Med 2013; 8:145-55. [PMID: 23477395 DOI: 10.2217/rme.13.2] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
AIMS To assess the biodistribution of bone marrow mononuclear cells (BMMNC) delivered by different routes in patients with subacute middle cerebral artery ischemic stroke. PATIENTS & METHODS This was a nonrandomized, open-label Phase I clinical trial. After bone marrow harvesting, BMMNCs were labeled with technetium-99m and intra-arterially or intravenously delivered together with the unlabeled cells. Scintigraphies were carried out at 2 and 24 h after cell transplantation. Clinical follow-up was continued for 6 months. RESULTS Twelve patients were included, between 19 and 89 days after stroke, and received 1-5 × 10(8) BMMNCs. The intra-arterial group had greater radioactive counts in the liver and spleen and lower counts in the lungs at 2 and 24 h, while in the brain they were low and similar for both routes. CONCLUSION BMMNC labeling with technetium-99m allowed imaging for up to 24 h after intra-arterial or intravenous injection in stroke patients.
Collapse
Affiliation(s)
- Paulo Henrique Rosado-de-Castro
- Department of Radiology, School of Medicine, Universidade Federal do Rio de Janeiro, Rua Professor Rodolpho Paulo Rocco 255, Cidade Universitária, Ilha do Fundão, 21941-913, Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
193
|
Wan H, Li F, Zhu L, Wang J, Yang Z, Pan Y. Update on therapeutic mechanism for bone marrow stromal cells in ischemic stroke. J Mol Neurosci 2013; 52:177-85. [PMID: 24048741 DOI: 10.1007/s12031-013-0119-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 09/09/2013] [Indexed: 02/08/2023]
Abstract
Cerebral ischemia is a major cause of morbidity and mortality in the aged population, as well as a tremendous burden on the healthcare system. Despite timely treatment with thrombolysis and percutaneous intravascular interventions, many patients are often left with irreversible neurological deficits. Bone marrow stromal cells (BMSCs), also referred to as mesenchymal stem cells (MSCs), are a type of nonhematopoietic stem cells which exists in bone marrow mesh, with the potential to self-renew. Unlike cells in the central nervous system, BMSCs differentiate not only into mesodermal cells, but also endodermal and ectodermal cells. Moreover, it has been reported that BMSCs develop into cells with neural and vascular markers and play a role in recovery from ischemic stroke. These findings have fuelled excitement in regenerative medicine for neurological diseases, especially for ischemic stroke. There is now preclinical evidence to suggest that BMSCs grafted into the brain of ischemic models abrogate neurological deficits. Based on the overwhelming evidence from animal studies as well as in clinical trials, BMSC transplantation is considered a promising strategy for treatment of ischemic stroke. The goal of this review is to present an integrated consideration of molecular mechanisms in a chronological fashion and discuss an optimal BMSC delivery route for ischemic stroke.
Collapse
Affiliation(s)
- Huan Wan
- Department of Neurology, First Hospital and Clinical College, Harbin Medical University, Room 501, Building 3, 23 Youzheng, Harbin, 150001, China
| | | | | | | | | | | |
Collapse
|
194
|
Eckert MA, Vu Q, Xie K, Yu J, Liao W, Cramer SC, Zhao W. Evidence for high translational potential of mesenchymal stromal cell therapy to improve recovery from ischemic stroke. J Cereb Blood Flow Metab 2013; 33:1322-34. [PMID: 23756689 PMCID: PMC3764389 DOI: 10.1038/jcbfm.2013.91] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 05/06/2013] [Accepted: 05/08/2013] [Indexed: 12/27/2022]
Abstract
Although ischemic stroke is a major cause of morbidity and mortality, current therapies benefit only a small proportion of patients. Transplantation of mesenchymal stromal cells (MSC, also known as mesenchymal stem cells or multipotent stromal cells) has attracted attention as a regenerative therapy for numerous diseases, including stroke. Mesenchymal stromal cells may aid in reducing the long-term impact of stroke via multiple mechanisms that include induction of angiogenesis, promotion of neurogenesis, prevention of apoptosis, and immunomodulation. In this review, we discuss the clinical rationale of MSC for stroke therapy in the context of their emerging utility in other diseases, and their recent clinical approval for treatment of graft-versus-host disease. An analysis of preclinical studies examining the effects of MSC therapy after ischemic stroke indicates near-universal agreement that MSC have significant favorable effect on stroke recovery, across a range of doses and treatment time windows. These results are interpreted in the context of completed and ongoing human clinical trials, which provide support for MSC as a safe and potentially efficacious therapy for stroke recovery in humans. Finally, we consider principles of brain repair and manufacturing considerations that will be useful for effective translation of MSC from the bench to the bedside for stroke recovery.
Collapse
Affiliation(s)
- Mark A Eckert
- Departments of Pharmaceutical Sciences and Biomedical Engineering, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, University of California, Irvine, California, USA
| | - Quynh Vu
- Department of Neurology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California, USA
| | - Kate Xie
- Department of Neurology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California, USA
| | - Jingxia Yu
- Departments of Pharmaceutical Sciences and Biomedical Engineering, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, University of California, Irvine, California, USA
| | - Wenbin Liao
- Department of Pathology, State University of New York at Stony Brook, Stony Brook, New York, USA
| | - Steven C Cramer
- Departments of Neurology and Anatomy and Neurobiology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California, USA
| | - Weian Zhao
- Departments of Pharmaceutical Sciences and Biomedical Engineering, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, University of California, Irvine, California, USA
| |
Collapse
|
195
|
Chen L, Xi H, Huang H, Zhang F, Liu Y, Chen D, Xiao J. Multiple cell transplantation based on an intraparenchymal approach for patients with chronic phase stroke. Cell Transplant 2013; 22 Suppl 1:S83-91. [PMID: 23992950 DOI: 10.3727/096368913x672154] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Stroke is the third leading cause of death worldwide and a huge perpetrator in adult disability. This pilot clinical study investigates the possible benefits of transplanting multiple cells in chronic stroke. A total of 10 consecutive stroke patients were treated by combination cell transplantation on the basis of an intraparenchymal approach from November 2003 to April 2011. There were six males and four females. Their age ranged from 42 to 87 years, and the course of disease varied from 6 months to 20 years. Six patients suffered cerebral infarction, and four patients suffered a brain hemorrhage. The olfactory ensheathing cells, neural progenitor cells, umbilical cord mesenchymal cells, and Schwann cells were injected through selected routes including intracranial parenchymal implantation, intrathecal implantation, and intravenous administration, respectively. The clinical neurological function was assessed carefully and independently before treatment and during a long-term follow-up using the Clinic Neurologic Impairment Scale and the Barthel index. All patients were followed up successfully from 6 months to 2 years after cell transplantation. Every subject achieved neurological function amelioration including improved speech, muscle strength, muscular tension, balance, pain, and breathing; most patients had an increased Barthel index score and Clinic Neurologic Impairment Scale score. These preliminary results demonstrate the novel strategy of combined multiple cell therapy based on intraparenchymal delivery: it appears to be relatively clinically safe and at least initially beneficial for chronic stroke patients. This manuscript is published as part of the International Association of Neurorestoratology (IANR) supplement issue of Cell Transplantation.
Collapse
|
196
|
Yuan J, Huang G, Xiao Z, Lin L, Han T. Overexpression of β-NGF promotes differentiation of bone marrow mesenchymal stem cells into neurons through regulation of AKT and MAPK pathway. Mol Cell Biochem 2013; 383:201-11. [PMID: 23934089 DOI: 10.1007/s11010-013-1768-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 08/02/2013] [Indexed: 02/03/2023]
Abstract
Bone marrow stromal stem cells (BMSCs) are fibroblastic in shape and capable of self-renewal and have the potential for multi-directional differentiation. Nerve growth factor (NGF), a homodimeric polypeptide, plays an important role in the nervous system by supporting the survival and growth of neural cells, regulating cell growth, promoting differentiation into neuron, and neuron migration. Adenoviral vectors are DNA viruses that contain 36 kb of double-stranded DNA allowing for transmission of the genes to the host nucleus but not inserting them into the host chromosome. The present study aimed to investigate the induction efficiency and differentiation of neural cells from BMSCs by β-NGF gene transfection with recombinant adenoviral vector (Ad-β-NGF) in vitro. The results of immunochemical assay confirmed the induced cells as neuron cells. Moreover, flow cytometric analysis, Annexin-V-FITC/PI, and BrdU assay revealed that chemical inducer β-mercaptoethanol (β-met) triggered apoptosis of BMSCs, as evidenced by inhibition of DNA fragmentation, nuclear condensation, translocation of phospholipid phosphatidylserine, and activation of caspase-3. Furthermore, the results of western blotting showed that β-met suppressed AKT signaling pathway and regulated the MAPKs during differentiation of BMSCs. In contrast, Ad-β-NGF effectively induced the differentiation of BMSCs without causing any cytopathic phenomenon and apoptotic cell death. Moreover, Ad-β-NGF recovered the expression level of phosphorylated AKT and MAPKs in cells exposed to chemical reagents. Taken together, these results suggest that β-NGF gene transfection promotes the differentiation of BMSCs into neurons through regulation of AKT and MAPKs signaling pathways.
Collapse
Affiliation(s)
- Jun Yuan
- Department of Neurovascular Surgery, First Hospital Affiliated to Shantou University, 57# Changping Road, Jinping District, Shantou, 515041, China
| | | | | | | | | |
Collapse
|
197
|
Hu M, Ludlow D, Alexander JS, McLarty J, Lian T. Improved wound healing of postischemic cutaneous flaps with the use of bone marrow-derived stem cells. Laryngoscope 2013; 124:642-8. [PMID: 23818296 DOI: 10.1002/lary.24293] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 06/02/2013] [Accepted: 06/13/2013] [Indexed: 12/31/2022]
Abstract
OBJECTIVES/HYPOTHESIS To determine if the intravascular delivery of mesenchymal stem cells improves wound healing and blood perfusion to postischemic cutaneous flap tissues. STUDY DESIGN Randomized controlled study. METHODS A murine model of a cutaneous flap was created based on the inferior epigastric vessels. Mice (n = 14) underwent 3.5 hours of ischemia followed by reperfusion. Bone marrow stromal cells (BMSCs) 1 × 10(6) were injected intravenously. Wound healing was then assessed measuring percent flap necrosis, flap perfusion, and tensile strength of the flap after a period of 14 days. Localization of BMSCs was determined with radiolabeled and fluorescent labeled BMSCs. RESULTS Postischemic cutaneous flap tissues treated with BMSCs demonstrated significantly less necrosis than control flaps (P <0.01). Beginning on postoperative day 5, BMSC-treated flaps demonstrated greater blood perfusion than untreated flaps (P <0.01). Tensile strength of BMSC-treated cutaneous flaps was significantly higher (P <0.01), with a mean strength of 283.4 ± 28.4 N/m than control flaps with a mean of 122.4 ± 23.5 N/m. Radiolabeled BMSCs localized to postischemic flaps compared to untreated tissues (P = 0.001). Fluorescent microscopy revealed incorporation of BMSCs into endothelial and epithelial tissues of postischemic flaps. CONCLUSIONS This study demonstrates that the intravascular delivery of BMSCs increases wound healing and promotes flap survival following ischemia-reperfusion injury of cutaneous tissue flaps.
Collapse
Affiliation(s)
- Melissa Hu
- Department of Otolaryngology-Head and Neck Surgery, LSUH-S, Shreveport, Louisiana, U.S.A
| | | | | | | | | |
Collapse
|
198
|
Abstract
Stroke is the most common cause of disability in the United States, and one of the leading causes of mortality and disability in the world. The hope that damage to the CNS can be reversed or at least ameliorated is the central idea behind the research into neural repair. The ultimate repair for the brain should restore the entire lost structure and it's function. However, partial benefit is possible from addressing some of the needs of the injured brain. These partial solutions are the basis of current research into brain repair after stroke. An opportunity arises for two kinds of intervention: (1) replacement of neurons; (2) support of existing neurons, to prevent excessive degeneration and promote rewiring and plasticity. Transplantation for stroke in the rat model was regularly reported starting in 1992, demonstrating graft survival and even evidence of connection with the host brain. These studies determined several parameters for future work in stroke models, but ultimately had limited efficacy and did not progress to clinical experiments. A variety of cell types have been tried for restoration of brain function after stroke, mostly in rodent models. Human fetal cells had shown some promise in clinical studies for the treatment of Parkinson's disease. The technical and ethical difficulties associated with these cells promoted a search for alternatives. These include porcine fetal cells, human cultured stem cells, immortalized cell lines, marrow stromal cells, Sertoli cells pineal cells, and other sources. Human clonal cell lines have few ethical limitations, but some questions remain regarding their safety and efficacy. Autologous somatic stem cells are a very attractive source--there are no ethical concerns and graft rejection is not an issue. However, it is not clear that somatic cells can are plastic enough and can be safely induced to a neural fate. Restorative treatment for stroke is a new field of study. Naturally, new ideas abound and many strategies have been suggested and tried. Methods and controversies abound, and include: local delivery of cells to the area of the stroke versus grafting to an area of the brain far removed form the stroke; cell therapy for reconstitution of structure and function versus use of cell grafts to support intrinsic repair and recovery mechanisms; intravascular administration of bone marrow or other stem cells; and combination grafts, or co-grafting of several cell types or cells and other substances. The various strategies address the issue of restorative treatments form different perspectives. Some interventions occur early after stroke, or are intended to preserve existing neural structures. For example, treatment strategies that aim to provide trophic support may demonstrate early beneficial results. Other strategies aim for growth and integration of new neurons to replace those lost after stroke. In this case, early beneficial results are not likely. Functional integration of grafted neurons, if it can ever happen, is likely to require training and exercise of the appropriate capacities. Further advances in preclinical studies of neural transplantation will require improved animal models with increased sensitivity to subtle behavioral and imaging changes. Non-human primate models have been established and may increase in importance as a phase before clinical trials. The future of brain repair for stroke is likely to require some form of combination therapy designed to replace the lost cells and supporting structure, attract new blood supply, support and enhance intrinsic repair and plasticity mechanisms.
Collapse
Affiliation(s)
- Ben Roitberg
- Department of Neurosurgery, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
199
|
Liu Y, Tang GH, Sun YH, Lin XJ, Wei C, Yang GY, Liu JR. The protective role of Tongxinluo on blood-brain barrier after ischemia-reperfusion brain injury. JOURNAL OF ETHNOPHARMACOLOGY 2013; 148:632-639. [PMID: 23707212 DOI: 10.1016/j.jep.2013.05.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 05/03/2013] [Accepted: 05/14/2013] [Indexed: 06/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tongxinluo (TXL), a renowned traditional Chinese medicine, consists of several different kinds of ingredients and has been widely used to treat myocardial infarction and ischemic stroke. However, the underlying neuroprotective mechanisms are not fully understood. AIM OF THE STUDY We focus on the effect of TXL on blood-brain barrier (BBB) including edema formation and tight junction (TJ) protein rearrangement, and inflammatory response after transient middle cerebral artery occlusion (tMCAO). We further explore the protective mechanism of TXL on ischemia-induced BBB damage. MATERIALS AND METHODS Adult CD1 male mice (n=168) were randomly divided into TXL pre-treatment group, TXL pre-post treatment group, TXL post-treatment group, control group and sham group. Mice in TXL pre-treatment group were given TXL solution by 1g/kg/day orally for 7 days before tMCAO. Mice in pre-post treatment group were continuously given TXL 7 days before and 14 days after tMCAO. Mice in TXL post-treatment group were given TXL solution immediately after tMCAO. Rotarod test and neurological severity scores were evaluated at 1-14 days following tMCAO. Brains were harvested for examining infarct volume, edema formation, and immunofluorescent staining at 1 and 3 days after tMCAO. Cytokines IL-6, IL-1β and TNF-α mRNA expression, and BBB permeability were further examined by RT-PCR and immunostaining. RESULTS TXL pre-post treatment improved neurobehavioral outcomes and reduced infarct volume compared to the control (p<0.05). Meanwhile, hemispheric swelling, Evans blue and IgG protein extravasation reduced, while TJ protein expression up-regulated in pre-post treatment group (p<0.05). Further study indicated that infarct volume was smaller and BBB damage was less severe in TXL pre-post treatment group compared to TXL pre-treatment alone. It was noted that fewer myeloperoxidase (MPO) positive cells and less cytokines IL-6, IL-1β and TNF-α expression in pre-post treatment group compared to the control group (p<0.05). CONCLUSIONS TXL pre-treatment and pre-post treatment effectively protected the brain from BBB disruption via alleviating inflammatory response. Moreover, pre-post treatment has better outcomes, suggesting that continuous administration of TXL before and throughout ischemia period is necessary because of multiple functions of TXL.
Collapse
Affiliation(s)
- Ye Liu
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | | | | | | | | | | | | |
Collapse
|
200
|
Laroni A, Novi G, Kerlero de Rosbo N, Uccelli A. Towards clinical application of mesenchymal stem cells for treatment of neurological diseases of the central nervous system. J Neuroimmune Pharmacol 2013; 8:1062-76. [PMID: 23579931 DOI: 10.1007/s11481-013-9456-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Accepted: 03/31/2013] [Indexed: 12/13/2022]
Abstract
The diagnosis of a neurological disease of the central nervous system (CNS) is often associated with the anticipation of an irreversible and untreatable disability. This is the case also of multiple sclerosis (MS) where approved treatments effectively modulate the autoimmune attack to myelin antigens, but poorly affect neurodegeneration and do not promote tissue repair. Thus, stem cell-based therapies are increasingly being considered a possible strategy for diseases of the CNS. Mesenchymal stem cells (MSC), the safety of which has been demonstrated in the last 20 years through clinical trials and case studies, are of particular interest in view not only of their neuroprotective, but also of their immunomodulatory properties. Here, we review the therapeutic features of MSC that make them relevant in the treatment of CNS illnesses and discuss the pioneer clinical experience with MSC-based therapy in neurological diseases.
Collapse
Affiliation(s)
- Alice Laroni
- Department of Neurosciences Ophthalmology, Genetics, Rehabilitation and Child Health, University of Genoa, Genoa, Italy
| | | | | | | |
Collapse
|