251
|
Jang KS, Lee KS, Yang SH, Jeun SS. In vivo Tracking of Transplanted Bone Marrow-Derived Mesenchymal Stem Cells in a Murine Model of Stroke by Bioluminescence Imaging. J Korean Neurosurg Soc 2010; 48:391-8. [PMID: 21286474 DOI: 10.3340/jkns.2010.48.5.391] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2009] [Revised: 06/26/2010] [Accepted: 11/25/2010] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE This study was designed to validate the cell trafficking efficiency of the in vivo bioluminescence image (BLI) study in the setting of transplantation of the luciferase expressing bone marrow-derived mesenchymal stem cells (BMSC), which were delivered at each different time after transient middle cerebral artery occlusion (MCAO) in a mouse model. METHODS Transplanting donor BMSC were prepared by primary cell culture from transgenic mouse expressing luciferase (LUC). Transient focal infarcts were induced in 4-6-week-old male nude mice. The experiment mice were divided into five groups by the time of MSC transplantation : 1) sham-operation group, 2) 2-h group, 3) 1-day group, 4) 3-day group, and 5) 1- week group. BLI for detection of spatial distribution of transplanted MSC was performed by detecting emitted photons. Migration of the transplanted cells to the infarcted area was confirmed by histological examinations. Differences between groups were evaluated by paired t-test. RESULTS A focal spot of bioluminescence was observed at the injection site on the next day after transplantation by signal intensity of bioluminescence. After 4 weeks, the mean signal intensities of 2-h, 1-day, 3-day, and 1-week group were 2.6×10(7) ± 7.4×10(6), 6.1×10(6) ± 1.2×10(6), 1.7×10(6) ± 4.4×10(5), and 8.9×10(6) ± 9.5×10(5), respectively. The 2-h group showed significantly higher signal intensity (p < 0.01). The engrafted BMSC showed around the infarct border zones on immunohistochemical examination. The counts of LUC-positive cells revealed the highest number in the 2-h group, in agreement with the results of BLI experiments (p < 0.01). CONCLUSION In this study, the results suggested that the transplanted BMSC migrated to the infarct border zone in BLI study and the higher signal intensity of LUC-positive cells seen in 2 hrs after MSC transplantation in MCAO mouse model. In addition, noninvasive imaging in real time is an ideal method for tracking stem cell transplantation. This method can be widely applied to various research fields of cell transplantation therapy.
Collapse
Affiliation(s)
- Kyung-Sool Jang
- Department of Neurosurgery, Institute of Catholic Integrative Medicine (ICIM) of Incheon St. Mary's Hospital, The Catholic University of Korea, Incheon, Korea
| | | | | | | |
Collapse
|
252
|
Doeppner TR, Hermann DM. Mesenchymal stem cells in the treatment of ischemic stroke: progress and possibilities. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2010; 3:157-63. [PMID: 24198521 PMCID: PMC3781740 DOI: 10.2147/sccaa.s7820] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Stroke is a major cause of death and long-term disability in industrialized countries, and the only causal therapy for stroke comprises recombinant tissue plasminogen activator(rt-PA)-mediated recanalization of the occluded vessel. New experimental strategies focus on neuroregenerative approaches, among which the application of mesenchymal stem cells (MSCs) has gained increasing attention. MSCs, like other stem cells, have the capacity of unlimited self-renewal giving rise to differentiated cells from various cell lineages. Bone marrow (BM)-derived MSCs are the most frequently used MSC type in experimental stroke studies. Application of BM-derived MSCs and, in some studies, transplantation of MSCs from other tissue sources resulted in an improved functional recovery in experimental animals, although stroke volumes were not always affected by MSC transplantation. The underlying precise mechanisms of this phenomenon remain elusive, although MSC transplantation is considered to affect many diverse events, eg, by modulating the inflammatory milieu, stimulating endogenous neurogenesis and angiogenesis, and reducing glial scar formation. On the contrary, neuronal differentiation and integration of transplanted MSCs do not seem to affect stroke outcome significantly. On the basis of these preclinical studies, first clinical trials confirmed improved functional recovery in patients who had received BM-derived MSCs systemically, although the number of patients enrolled in these studies was low and there were no adequate control groups. In this review, we describe some fundamental biological characteristics of MSCs and further review some preclinical experimental studies, with special emphasis on BM-derived MSCs. We also review clinical trials in which MSCs have been used and conclude with a short outlook on the application of MSCs in stroke research.
Collapse
Affiliation(s)
- Thorsten R Doeppner
- Department of Neurology, University of Duisburg-Essen Medical School, Essen, North Rhine-Westphalia, Germany
| | | |
Collapse
|
253
|
Tóth ZE, Leker RR, Shahar T, Bratincsak A, Szalayova I, Key S, Palkovits M, Cassiani-Ingoni R, Mezey E. Bone marrow-derived nonreactive astrocytes in the mouse brain after permanent middle cerebral artery occlusion. Stem Cells Dev 2010; 20:539-46. [PMID: 20604679 DOI: 10.1089/scd.2010.0237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We studied the effect of permanent unilateral middle cerebral artery occlusion (PMCAO) on the generation of bone marrow (BM)-derived astrocytes in female mice previously transplanted with enhanced green fluorescent protein-expressing BM from male donors. In addition to an untreated PMCAO group, one group of mice also received intracerebral infusion of transforming growth factor-alpha, resulting in a decrease in the size of the infarct. Two months after PMCAO, we found a specific type of astrocyte of BM origin in the side of the injury, near the lesion. These astrocytes did not express glial fibrillary acidic protein (GFAP) by conventional fluorescence immunostaining; however, GFAP was easily detectable by tyramide signal amplification. These cells also expressed S100β, confirming their astrocytic character. Unlike the endogenous reactive astrocytes, these BM-derived astrocytes did not proliferate during the first week of ischemia and did not contribute to the glial scar formation. Transforming growth factor-alpha infusion increased the number of BM-derived astrocytes, without affecting their distribution. Interestingly, exclusively by tyramide signal amplification staining, we found that endogenous astrocytes displaying an identical morphology were also present in control mouse and human brains. Our data demonstrate that a subpopulation of nonreactive astrocytes expressing low levels of GFAP can originate from transplanted BM in the ischemic brain. We believe that these cells represent a subpopulation of astrocytes earlier considered to be GFAP negative. The high number of astrocytes with identical morphology and chemical character in control brains suggest that these type of astrocytes may have important functional role in the central nervous system that calls for further studies.
Collapse
Affiliation(s)
- Zsuzsanna E Tóth
- National Institute of Dental and Craniofacial Research, National Institute of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
254
|
Tanaka Y, Tanaka R, Liu M, Hattori N, Urabe T. Cilostazol attenuates ischemic brain injury and enhances neurogenesis in the subventricular zone of adult mice after transient focal cerebral ischemia. Neuroscience 2010; 171:1367-76. [PMID: 20933581 DOI: 10.1016/j.neuroscience.2010.10.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 09/29/2010] [Accepted: 10/02/2010] [Indexed: 11/30/2022]
Abstract
Evidence suggests that neurogenesis occurs in the adult mammalian brain, and that various stimuli, for example, ischemia/hypoxia, enhance the generation of neural progenitor cells in the subventricular zone (SVZ) and their migration into the olfactory bulb. In a mouse stroke model, focal ischemia results in activation of neural progenitor cells followed by their migration into the ischemic lesion. The present study assessed the in vivo effects of cilostazol, a type 3 phosphodiesterase inhibitor known to activate the cAMP-responsive element binding protein (CREB) signaling, on neurogenesis in the ipsilateral SVZ and peri-infarct area in a mouse model of transient middle cerebral artery occlusion. Mice were divided into sham operated (n=12), vehicle- (n=18) and cilostazol-treated (n=18) groups. Sections stained for 5-bromodeoxyuridine (BrdU) and several neuronal and a glial markers were analyzed at post-ischemia days 1, 3 and 7. Cilostazol reduced brain ischemic volume (P<0.05) and induced earlier recovery of neurologic deficit (P<0.05). Cilostazol significantly increased the density of BrdU-positive newly-formed cells in the SVZ compared with the vehicle group without ischemia. Increased density of doublecortin (DCX)-positive and BrdU/DCX-double positive neural progenitor cells was noted in the ipsilateral SVZ and peri-infarct area at 3 and 7 days after focal ischemia compared with the vehicle group (P<0.05). Cilostazol increased DCX-positive phosphorylated CREB (pCREB)-expressing neural progenitor cells, and increased brain derived neurotrophic factor (BDNF)-expressing astrocytes in the ipsilateral SVZ and peri-infarct area. The results indicated that cilostazol enhanced neural progenitor cell generation in both ipsilateral SVZ and peri-infarct area through CREB-mediated signaling pathway after focal ischemia.
Collapse
Affiliation(s)
- Y Tanaka
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | | | | |
Collapse
|
255
|
Gobbel GT, Kondziolka D, Fellows-Mayle W, Uram M. Cellular transplantation for the nervous system: impact of time after preparation on cell viability and survival. J Neurosurg 2010; 113:666-72. [PMID: 19911893 DOI: 10.3171/2009.10.jns09252] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECT Cell transplantation has shown promise for the treatment of various neurological disorders, but the factors that influence cell survival and integration following transplantation are poorly understood. In fact, little is known regarding how simple but potentially critical variables, including the method of cellular preparation and administration, might affect transplant success. The goal of the present study was to determine the impact of time between tissue preparation and implantation on cellular viability. Time can vary with cell preparation, delivery to the operating room, and surgical technique. This study was also designed to evaluate the sensitivity of various methods of assessing implant viability. METHODS Cell lines of neural progenitor cells and bone marrow stromal cells were generated from healthy adult mice. On the day of experimentation, the cells were collected, suspended in a balanced salt solution, and sequentially assessed for viability for up to 3.5 hours based on their appearance under phase-contrast microscopy, their ability to retain a fluorescent dye, and their attachment to a cultivation surface for 24 hours. RESULTS When viability was measured based on the ability of cells to retain a fluorescent dye, there was a decrease in viability of 10-15% each hour. Based on the ability of the cells to attach to a culture surface and grow for 24 hours, viability decreased more rapidly at approximately 20% per hour. In addition, only about one-third of the cells judged viable based on phase-contrast microscopy or acute dye retention were found to be viable based on plating, and only 10% of the cells initially judged as viable were still capable of survival after 3 hours in suspension. CONCLUSIONS The authors' results indicate that that there can be significant losses in viability between preparation and implantation and that more sophisticated methods of evaluation, such as the ability of cells to attach to a substrate and grow, may be required to detect decreases in viability. The time between preparation and implantation will be an important factor in clinical trial design.
Collapse
Affiliation(s)
- Glenn T Gobbel
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.
| | | | | | | |
Collapse
|
256
|
Kidd S, Caldwell L, Dietrich M, Samudio I, Spaeth EL, Watson K, Shi Y, Abbruzzese J, Konopleva M, Andreeff M, Marini FC. Mesenchymal stromal cells alone or expressing interferon-beta suppress pancreatic tumors in vivo, an effect countered by anti-inflammatory treatment. Cytotherapy 2010; 12:615-25. [PMID: 20230221 DOI: 10.3109/14653241003631815] [Citation(s) in RCA: 143] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND AIMS Because of the inflammatory nature and extensive stromal compartment in pancreatic tumors, we investigated the role of mesenchymal stromal cells (MSC) to engraft selectively in pancreatic carcinomas and serve as anti-tumor drug delivery vehicles to control pancreatic cancer progression. METHODS Human pancreatic carcinoma cells, PANC-1, expressing renilla luciferase were orthotopically implanted into SCID mice and allowed to develop for 10 days. Firefly luciferase-transduced MSC or MSC expressing interferon (IFN)-beta were then injected intraperitoneally weekly for 3 weeks. Mice were monitored by bioluminescent imaging for expression of renilla (PANC-1) and firefly (MSC) luciferase. RESULTS MSC selectively homed to sites of primary and metastatic pancreatic tumors and inhibited tumor growth (P=0.032). The production of IFN-beta within the tumor site by MSC-IFN-beta further suppressed tumor growth (P=0.0000083). Prior studies indicated that MSC home to sites of inflammation; therefore, we sought to alter the tumor microenvironment through treatment with a potent anti-inflammatory agent. After treatment, inflammation-associated mediators were effectively down-regulated, including NFkappaB, vascular endothelial growth factor (VEGF) and interleukin (IL)-6 as well as chemokines involved in MSC migration (CCL3 and CCL25). Treatment with the anti-inflammatory agent CDDO-Me before and after MSC-IFN-beta injections resulted in reduction of MSC in the tumors and reversed the positive effect of tumor inhibition by MSC-IFN-beta alone (P=0.041). CONCLUSIONS These results suggest that MSC exhibit innate anti-tumor effects against PANC-1 cells and can serve as delivery vehicles for IFN-beta for the treatment of pancreatic cancer. However, these beneficial effects may be lost in therapies combining MSC with anti-inflammatory agents.
Collapse
Affiliation(s)
- Shannon Kidd
- Section of Molecular Hematology and Therapy, Department of Stem Cell Transplantation and Cellular Therapy, UT-MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
257
|
Rastegar F, Shenaq D, Huang J, Zhang W, Zhang BQ, He BC, Chen L, Zuo GW, Luo Q, Shi Q, Wagner ER, Huang E, Gao Y, Gao JL, Kim SH, Zhou JZ, Bi Y, Su Y, Zhu G, Luo J, Luo X, Qin J, Reid RR, Luu HH, Haydon RC, Deng ZL, He TC. Mesenchymal stem cells: Molecular characteristics and clinical applications. World J Stem Cells 2010; 2:67-80. [PMID: 21607123 PMCID: PMC3097925 DOI: 10.4252/wjsc.v2.i4.67] [Citation(s) in RCA: 161] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Revised: 06/26/2010] [Accepted: 07/03/2010] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are non-hematopoietic stem cells with the capacity to differentiate into tissues of both mesenchymal and non-mesenchymal origin. MSCs can differentiate into osteoblastic, chondrogenic, and adipogenic lineages, although recent studies have demonstrated that MSCs are also able to differentiate into other lineages, including neuronal and cardiomyogenic lineages. Since their original isolation from the bone marrow, MSCs have been successfully harvested from many other tissues. Their ease of isolation and ex vivo expansion combined with their immunoprivileged nature has made these cells popular candidates for stem cell therapies. These cells have the potential to alter disease pathophysiology through many modalities including cytokine secretion, capacity to differentiate along various lineages, immune modulation and direct cell-cell interaction with diseased tissue. Here we first review basic features of MSC biology including MSC characteristics in culture, homing mechanisms, differentiation capabilities and immune modulation. We then highlight some in vivo and clinical evidence supporting the therapeutic roles of MSCs and their uses in orthopedic, autoimmune, and ischemic disorders.
Collapse
Affiliation(s)
- Farbod Rastegar
- Farbod Rastegar, Deana Shenaq, Jiayi Huang, Wenli Zhang, Bing-Qiang Zhang, Bai-Cheng He, Liang Chen, Guo-Wei Zuo, Qing Luo, Qiong Shi, Eric R Wagner, Enyi Huang, Yanhong Gao, Jian-Li Gao, Stephanie H Kim, Jian-Zhong Zhou, Yang Bi, Yuxi Su, Gaohui Zhu, Jinyong Luo, Xiaoji Luo, Jiaqiang Qin, Russell R Reid, Hue H Luu, Rex C Haydon, Zhong-Liang Deng, Tong-Chuan He, Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
258
|
Human microglia transplanted in rat focal ischemia brain induce neuroprotection and behavioral improvement. PLoS One 2010; 5:e11746. [PMID: 20668522 PMCID: PMC2909196 DOI: 10.1371/journal.pone.0011746] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Accepted: 06/24/2010] [Indexed: 11/19/2022] Open
Abstract
Background and Purpose Microglia are resident immunocompenent and phagocytic cells of central nervous system (CNS), which produce various cytokines and growth factors in response to injury and thereby regulate disease pathology. The purpose of this study is to investigate the effects of microglial transplantation on focal cerebral ischemia model in rat. Methods Transient middle cerebral artery occlusion (MCAO) in rats was induced by the intraluminal filament technique. HMO6 cells, human microglial cell line, were transplanted intravenously at 48 hours after MCAO. Functional tests were performed and the infarct volume was measured at 7 and 14 days after MCAO. Migration and cell survival of transplanted microglial cells and host glial reaction in the brain were studied by immunohistochemistry. Gene expression of neurotrophic factors, cytokines and chemokines in transplanted cells and host rat glial cells was determined by laser capture microdissection (LCM) and quantitative real time-PCR. Results HMO6 human microglial cells transplantion group demonstrated significant functional recovery compared with control group. At 7 and 14 days after MCAO, infarct volume was significantly reduced in the HMO group. In the HMO6 group, number of apoptotic cells was time-dependently reduced in the infarct core and penumbra. In addition, number of host rat microglia/macrophages and reactive astrocytes was significantly decreased at 7 and 14 days after MCAO in the penumbra. Gene expression of various neurotrophic factors (GDNF, BDNF, VEGF and BMP7) and anti-inflammatory cytokines (IL4 and IL5) was up-regulated in transplanted HMO6 cells of brain tissue compared with those in culture. The expression of GDNF and VEGF in astrocytes in penumbra was significantly up-regulated in the HMO6 group. Conclusions Our results indicate that transplantation of HMO6 human microglial cells reduces ischemic deficits and apoptotic events in stroke animals. The results were mediated by modulation of gliosis and neuroinflammation, and neuroprotection provided by neurotrophic factors of endogenous and transplanted cells-origin.
Collapse
|
259
|
Petrie Aronin CE, Tuan RS. Therapeutic potential of the immunomodulatory activities of adult mesenchymal stem cells. ACTA ACUST UNITED AC 2010; 90:67-74. [PMID: 20301222 DOI: 10.1002/bdrc.20174] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Adult mesenchymal stem cells (MSCs) include a select population of resident cells within adult tissues, which retain the ability to differentiate along several tissue-specific lineages under defined media conditions and have finite expansion potential in vitro. These adult progenitor populations have been identified in various tissues, but it remains unclear exactly what role both transplanted and native MSCs play in processes of disease and regeneration. Interestingly, increasing evidence reveals a unique antiinflammatory immunomodulatory phenotype shared among this population, lending support to the idea that MSCs play a central role in early tissue remodeling responses where a controlled inflammatory response is required. However, additional evidence suggests that MSCs may not retain infinite immune privilege and that the context with which these cells are introduced in vivo may influence their immune phenotype. Therefore, understanding this dynamic microenvironment in which MSCs participate in complex feedback loops acting upon and being influenced by a plethora of secreted cytokines, extracellular matrix molecules, and fragments will be critical to elucidating the role of MSCs in the intertwined processes of immunomodulation and tissue repair.
Collapse
Affiliation(s)
- Caren E Petrie Aronin
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | | |
Collapse
|
260
|
Intravenous infusion of bone marrow mesenchymal stem cells improves brain function after resuscitation from cardiac arrest. Crit Care Med 2010; 36:S486-91. [PMID: 20449915 DOI: 10.1097/ccm.0b013e31818a8ff0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Allogeneic bone marrow mesenchymal stem cells were previously shown to improve myocardial function when administered intravenously after resuscitation from cardiac arrest in rats. Coincidental evidence of improved brain function prompted the present study. DESIGN Prospective, randomized, controlled study. SETTING University-affiliated research institute. SUBJECTS Male Sprague-Dawley rats. INTERVENTIONS Using an established model in 20 male Sprague-Dawley rats in which 6 mins of untreated cardiac arrest was followed by cardiopulmonary resuscitation, animals were randomized to receive 5 x 10(6) mesenchymal stem cells labeled with PKH26 in phosphate buffer solution or phosphate buffer solution alone as a placebo at 2 hrs after restoration of spontaneous circulation. The stem cells or buffer diluent were injected into a catheter advanced from the jugular vein into the right atrium. MEASUREMENTS AND MAIN RESULTS Outcome measurements in addition to 35-day survival included somatosensor testing of capability for removal of an adhesive patch applied to both front paws, testing of motor function using a rotating cylinder, and observational scoring of the severity of neurologic impairment. Labeled mesenchymal stem cells were subsequently identified and counted in 5 microm sections obtained from defined sites in the harvested brain. Immunohistochemistry was used to identify neural cells differentiation of mesenchymal stem cells. Adhesive removal, motor function test, neurologic severity score, and 35-day survival were each significantly improved in comparison with control animals. Labeled mesenchymal stem cells were identified in the hippocampus, cortex, pons, medulla, and cerebellum and expressed protein markers phenotypic neural cells. CONCLUSIONS Mesenchymal stem cells injected into the right atrium of rats after resuscitation from cardiac arrest were identified in brains harvested 35 days later. Brain function was significantly improved. Accordingly, venous injection of mesenchymal stem cells after cardiopulmonary resuscitation has promise of minimizing the severity of postresuscitation neurologic impairment.
Collapse
|
261
|
Yilmaz G, Alexander JS, Erkuran Yilmaz C, Granger DN. Induction of neuro-protective/regenerative genes in stem cells infiltrating post-ischemic brain tissue. EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2010; 2:11. [PMID: 20509949 PMCID: PMC2893124 DOI: 10.1186/2040-7378-2-11] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Accepted: 05/28/2010] [Indexed: 12/25/2022]
Abstract
BACKGROUND Although the therapeutic potential of bone marrow-derived stromal stem cells (BMSC) has been demonstrated in different experimental models of ischemic stroke, it remains unclear how stem cells (SC) induce neuroprotection following stroke. In this study, we describe a novel method for isolating BMSC that infiltrate postischemic brain tissue and use this method to identify the genes that are persistently activated or depressed in BMSC that infiltrate brain tissue following ischemic stroke. METHODS Ischemic strokes were induced in C57BL/6 mice by middle cerebral artery occlusion for 1 h, followed by reperfusion. BMSC were isolated from H-2 Kb-tsA58 (immortomouse) mice, and were administered (i.v.) 24 h after reperfusion. At the peak of therapeutic improvement (14 days after the ischemic insult), infarcted brain tissue was isolated, and the BMSC were isolated by culturing at 33 degrees C. Microarray analysis and RT-PCR were performed to compare differential gene expression between naïve and infiltrating BMSC populations. RESULTS Z-scoring revealed dramatic differences in the expression of extracellular genes between naïve and infiltrating BMSC. Pair-wise analysis detected 80 extracellular factor genes that were up-regulated (>/= 2 fold, P < 0.05, Benjamini-Hochberg correction) between naïve and infiltrated BMSC. Although several anticipated neuroregenerative, nerve guidance and angiogenic factor (e.g., bFGF, bone morphogenetic protein, angiopoietins, neural growth factor) genes exhibited an increased expression, a remarkable induction of genes for nerve guidance survival (e.g., cytokine receptor-like factor 1, glypican 1, Dickkopf homolog 2, osteopontin) was also noted. CONCLUSIONS BMSC infiltrating the post-ischemic brain exhibit persistent epigenetic changes in gene expression for numerous extracellular genes, compared to their naïve counterparts. These genes are relevant to the neuroprotection, regeneration and angiogenesis previously described following stem cell therapy in animal models of ischemic stroke.
Collapse
Affiliation(s)
- Gokhan Yilmaz
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, Shreveport, LA, USA.
| | | | | | | |
Collapse
|
262
|
Li XY, Li J, Feng DF, Gu L. Diffuse axonal injury induced by simultaneous moderate linear and angular head accelerations in rats. Neuroscience 2010; 169:357-69. [PMID: 20451584 DOI: 10.1016/j.neuroscience.2010.04.075] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2010] [Revised: 04/12/2010] [Accepted: 04/26/2010] [Indexed: 11/19/2022]
Abstract
Diffuse axonal injury (DAI) is one of the most common and important pathologic features of human traumatic brain injury (TBI), accounting for high mortality and development of persistent post-traumatic neurologic sequelae. Although a relatively high number of therapies have been shown to be effective in experimental models, there are currently few treatments that are effective for improving the prognosis of clinical DAI. A major reason is the failure of current models to validly reproduce the pathophysiological characteristics observed after clinical DAI. In the present study, we employed a specially designed, highly controllable model to induce a sudden rotation in the coronal plane (75 degrees rotation at 1.6x10(4) degrees/s) combined with lateral translation (1.57 cm displacement at 3.4x10(2) cm/s) to the rat's head. We were interested in discovering whether the combined accelerations could reproduce the pathophysiological changes analogous to those seen in human DAI. The axonal injury as assessed with amyloid protein precursor (APP) as a marker was consistently present in all injured rats. The commonly injured brain regions included the subcortical regions, deep white matter, corpus callosum and brain stem. The evolution of APP accumulations in brain sections depicted the detailed progression of axonal pathology. Ultrastructural studies gave further insights into the presence and progression of axonal injury. All injured rats exhibited transient physiological dysfunction, as well as immediate and dramatic neurological impairment that still persisted at 14 days after injury. These results suggest that this model reproduced the major pathophysiological changes analogous to those observed after severe clinical TBI and provides an attractive vehicle for experimental brain injury research.
Collapse
Affiliation(s)
- X Y Li
- Department of Neurosurgery, No.3 People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | | | | | | |
Collapse
|
263
|
Seyfried DM, Han Y, Yang D, Ding J, Shen LH, Savant-Bhonsale S, Chopp M. Localization of bone marrow stromal cells to the injury site after intracerebral hemorrhage in rats. J Neurosurg 2010; 112:329-35. [PMID: 19284233 DOI: 10.3171/2009.2.jns08907] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
OBJECT Previous studies demonstrated that intravascular injection of bone marrow stromal cells (BMSCs) significantly improved neurological functional recovery in a rat model of intracerebral hemorrhage (ICH). To further investigate the fate of transplanted cells, we examined the effect of male rat BMSCs administered to female rats after ICH. METHODS Twenty-seven female Wistar rats were subjected to ICH surgery. At 24 hours after ICH, these rats were randomly divided into 3 groups and injected intravenously with 1 ml phosphate-buffered saline or 0.5 million or 1 million male rat BMSCs in phosphate-buffered saline. To evaluate the neurological functional outcome, each rat was subjected to a series of behavioral tests (modified neurological severity score and corner turn test) at 1, 7, and 14 days after ICH. The rats were anesthetized intraperitoneally and killed, and the brain tissues were processed at Day 14 after ICH. Immunohistochemistry and in situ hybridization were used to identify cell-specific markers. RESULTS The male rat BMSCs significantly improved the neurological functional outcome and also significantly diminished tissue loss when intravenously transplanted into the rats after ICH. Immunoassay for bromodeoxyuridine (BrdU) and neuronal markers demonstrated a significant increase in the number of BrdU-positive cells, which indicated endogenous neurogenesis, and a significant increase in the number of cells positive for immature neuronal markers. In situ hybridization showed that more BMSCs resided around the hematoma of the rats treated with the 1-million-cell dose compared with the 0.5-million-cell-dose group. In addition, a subfraction of Y chromosome-positive cells were co-immunostained with the neuronal marker microtubule-associated protein-2 or the astrocytic marker glial fibrillary acidic protein. CONCLUSIONS Male rat BMSCs improve neurological outcome and increase histochemical parameters of neurogenesis when administered to female rats after ICH. This study has shown that the intravenously administered male rat BMSCs enter the brain, migrate to the perihematomal area, and express parenchymal markers.
Collapse
Affiliation(s)
- Donald M Seyfried
- Department of Neurosurgery, Henry Ford Health System, Detroit, Michigan, USA
| | | | | | | | | | | | | |
Collapse
|
264
|
Xin H, Li Y, Shen LH, Liu X, Wang X, Zhang J, Pourabdollah-Nejad D S, Zhang C, Zhang L, Jiang H, Zhang ZG, Chopp M. Increasing tPA activity in astrocytes induced by multipotent mesenchymal stromal cells facilitate neurite outgrowth after stroke in the mouse. PLoS One 2010; 5:e9027. [PMID: 20140248 PMCID: PMC2815778 DOI: 10.1371/journal.pone.0009027] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Accepted: 01/11/2010] [Indexed: 10/31/2022] Open
Abstract
We demonstrate that tissue plasminogen activator (tPA) and its inhibitors contribute to neurite outgrowth in the central nervous system (CNS) after treatment of stroke with multipotent mesenchymal stromal cells (MSCs). In vivo, administration of MSCs to mice subjected to middle cerebral artery occlusion (MCAo) significantly increased activation of tPA and downregulated PAI-1 levels in the ischemic boundary zone (IBZ) compared with control PBS treated mice, concurrently with increases of myelinated axons and synaptophysin. In vitro, MSCs significantly increased tPA levels and concomitantly reduced plasminogen activator inhibitor 1 (PAI-1) expression in astrocytes under normal and oxygen and glucose deprivation (OGD) conditions. ELISA analysis of conditioned medium revealed that MSCs stimulated astrocytes to secrete tPA. When primary cortical neurons were cultured in the conditioned medium from MSC co-cultured astrocytes, these neurons exhibited a significant increase in neurite outgrowth compared to conditioned medium from astrocytes alone. Blockage of tPA with a neutralizing antibody or knock-down of tPA with siRNA significantly attenuated the effect of the conditioned medium on neurite outgrowth. Addition of recombinant human tPA into cortical neuronal cultures also substantially enhanced neurite outgrowth. Collectively, these in vivo and in vitro data suggest that the MSC mediated increased activation of tPA in astrocytes promotes neurite outgrowth after stroke.
Collapse
Affiliation(s)
- Hongqi Xin
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Yi Li
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Li Hong Shen
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Xianshuang Liu
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Xinli Wang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Jing Zhang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
| | | | - Chunling Zhang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Li Zhang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Hao Jiang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
- Department of Physics, Oakland University, Rochester, Michigan, United States of America
- * E-mail:
| |
Collapse
|
265
|
Jiang H, Gomez-Manzano C, Lang FF, Alemany R, Fueyo J. Oncolytic adenovirus: preclinical and clinical studies in patients with human malignant gliomas. Curr Gene Ther 2010; 9:422-7. [PMID: 19860656 DOI: 10.2174/156652309789753356] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Oncolytic adenoviruses are emerging as a promising alternative therapy for glioma patients and are currently being tested in clinic. In this review, we summarize our experience with gene-based therapy targeting RB pathway in gliomas. Our study has evolved from the development of RB-expressing adenoviral vectors to the characterization of the oncolytic effects on gliomas of the replication competent adenoviruses Delta-24, Delta-24-RGD and ICOVIR. We also review the successful combination of the viruses with chemotherapies that are routinely used in glioma patients, the efficacy of Delta-24-RGD against brain tumor stem cells, the newly described adenovirus-induced autophagy and the potential for the systemic delivery of the oncolytic viruses with human mesenchymal stem cells. Finally, we comment on the preclinical and clinical studies of p53 expressing adenoviral vector and the lessons learned from the experience of Onyx-015, the first oncolytic adenovirus tested in clinical setting.
Collapse
Affiliation(s)
- Hong Jiang
- Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, Texas, USA.
| | | | | | | | | |
Collapse
|
266
|
Chung DJ, Choi CB, Lee SH, Kang EH, Lee JH, Hwang SH, Han H, Lee JH, Choe BY, Lee SY, Kim HY. Intraarterially delivered human umbilical cord blood-derived mesenchymal stem cells in canine cerebral ischemia. J Neurosci Res 2010; 87:3554-67. [PMID: 19642203 DOI: 10.1002/jnr.22162] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The present study examined the effects of human umbilical cord blood-derived mesenchymal stem cells (HUCB-derived MSCs) delivered through the basilar artery in a canine thromboembolic brain ischemia model. Cerebral ischemia was induced through occlusion of the middle cerebral artery by injecting thrombus emboli into 10 beagles. In the HUCBC group (n = 5), 1 x 10(6) HUCB-derived MSCs were transplanted through the basilar artery 1 day after ischemic induction using an endovascular interventional approach. In the control group (n = 5), phosphate-buffered saline (PBS) was injected in the same manner in as the HUCBC group. Upon neurobehavioral examination, earlier recovery was observed in the HUCBC group. The HUCBC group showed a decrease in the infarction volume at 1 week after cerebral ischemic induction, whereas the control group showed an increase in the infarction volume at 1 week, by magnetic resonance image analysis. Transplanted cells had differentiated into neurons and astrocytes and were observed in and around endothelial cells that were positive for von Willebrand factor (vWF). HUCB-derived MSCs expressed neuroprotective factors, such as brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF), at 4 weeks after the transplantation. The transplanted cells demonstrated their efficacy by reducing the infarction lesion volume and through earlier recovery from the neurological deficit. These results suggest that intraarterial transplantation of HUCB-derived MSCs could be useful in clinical treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Dai-Jung Chung
- Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
267
|
Horiuchi M, Mogi M, Iwai M. The angiotensin II type 2 receptor in the brain. J Renin Angiotensin Aldosterone Syst 2009; 11:1-6. [PMID: 19861353 DOI: 10.1177/1470320309347793] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Recent clinical studies indicate that blockade of the renin-angiotensin system is important to prevent stroke, and accumulating results of basic research also indicate the possible involvement of the central renin-angiotensin system in ischaemic brain damage and cognition. When the angiotensin II type 1 receptor is blocked by an angiotensin type 1 receptor blocker, unbound angiotensin II acts preferentially on the angiotensin II type 2 (AT(2)) receptor. These results suggest the pathophysiological importance of the AT(2) receptor in the clinical use of angiotensin type 1 receptor blockers, which are widely used in patients with hypertension with the expectation of a decrease in the onset of cardiovascular and cerebrovascular disease. We review here the possible roles of AT(2) receptor activation in the brain, focusing on ischaemic stroke, cognitive function and neurogenesis, and potential effects of specific AT(2) receptor agonists.
Collapse
Affiliation(s)
- Masatsugu Horiuchi
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Shitsukawa, Tohon, Ehime 791-0295, Japan.
| | | | | |
Collapse
|
268
|
Barbosa da Fonseca LM, Gutfilen B, Rosado de Castro PH, Battistella V, Goldenberg RCS, Kasai-Brunswick T, Chagas CLR, Wajnberg E, Maiolino A, Salles Xavier S, Andre C, Mendez-Otero R, de Freitas GR. Migration and homing of bone-marrow mononuclear cells in chronic ischemic stroke after intra-arterial injection. Exp Neurol 2009; 221:122-8. [PMID: 19853605 DOI: 10.1016/j.expneurol.2009.10.010] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Revised: 10/06/2009] [Accepted: 10/15/2009] [Indexed: 12/15/2022]
Abstract
Cell-based treatments have been considered a promising therapy for neurological diseases. However, currently there are no clinically available methods to monitor whether the transplanted cells reach and remain in the brain. In this study we investigated the feasibility of detecting the distribution and homing of autologous bone-marrow mononuclear cells (BMMCs) labeled with Technetium-99 m ((99m)Tc) in a cell-based therapy clinical study for chronic ischemic stroke. Six male patients (ages 24-65 years) with ischemic cerebral infarcts within the middle cerebral artery (MCA) between 59 and 82 days were included. Cell dose ranged from 1.25x10(8) to 5x10(8). Approximately 2x10(7) cells were labeled with (99m)Tc and intra-arterially delivered together with the unlabeled cells via a catheter navigated to the MCA. None of the patients showed any complications on the 120-day follow-up. Whole body scintigraphies indicated cell homing in the brain of all patients at 2 h, while the remaining uptake was mainly distributed to liver, lungs, spleen, kidneys and bladder. Moreover, quantification of uptake in Single-Photon Emission Computed Tomography (SPECT) at 2 h showed preferential accumulation of radioactivity in the hemisphere affected by the ischemic infarct in all patients. However, at 24 h homing could only distinguished in the brains of 2 patients, while in all patients uptake was still seen in the other organs. Taken together, these results indicate that labeling of BMMCs with (99m)Tc is a safe and feasible technique that allows monitoring the migration and engraftment of intra-arterially transplanted cells for at least 24 h.
Collapse
Affiliation(s)
- Lea Mirian Barbosa da Fonseca
- Hospital Universitário Clementino Fraga Filho, Departamento de Radiologia, subsolo, Universidade Federal do Rio de Janeiro, Ilha do Fundão, Rio de Janeiro, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
269
|
Detante O, Moisan A, Dimastromatteo J, Richard MJ, Riou L, Grillon E, Barbier E, Desruet MD, De Fraipont F, Segebarth C, Jaillard A, Hommel M, Ghezzi C, Remy C. Intravenous administration of 99mTc-HMPAO-labeled human mesenchymal stem cells after stroke: in vivo imaging and biodistribution. Cell Transplant 2009; 18:1369-79. [PMID: 19849895 DOI: 10.3727/096368909x474230] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Human mesenchymal stem cells (hMSC) are a promising source for cell therapy after stroke. To deliver these cells, an IV injection appears safer than a local graft. We aimed to assess the whole-body biodistribution of IV-injected (99m)Tc-HMPAO-labeled hMSC in normal rats (n = 9) and following a right middle cerebral artery occlusion (MCAo, n = 9). Whole-body nuclear imaging, isolated organ counting (at 2 and 20 h after injection) and histology were performed. A higher activity was observed in the right damaged hemisphere of the MCAo group [6.5 +/- 0.9 x 10(-3) % of injected dose (ID)/g] than in the control group (3.6 +/- 1.2 x 10(-3) %ID/g), 20 h after injection. In MCAo rats, right hemisphere activity was higher than that observed in the contralateral hemisphere at 2 h after injection (11.6 +/- 2.8 vs. 9.8 +/- 1.7 x 10(-3) %ID/g). Following an initial hMSC lung accumulation, there was a decrease in pulmonary activity from 2 to 20 h after injection in both groups. The spleen was the only organ in which activity increased between 2 and 20 h. The presence of hMSC was documented in the spleen, liver, lung, and brain following histology. IV-injected hMSC are transiently trapped in the lungs, can be sequestered in the spleen, and are predominantly eliminated by kidneys. After 20 h, more hMSC are found in the ischemic lesion than into the undamaged cerebral tissue. IV delivery of hMSC could be the initial route for a clinical trial of tolerance.
Collapse
|
270
|
|
271
|
Yu SJ, Soncini M, Kaneko Y, Hess DC, Parolini O, Borlongan CV. Amnion: a potent graft source for cell therapy in stroke. Cell Transplant 2009; 18:111-8. [PMID: 19499700 DOI: 10.3727/096368909788341243] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Regenerative medicine is a new field primarily based on the concept of transplanting exogenous or stimulating endogenous stem cells to generate biological substitutes and improve tissue functions. Recently, amnion-derived cells have been reported to have multipotent differentiation ability, and these cells have attracted attention as a novel cell source for cell transplantation therapy. Cells isolated from amniotic membrane can differentiate into all three germ layers, have low immunogenicity and anti-inflammatory function, and do not require the destruction of human embryos for their isolation, thus circumventing the ethical debate commonly associated with the use of human embryonic stem cells. Accumulating evidence now suggests that the amnion, which had been discarded after parturition, is a highly potent transplant material in the field of regenerative medicine. In this report, we review the current progress on the characterization of MSCs derived from the amnion as a remarkable transplantable cell population with therapeutic potential for multiple CNS disorders, especially stroke.
Collapse
Affiliation(s)
- Seong Jin Yu
- Department of Neurology, Medical College of Georgia, Augusta, GA, USA
| | | | | | | | | | | |
Collapse
|
272
|
Tadauchi A, Narita Y, Kagami H, Niwa Y, Ueda M, Goto H. Novel cell-based therapeutic strategy for ischemic colitis with use of bone marrow-derived mononuclear cells in rats. Dis Colon Rectum 2009; 52:1443-51. [PMID: 19617758 DOI: 10.1007/dcr.0b013e3181a79549] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
PURPOSE Ischemic colitis is a common disorder of the large bowel. In the clinical setting, some patients suffer refractory ischemic colitis regardless of conventional treatment. Meanwhile, bone marrow-derived mononuclear cells are known to accelerate neovascularization. The purpose of this study was to verify the effects of bone marrow-derived mononuclear cells on ischemic colitis in rats. METHODS An ischemic colitis model was established by partial obstruction of the rectum and interruption of the marginal vessel in the immunodeficient rat. Bone marrow-derived mononuclear cells from a Wistar rat were injected into the ischemic area one day later than the ischemia (Group MNC). As a control, phosphate-buffered saline was injected in the same manner (Group PBS). Seven days after cell transplantation, each rat was evaluated for histology and colic motility. RESULTS Compared with Group PBS scores, the Group MNC macroscopic and microscopic colitis severity scores were significantly reduced. Moreover, the density of the capillary and myenteric plexus was significantly higher in Group MNC than in Group PBS (9.55 +/- 0.74 vs. 4.61 +/- 0.22, respectively, P < 0.01; and 8.57 +/- 0.41 vs. 5.93 +/- 0.31, respectively, P < 0.02). The whole-gut transit time was significantly shorter in Group MNC compared with Group PBS (472.7 +/- 17.6 vs. 584.8 +/- 24.0 minutes, respectively, P < 0.01). Transplanted cells were detected in all layers of the intestinal wall; however, these cells did not differentiate into vascular or neural cells. CONCLUSIONS These results suggest that transplantation of bone marrow-derived mononuclear cells might enhance not only tissue regeneration and angiogenesis but also neurogenesis. Transplantation of bone marrow-derived mononuclear cells may be a useful therapeutic strategy for ischemic colitis.
Collapse
Affiliation(s)
- Akimitsu Tadauchi
- Department of Gastroenterology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | |
Collapse
|
273
|
Hayase M, Kitada M, Wakao S, Itokazu Y, Nozaki K, Hashimoto N, Takagi Y, Dezawa M. Committed neural progenitor cells derived from genetically modified bone marrow stromal cells ameliorate deficits in a rat model of stroke. J Cereb Blood Flow Metab 2009; 29:1409-20. [PMID: 19436312 DOI: 10.1038/jcbfm.2009.62] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Bone marrow stromal cells (MSCs) are an excellent source of cells for treating a variety of central nervous system diseases. In this study, we report the efficient induction of committed neural progenitor cells from rat and human MSCs (NS-MSCs) by introduction of cells with the intracellular domain of Notch-1 followed by growth in the free-floating culture system. NS-MSCs successfully formed spheres, in which cells highly expressed the neural precursor cell markers. The commitment of spheres to neural lineage cells was confirmed by their successful differentiation into neuronal cells when exposed to a differentiation medium. To determine the therapeutic potential of NS-MSCs, cells were transplanted into the cortex and striatum in a rat model of focal cerebral ischemia. The survival, distribution, and integration of NS-MSCs in the host brain were very high, and at day 100, grafted NS-MSCs were positive for dopaminergic, glutamatergic, and gamma-amino butyric acid(GABA)ergic neuronal markers. They extended long neurites for nearly 6.3 mm and many of these expressed synaptophysin. Significant behavioral recovery was also observed in limb-placing and water-maze tests. These suggest a high potential for this MSC approach in the replenishment of neural cells for stroke and for a wide range of neurodegenerative conditions that require various types of neural cells.
Collapse
Affiliation(s)
- Makoto Hayase
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507 , Japan
| | | | | | | | | | | | | | | |
Collapse
|
274
|
Treatment of stroke and intracerebral hemorrhage with cellular and pharmacological restorative therapies. CEREBRAL HEMORRHAGE 2009; 105:79-83. [DOI: 10.1007/978-3-211-09469-3_16] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
275
|
Abstract
Functional recovery after cerebral ischemia is mediated by the regeneration of vascular networks and the restoration of synaptic architecture. Netrins have been implicated in neuronal pathfinding and angiogenesis. In this study, we investigated the expression of Netrin-4 and its putative receptors, deleted in colorectal cancer (DCC), Unc5A, and Unc5B after distal middle cerebral artery occlusion in mice. Netrin-4 protein was also administered intracerebroventricularly to examine its effect on angiogenesis and behavioral recovery. Netrin-4 protein was highly upregulated in the ischemic core as soon as 1 day after cerebral ischemia, with subsequent downregulation after 1 week. Its expression was limited to the area of blood-brain barrier damage and was seen on both blood vessels and astrocytic foot processes. Although there was not a significant upregulation of the putative Netrin-4 receptor Unc5A and Unc5B, there was a significant increase in expression of the DCC receptor on neuronal processes in the peri-infarct cortex. Intracerebroventricular administration of Netrin-4 into the ischemic brain increased blood vessel density, endothelial proliferation, and improved behavioral recovery at 1 week after stroke, but did not have an effect on blood-brain barrier permeability or infarct size. These findings suggest that Netrin-4 may improve poststroke functional recovery by enhancing blood vessel proliferation.
Collapse
|
276
|
Li Y, Chopp M. Marrow stromal cell transplantation in stroke and traumatic brain injury. Neurosci Lett 2009; 456:120-3. [PMID: 19429146 DOI: 10.1016/j.neulet.2008.03.096] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Revised: 02/24/2008] [Accepted: 03/01/2008] [Indexed: 12/19/2022]
Abstract
There is a paucity of therapies for most central nervous system (CNS) disorders. Bone marrow stromal cells (MSCs) are a mixed cell population, including stem and progenitor cells, and are currently a strong candidate for cell-based therapy in "brain attack", including stroke, and traumatic brain injury (TBI), since they are easily isolated and can be expanded in culture from patients without ethical and technical problems. Although it has been suggested that trans-differentiation of MSCs into cells of neural lineage may occur in vitro, no one has yet observed that MSCs give rise to fully differentiated and functional neurons in vivo. The overwhelming body of data indicate that bioactive factors secreted by MSCs in response to the local environment underlie the tissue restorative effects of MSCs. The MSCs that are employed in this therapy are not necessarily stem cells, but progenitor and differentiated cells that escape immune system surveillance and survive in the CNS even for transplantation of allogeneic or xenogeneic MSCs. The injured CNS is stimulated by the MSCs to amplify its intrinsic restorative processes. Treatment of damaged brain with MSCs promotes functional recovery, and facilitates CNS endogenous plasticity and remodeling. The current mini-review is mainly based on our data and focuses on possible cellular and molecular mechanisms of interaction of MSCs with glia, neurons and vessels after brain attack. The transplantation of MSCs opens up new avenues of cell therapy and may provide an effective treatment for various CNS diseases.
Collapse
Affiliation(s)
- Yi Li
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, United States
| | | |
Collapse
|
277
|
Yao RQ, Zhang L, Wang W, Li L. Cornel iridoid glycoside promotes neurogenesis and angiogenesis and improves neurological function after focal cerebral ischemia in rats. Brain Res Bull 2009; 79:69-76. [PMID: 19150488 DOI: 10.1016/j.brainresbull.2008.12.010] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2008] [Revised: 12/16/2008] [Accepted: 12/16/2008] [Indexed: 01/17/2023]
Abstract
The aim of this study was to investigate the effects of cornel iridoid glycoside (CIG), an ingredient extracted from a traditional Chinese herb Cornus officinalis, on neurological function and neurogenesis after ischemic stroke. CIG was intragastrically administered to rats in doses of 20, 60 and 180 mg/kg/day, starting 3 h after the onset of middle cerebral artery occlusion (MCAO). The behavioral test was performed by using the modified neurological severity score (mNSS). Rats were sacrificed 7, 14, or 28 days after ischemia occurred. Neurogenesis and angiogenesis were detected by using immunofluorescence staining. The messenger ribonucleic acid (mRNA) expression of vascular endothelial growth factor (VEGF) and its receptor Flk-1 was measured by RT-PCR, and the protein expression of VEGF was determined by Western blotting analysis. The treatment with CIG at the doses of 60 and 180 mg/kg/day significantly improved neurological function, and increased the number of bromodeoxyuridine (BrdU)-positive cells and nestin-positive cells in the subventricular zone of rats 7, 14 and 28 days after ischemia. The number of newly mature neurons and blood vessels in striatum, as indicated by BrdU/NeuN and vWF immunoreactivity, respectively, was also increased in CIG-treated rats 28 days after stroke. CIG treatment obviously enhanced the mRNA expression of VEGF and its receptor Flk-1 and the protein expression of VEGF 7 and 28 days after ischemia. The results indicated that CIG promoted neurogenesis and angiogenesis and improved neurological function after ischemia in rats, and the mechanism might be related to CIG's increasing VEGF and Flk-1 in the brain.
Collapse
Affiliation(s)
- R-Q Yao
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, 45 Chang-chun Street, Beijing 100053, PR China
| | | | | | | |
Collapse
|
278
|
Matsuda R, Yoshikawa M, Kimura H, Ouji Y, Nakase H, Nishimura F, Nonaka JI, Toriumi H, Yamada S, Nishiofuku M, Moriya K, Ishizaka S, Nakamura M, Sakaki T. Cotransplantation of Mouse Embryonic Stem Cells and Bone Marrow Stromal Cells following Spinal Cord Injury Suppresses Tumor Development. Cell Transplant 2009; 18:39-54. [DOI: 10.3727/096368909788237122] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Embryonic stem (ES) cells are a potential source for treatment of spinal cord injury (SCI). Although one of the main problems of ES cell-based cell therapy is tumor formation, there is no ideal method to suppress tumor development. In this study, we examined whether transplantation with bone marrow stromal cells (BMSCs) prevented tumor formation in SCI model mice that received ES cell-derived grafts containing both undifferentiated ES cells and neural stem cells. Embryoid bodies (EBs) formed in 4-day hanging drop cultures were treated with retinoic acid (RA) at a low concentration of 5 × 10–9 M for 4 days, in order to allow some of the ES cells to remain in an undifferentiated state. RA-treated EBs were enzymatically digested into single cells and used as ES cell-derived graft cells. Mice transplanted with ES cell-derived graft cells alone developed tumors at the grafted site and behavioral improvement ceased after day 21. In contrast, no tumor development was observed in mice cotransplanted with BMSCs, which also showed sustained behavioral improvement. In vitro results demonstrated the disappearance of SSEA-1 expression in cytochemical examinations, as well as attenuated mRNA expressions of the undifferentiated markers Oct3/4, Utf1, Nanog, Sox2, and ERas by RT-PCR in RA-treated EBs cocultured with BMSCs. In addition, MAP2-immunopositive cells appeared in the EBs cocultured with BMSCs. Furthermore, the synthesis of NGF, GDNF, and BDNF was confirmed in cultured BMSCs, while immunohistochemical examinations demonstrated the survival of BMSCs and their maintained ability of neurotrophic factor production at the grafted site for up to 5 weeks after transplantation. These results suggest that BMSCs induce undifferentiated ES cells to differentiate into a neuronal lineage by neurotrophic factor production, resulting in suppression of tumor formation. Cotransplantation of BMSCs with ES cell-derived graft cells may be useful for preventing the development of ES cell-derived tumors.
Collapse
Affiliation(s)
- Ryosuke Matsuda
- Department of Neurosurgery, Nara Medical University, Nara 634–8521, Japan
| | - Masahide Yoshikawa
- Department of Parasitology, Nara Medical University, Nara 634–8521, Japan
| | - Hajime Kimura
- Department of Neurosurgery, Nara Medical University, Nara 634–8521, Japan
| | - Yukiteru Ouji
- Department of Parasitology, Nara Medical University, Nara 634–8521, Japan
| | - Hiroyuki Nakase
- Department of Neurosurgery, Nara Medical University, Nara 634–8521, Japan
| | - Fumihiko Nishimura
- Department of Neurosurgery, Nara Medical University, Nara 634–8521, Japan
| | - Jun-Ichi Nonaka
- Department of Neurosurgery, Nara Medical University, Nara 634–8521, Japan
| | - Hayato Toriumi
- Department of Neurosurgery, Nara Medical University, Nara 634–8521, Japan
| | - Shuichi Yamada
- Department of Neurosurgery, Nara Medical University, Nara 634–8521, Japan
| | - Mariko Nishiofuku
- Department of Parasitology, Nara Medical University, Nara 634–8521, Japan
| | - Kei Moriya
- Department of Parasitology, Nara Medical University, Nara 634–8521, Japan
| | - Shigeaki Ishizaka
- Department of Parasitology, Nara Medical University, Nara 634–8521, Japan
| | | | - Toshisuke Sakaki
- Department of Neurosurgery, Nara Medical University, Nara 634–8521, Japan
| |
Collapse
|
279
|
Abstract
Stem cells, as subjects of study for use in treating neurological diseases, are envisioned as a replacement for lost neurons and glia, a means of trophic support, a therapeutic vehicle, and, more recently, a tool for in vitro modeling to understand disease and to screen and personalize treatments. In this review we analyze the requirements of stem cell-based therapy for clinical translation, advances in stem cell research toward clinical application for neurological disorders, and different animal models used for analysis of these potential therapies. We focus on Parkinson's disease (typically defined by the progressive loss of dopaminergic nigral neurons), stroke (neurodegeneration associated with decreased blood perfusion in the brain), and multiple sclerosis (an autoimmune disorder that generates demyelination, axonal damage, astrocytic scarring, and neurodegeneration in the brain and spinal cord). We chose these disorders for their diversity and the number of people affected by them. An additional important consideration was the availability of multiple animal models in which to test stem cell applications for these diseases. We also discuss the relationship between the limited number of systematic stem cell studies performed in animals, in particular nonhuman primates and the delayed progress in advancing stem cell therapies to clinical success.
Collapse
Affiliation(s)
- Valerie L Joers
- Neuroscience Training Program, University of Wisconsin, Madison, WI, USA
| | | |
Collapse
|
280
|
Croft AP, Przyborski SA. Mesenchymal stem cells expressing neural antigens instruct a neurogenic cell fate on neural stem cells. Exp Neurol 2008; 216:329-41. [PMID: 19159625 DOI: 10.1016/j.expneurol.2008.12.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Revised: 12/08/2008] [Accepted: 12/09/2008] [Indexed: 01/01/2023]
Abstract
The neurogenic response to injury in the postnatal brain is limited and insufficient for restoration of function. Recent evidence suggests that transplantation of mesenchymal stem cells (MSCs) into the injured brain is associated with improved functional recovery, mediated in part through amplification in the endogenous neurogenic response to injury. In the current study we investigate the interactions between bone marrow-derived MSCs and embryonic neural stem cells (NSCs) plus their differentiated progeny using an in vitro co-culture system. Two populations of MSCs were used, MSCs induced to express neural antigens (nestin+, Tuj-1+, GFAP+) and neural antigen negative MSCs. Following co-culture of induced MSCs with differentiating NSC/progenitor cells a significant increase in Tuj-1+ neurons was detected compared to co-cultures of non-induced MSCs in which an increase in astrocyte (GFAP+) differentiation was observed. The effect was mediated by soluble interactions between the two cell populations and was independent of any effect on cell death and proliferation. Induced and non-induced MSCs also promoted the survival of Tuj-1+ cell progeny in long-term cultures and both promoted axonal growth, an effect also seen in differentiating neuroblastoma cells. Therefore, MSCs provide instructive signals that are able to direct the differentiation of NSCs and promote axonal development in neuronal progeny. The data indicates that the nature of MSC derived signals is dependent not only on their microenvironment but on the developmental status of the MSCs. Pre-manipulation of MSCs prior to transplantation in vivo may be an effective means of enhancing the endogenous neurogenic response to injury.
Collapse
Affiliation(s)
- Adam P Croft
- School of Biological and Biomedical Sciences, Durham University, South Road, Durham DH1 3LE, UK
| | | |
Collapse
|
281
|
Abstract
We investigated vascular changes after stroke using magnetic resonance imaging (MRI) microvascular density (MVD) measurement. T(2) and T(2)(*) were measured in eight rats before and after injecting an intravascular superparamagnetic iron oxide contrast agent to derive the corresponding transverse relaxation shift. Reliability of MRI for measurement of MVD was compared with corresponding sections immunostained with von Willebrand factor (vWF) 2 weeks after stroke. The intracorrelation coefficient (ICC) and its 95% lower bound (LB) was high in the ischemic recovery region (ICC=0.753), moderate in the contralateral area of normal brain tissue (ICC=0.70), and low in the ischemic core (ICC=0.24). A very good agreement (ICC=0.85) and correlation (r=0.90) were observed using only the recovery region and normal contralateral hemisphere (ICC=0.85; 95% LB=0.78; P<0.05). The mean MRI MVD in the center of the core lesion (26+/-9 per mm(2)) was lower than in the recovery region (209+/-60 per mm(2)) or contralateral normal hemisphere (313+/-32 per mm(2)). However, large errors in MRI MVD were encountered in the ischemic core. Our data demonstrate that MRI MVD measurements can quantitatively evaluate microvascular changes in the brain tissue after stroke, if the MVD is not extremely low as in the ischemic core.
Collapse
|
282
|
Dharmasaroja P. Bone marrow-derived mesenchymal stem cells for the treatment of ischemic stroke. J Clin Neurosci 2008; 16:12-20. [PMID: 19017556 DOI: 10.1016/j.jocn.2008.05.006] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2008] [Revised: 05/12/2008] [Accepted: 05/17/2008] [Indexed: 02/01/2023]
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs) have great potential as therapeutic agents in stroke management, since they are easily accessible and can be rapidly expanded ex vivo for autologous transplantation. Increasing evidence suggests that bone marrow cells migrate throughout the brain and differentiate into neurons and glial cells. Both non-human and human MSCs have been used to treat stroke in murine models with satisfactory results. Several factors, such as transdifferentiation, induction of neurogenesis and angiogenesis, neuroprotection, and activation of endogenous neurorestorative processes, contribute to the benefits of MSCs in the ischemic brain. Many variables, including types of MSCs, cell dose, timing of treatment, route of cell delivery, and characteristics of stroke patients, influence the efficacy of MSC treatment of stroke. Although the first trials of autologous MSC therapy in stroke patients showed promising results, the optimal approach for different clinical settings has yet to be determined. The fundamental properties of MSCs and their potential short-term and long-term toxicities also need to be determined before moving forward to use of these cells in clinical practice.
Collapse
Affiliation(s)
- Permphan Dharmasaroja
- Department of Anatomy and Center for Neuroscience, Faculty of Science, Mahidol University, Rama VI Road, Rajthevi, Bangkok 10400, Thailand.
| |
Collapse
|
283
|
Sauerzweig S, Baldauf K, Braun H, Reymann KG. Time-dependent segmentation of BrdU-signal leads to late detection problems in studies using BrdU as cell label or proliferation marker. J Neurosci Methods 2008; 177:149-59. [PMID: 19007815 DOI: 10.1016/j.jneumeth.2008.10.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2008] [Revised: 09/22/2008] [Accepted: 10/06/2008] [Indexed: 01/05/2023]
Abstract
Bromodeoxyuridine incorporates into DNA during mitosis. A long-term stability of the incorporated BrdU is important for the recovery of BrdU-labeled cells. For testing the stability of BrdU incorporation into DNA we pulse-labeled mesenchymal stem cells with BrdU and observed these cells in vitro over 4 weeks. During this time the BrdU-signal was permanently decreasing. Starting with cells containing evenly stained BrdU-nuclei, so-called filled cells, already 3 days after BrdU removal we detected cells containing so-called segmented and punctated BrdU-signals. The number of those labeled cells continuously increased over time. Interestingly, the loss of BrdU in the nucleus was accompanied by an increasing labeling of the cytosol. Further, we injected BrdU intraperitoneally into rats after ischemia and detected BrdU-positive cells in the hippocampus 3 and 23 days after the last BrdU injection. While after 3 days most of the BrdU-positive cells in the hippocampus displayed a filled BrdU-signal, 23 days after BrdU removal an increased number of segmented and punctated BrdU-positive nuclei was detected. The gradual degradation of the BrdU-signal was not caused by cell death. The consequence of this BrdU degradation would be an underestimation of cell proliferation and an overestimation of cell death of newly generated cells.
Collapse
Affiliation(s)
- Steven Sauerzweig
- Leibniz Institute for Neurobiology (IfN), Brenneckestr. 6, 39118 Magdeburg, Germany.
| | | | | | | |
Collapse
|
284
|
Fu X, He Y, Xie C, Liu W. Bone marrow mesenchymal stem cell transplantation improves ovarian function and structure in rats with chemotherapy-induced ovarian damage. Cytotherapy 2008; 10:353-63. [PMID: 18574768 DOI: 10.1080/14653240802035926] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Many investigations have reported that mesenchymal stem cell (MSC) transplantation can ameliorate the structure and function of injured tissues. The purpose of this study was to explore the therapeutic potency of MSC transplantation for chemotherapy-induced ovarian damage. METHODS MSC were isolated and cultured in vitro. The cytokines, including vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF) and insulin-like growth factor-1 (IGF-1), were detected in the MSC cultures using enzyme-linked immunosorbent assay (ELISA). Phosphoramide mustard (PM) was added to the media of granulosa cells (GC) cultured alone or co-cultured with MSC. GC apoptosis was assayed by Annexin-V and DNA fragmentation analysis. Chemotherapy-induced ovarian damage was induced in rats by intraperitoneal injection of cyclophosphamide (CTX). After the injection, MSC labeled with green fluorescent protein (GFP) were transplanted directly into bilateral ovaries. The rats were killed at 2, 4, 6 and 8 weeks after transplantation. Ovarian function was evaluated by estrous cycle changes and sexual hormone levels. The follicle number was counted, and GC apoptosis was analyzed by TUNEL. The expressions of Bcl-2 and Bax proteins were detected by Western blotting. RESULTS MSC released VEGF, HGF and IGF-1 in vitro. The GC apoptosis was diminished by co-culture with MSC, which also resulted in increased Bcl-2 expression. The ovarian function of the rats exposed to CTX injection was improved after MSC transplantation. MSC reduced apoptosis of GC and induced up-regulation of Bcl-2 in vivo. DISCUSSION MSC transplantation can improve ovarian function and structure damaged by chemotherapy. The paracrine mediators secreted by MSC might be involved in the repair of damaged ovaries.
Collapse
Affiliation(s)
- X Fu
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | | | | | | |
Collapse
|
285
|
Iwanami J, Mogi M, Li JM, Tsukuda K, Min LJ, Sakata A, Fujita T, Iwai M, Horiuchi M. Deletion of Angiotensin II Type 2 Receptor Attenuates Protective Effects of Bone Marrow Stromal Cell Treatment on Ischemia–Reperfusion Brain Injury in Mice. Stroke 2008; 39:2554-9. [DOI: 10.1161/strokeaha.107.513275] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Jun Iwanami
- From the Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Masaki Mogi
- From the Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Jian-Mei Li
- From the Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Kana Tsukuda
- From the Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Li-Juan Min
- From the Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Akiko Sakata
- From the Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Teppei Fujita
- From the Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Masaru Iwai
- From the Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Masatsugu Horiuchi
- From the Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| |
Collapse
|
286
|
Li JS. Effects of rhubarb aglycone on matrix metalloproteinase in cerebral ischemic tissue in rats with bone marrow mesenchymal stem cell transplantation. JOURNAL OF CHINESE INTEGRATIVE MEDICINE 2008; 6:810-816. [DOI: 10.3736/jcim20080808] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
287
|
Myocyte enhancer factor 2C as a neurogenic and antiapoptotic transcription factor in murine embryonic stem cells. J Neurosci 2008; 28:6557-68. [PMID: 18579729 DOI: 10.1523/jneurosci.0134-08.2008] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cell-based therapies require a reliable source of cells that can be easily grown, undergo directed differentiation, and remain viable after transplantation. Here, we generated stably transformed murine ES (embryonic stem) cells that express a constitutively active form of myocyte enhancer factor 2C (MEF2CA). MEF2C has been implicated as a calcium-dependent transcription factor that enhances survival and affects synapse formation of neurons as well as differentiation of cardiomyocytes. We now report that expression of MEF2CA, both in vitro and in vivo, under regulation of the nestin enhancer effectively produces "neuronal" progenitor cells that differentiate into a virtually pure population of neurons. Histological, electrophysiological, and behavioral analyses demonstrate that MEF2C-directed neuronal progenitor cells transplanted into a mouse model of cerebral ischemia can successfully differentiate into functioning neurons and ameliorate stroke-induced behavioral deficits.
Collapse
|
288
|
Abstract
Cell-based therapy for stroke represents a third wave of therapeutics for stroke and one focused on restorative processes with a longer time window of opportunity than neuroprotective therapies. An early time window, within the first week after stroke, is an opportunity for intravenously delivered bone marrow and perinatally derived cells that can home to areas of tissue injury and target brain remodeling. Allogeneic cells will likely be the most scalable and commercially viable product. Later time windows, months after stroke, may be opportunities for intracerebral transplantation of neuronally differentiated cell types. An integrated approach of cell-based therapy with early-phase clinical trials and continued preclinical work with focus on mechanisms of action is needed.
Collapse
Affiliation(s)
- David C Hess
- Department of Neurology, Medical College of Georgia, Augusta, GA 30912, USA.
| | | |
Collapse
|
289
|
Intracerebral transplantation of human adipose tissue stromal cells after middle cerebral artery occlusion in rats. J Clin Neurosci 2008; 15:907-12. [DOI: 10.1016/j.jocn.2007.03.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2007] [Accepted: 03/20/2007] [Indexed: 01/01/2023]
|
290
|
Zhang RL, Zhang C, Zhang L, Roberts C, Lu M, Kapke A, Cui Y, Ninomiya M, Nagafuji T, Albala B, Zhang ZG, Chopp M. Synergistic effect of an endothelin type A receptor antagonist, S-0139, with rtPA on the neuroprotection after embolic stroke. Stroke 2008; 39:2830-6. [PMID: 18669895 DOI: 10.1161/strokeaha.108.515684] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Using a model of embolic stroke, the present study tested the hypothesis that blockage of endothelin-1 with S-0139, a specific endothelin type A receptor (ET(A)) antagonist, enhances the neuroprotective effect of recombinant tissue plasminogen activator (rtPA) by suppressing molecules that mediate thrombosis and blood brain barrier (BBB) disruption induced by ischemia and rtPA. METHODS Rats (n=104) subjected to embolic middle cerebral artery (MCA) occlusion were randomly divided into 1 of 4 infusion groups with 26 rats per group: (1) the control group in which rats were administered saline, (2) the monotherapy rtPA group in which rtPA was intravenously administered at a dose of 10 mg/kg 4 hours after MCA occlusion, (3) the monotherapy S-0139 group in which S-0139 was intravenously given 2 hours after MCA occlusion, and (4) the combination of rtPA +S-0139 group in which S-0139 and rtPA were given 2 and 4 hours after MCA occlusion, respectively. Measurements of infarct volume and parenchymal hemorrhage, behavioral outcome, and immunostaining were performed on rats euthanized 1 and 7 days after stroke. RESULTS The combination therapy of S-0139 and rtPA significantly (P<0.01) reduced infarct volume (24.8+/-0.9% versus 33.8+/-1.5% in control) and hemorrhagic area (7.1+/-6.1 microm(2) versus 36.5+/-19.2 microm(2) in control) and improved functional recovery compared with control saline-treated animals. Immunostaining analysis revealed that the combination therapy had the synergistically suppressed ischemia- and rtPA-induced ICAM-1, protease-activated receptor 1 (PAR-1), as well as accumulation of platelets in cerebral microvessels. Furthermore, the combination treatment synergistically reduced loss of laminin, ZO1, and occludin in cerebral vessels. CONCLUSIONS These data suggest that S-0139 provides the neuroprotection by suppressing ischemia- and rtPA-triggered molecules that evoke thrombosis and BBB disruption.
Collapse
Affiliation(s)
- Rui Lan Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan 48202, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
291
|
Zhang J, Li Y, Zheng X, Gao Q, Liu Z, Qu R, Borneman J, Elias SB, Chopp M. Bone marrow stromal cells protect oligodendrocytes from oxygen-glucose deprivation injury. J Neurosci Res 2008; 86:1501-10. [PMID: 18214988 DOI: 10.1002/jnr.21617] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Oligodendrocyte (OLG) damage leads to demyelination, which is frequently observed in ischemic cerebrovascular diseases. In this study, we investigated the effect of bone marrow stromal cells (BMSCs) on OLGs subjected to oxygen-glucose deprivation (OGD). N20.1 cells (mouse OLG cell line) were transferred into an anaerobic chamber for 3 hr in glucose-free and serum-free medium. After OGD incubation, OLG cultures were divided into the following groups: 1) OGD alone, 2) OLG cocultured with BMSCs, 3) treatment with the phosphoinostide 3-kinase (PI3k) inhibitor LY294002, 4) LY294002-treated OLGs with BMSC cocultured, and 5) anti-p75 antibody-treated OLGs. After an additional 3 hr of reoxygenation incubation, OLG viability and apoptosis were measured. The mRNA expression in the BMSCs and OLGs was analyzed using quantitative real-time PCR (RT-PCR). Serine/threonine-specific protein kinase (Akt), phosphorylated Akt (p-Akt), p75, and caspase 3 protein expressions in OLGs were measured by Western blot. Our results suggest that BMSCs produce growth factors, activate the Akt pathway, and increase the survival of OLGs. BMSCs also reduce p75 and caspase 3 expressions in the OGD-OLGs, which leads to decreased OLG apoptosis. BMSCs participate in OLG protection that may occur with promoting growth factors/PI3K/Akt and inhibiting the p75/caspase pathways. Our study provides insight into white matter damage and the therapeutic benefits of BMSC-based remyelinating therapy after stroke and demyelinating diseases.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan 48202, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
292
|
Cancer gene therapy using mesenchymal stem cells expressing interferon-beta in a mouse prostate cancer lung metastasis model. Gene Ther 2008; 15:1446-53. [PMID: 18596829 DOI: 10.1038/gt.2008.101] [Citation(s) in RCA: 146] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Cell-based therapy for cancer is a promising new field. Among cell types that can be used for this purpose, mesenchymal stem cells (MSCs) appear to hold great advantage for reasons including easier propagation in culture, possible genetic modification to express therapeutic proteins and preferential homing to sites of cancer growth upon in vivo transfer. The present study evaluated the potential of genetically modified MSC, constitutively expressing interferon (IFN)-beta, in an immunocompetent mouse model of prostate cancer lung metastasis. A recombinant adeno-associated virus (rAAV) encoding mouse IFN-beta was constructed and initially tested in vitro for high-level expression and bioactivity of the transgenic protein. MSCs were transduced by the rAAV-IFN-beta or green fluorescent protein ex vivo and used as cellular vehicles to target lung metastasis of TRAMP-C2 prostate cancer cells in a therapy model. Cohorts of mice were killed on days 30 and 75 to determine the effect of therapy by measurement of tumor volume, histology, immunohistochemistry, enzyme-linked immunosorbent assay and flow cytometry. Results indicated a significant reduction in tumor volume in lungs following IFN-beta-expressing MSC therapy. Immunohistochemistry of the lung demonstrated increased tumor cell apoptosis and decreased tumor cell proliferation and blood vessel counts. A significant increase in the natural kill cell activity was observed following IFN-beta therapy correlating the antitumor effect. Systemic level of IFN-beta was not significantly elevated from this targeted cell therapy. These data demonstrate the potential of MSC-based IFN-beta therapy for prostate cancer lung metastasis.
Collapse
|
293
|
Seyfried DM, Han Y, Yang D, Ding J, Savant-Bhonsale S, Shukairy MS, Chopp M. Mannitol enhances delivery of marrow stromal cells to the brain after experimental intracerebral hemorrhage. Brain Res 2008; 1224:12-9. [PMID: 18573239 DOI: 10.1016/j.brainres.2008.05.080] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2007] [Revised: 05/22/2008] [Accepted: 05/23/2008] [Indexed: 01/27/2023]
Abstract
Previous studies show that intravascular injection of human bone marrow stromal cells (hBMSCs) significantly improves neurological functional recovery in a rat model of intracerebral hemorrhage (ICH). In the present study, we tested the hypothesis that mannitol improves the efficiency of intraarterial MSC delivery (i.e., fewer injected cells required for therapeutic efficacy) after ICH. There were four post-ICH groups (N=9): group 1, negative control with only intraarterial injection of 1 million human fibroblasts in phosphate-buffered saline (PBS); group 2, intravenous injection of mannitol alone in PBS (1.5 g/kg); group 3, intraarterial injection of 1 million hBMSCs alone in PBS; and group 4, intravenous injection of mannitol (1.5 g/kg) in PBS followed by intraarterial injection of 1 million hBMSCs in PBS. Group 4 exhibited significantly improved neurological functional outcome as assessed by neurological severity score (NSS) and corner test scores. Immunohistochemical staining of group 4 suggested increased synaptogenesis, proliferating immature neurons, and neuronal migration. The number of hBMSCs recruited to the injured region increased strikingly in group 4. Tissue loss was notably reduced in group 4. In summary, the beneficial effects of intraarterial infusion of MSCs are amplified with intravenous injection of mannitol. Preadministration of mannitol significantly increases the number of hBMSCs located in the ICH region, improves histochemical parameters of neural regeneration, and reduces the anatomical and pathological consequences of ICH.
Collapse
Affiliation(s)
- Donald M Seyfried
- Department of Neurosurgery, Henry Ford Health System, 2799 W Grand Boulevard, Detroit, MI 48202, USA.
| | | | | | | | | | | | | |
Collapse
|
294
|
Hedgehog Serves as a Mitogen and Survival Factor During Embryonic Stem Cell Neurogenesis. Stem Cells 2008; 26:1097-108. [DOI: 10.1634/stemcells.2007-0684] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
295
|
Walczak P, Zhang J, Gilad AA, Kedziorek DA, Ruiz-Cabello J, Young RG, Pittenger MF, van Zijl PCM, Huang J, Bulte JWM. Dual-modality monitoring of targeted intraarterial delivery of mesenchymal stem cells after transient ischemia. Stroke 2008; 39:1569-74. [PMID: 18323495 DOI: 10.1161/strokeaha.107.502047] [Citation(s) in RCA: 295] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND PURPOSE In animal models of stroke, functional improvement has been obtained after stem cell transplantation. Successful therapy depends largely on achieving a robust and targeted cell engraftment, with intraarterial (IA) injection being a potentially attractive route of administration. We assessed the suitability of laser Doppler flow (LDF) signal measurements and magnetic resonance (MR) imaging for noninvasive dual monitoring of targeted IA cell delivery. METHODS Transient cerebral ischemia was induced in adult Wistar rats (n=25) followed by IA or intravenous (IV) injection of mesenchymal stem cells (MSCs) labeled with superparamagnetic iron oxide. Cell infusion was monitored in real time with transcranial laser Doppler flowmetry while cellular delivery was assessed with MRI in vivo (4.7 T) and ex vivo (9.4 T). RESULTS Successful delivery of magnetically labeled MSCs could be readily visualized with MRI after IA but not IV injection. IA stem cell injection during acute stroke resulted in a high variability of cerebral engraftment. The amount of LDF reduction during cell infusion (up to 80%) was found to correlate well with the degree of intracerebral engraftment, with low LDF values being associated with significant morbidity. CONCLUSIONS High cerebral engraftment rates are associated with impeded cerebral blood flow. Noninvasive dual-modality imaging enables monitoring of targeted cell delivery, and through interactive adjustment may improve the safety and efficacy of stem cell therapy.
Collapse
Affiliation(s)
- Piotr Walczak
- Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 217 Traylor, 720 Rutland Ave, Baltimore, MD 21205-2195, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
296
|
Local stabilization of microtubule assembly improves recovery of facial nerve function after repair. Exp Neurol 2008; 209:131-44. [DOI: 10.1016/j.expneurol.2007.09.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2007] [Revised: 09/03/2007] [Accepted: 09/10/2007] [Indexed: 11/23/2022]
|
297
|
Intravenous administration of bone marrow stromal cells increases survivin and Bcl-2 protein expression and improves sensorimotor function following ischemia in rats. Neurosci Lett 2008; 430:109-14. [DOI: 10.1016/j.neulet.2007.10.046] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2007] [Revised: 10/08/2007] [Accepted: 10/25/2007] [Indexed: 11/23/2022]
|
298
|
Chopp M, Li Y. Transplantation of Bone Marrow Stromal Cells for Treatment of Central Nervous System Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 585:49-64. [PMID: 17120776 DOI: 10.1007/978-0-387-34133-0_4] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Michael Chopp
- Department of Neurology, Henry Ford Health Sciences Center, Detroit, Michigan, USA
| | | |
Collapse
|
299
|
SDF-1alpha/CXCR4-mediated migration of systemically transplanted bone marrow stromal cells towards ischemic brain lesion in a rat model. Brain Res 2007; 1195:104-12. [PMID: 18206136 DOI: 10.1016/j.brainres.2007.11.068] [Citation(s) in RCA: 204] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Revised: 11/28/2007] [Accepted: 11/29/2007] [Indexed: 12/21/2022]
Abstract
Transplantation of bone marrow-derived mesenchymal stem cells (BMSCs) can promote functional recovery of brain after stroke with the mechanism regulating the BMSCs migration to ischemic penumbra poorly understood. Interaction between stromal cell-derived factor-1alpha (SDF-1alpha) and its cognate receptor CXCR4 is crucial for homing and migration of multiple stem cell types. Their potential role in mediating BMSC migration in ischemic brain has not been demonstrated. In this study, ischemic brain lesion model was created in rats by permanent middle cerebral artery occlusion and green fluorescent protein (GFP)-labeled BMSCs were intravenously injected. Immunohistochemical staining showed that BMSCs were able to enter the route from olfactory areas to cortex of the rat brain. Significant recovery of modified Neurological Severity Score was observed at days 14 and 28. Interestingly, the SDF-1alpha mRNA and protein were predominantly localized in the ischemic penumbral, peaked by 3-7 days and retained at least 14 days post-transplantation. On the other hand, the CXCR4 expression by BMSCs was elevated under hypoxia. The pre-treatment with the CXCR4-specific antagonist AMD3100 significantly prevented the migration of BMSCs to the injured brain. Taken together, these observations indicate that systemically administered BMSCs can migrate to the ischemic lesion of brain along with the olfactory-thalamus and hippocampus-cortex route. The interaction of locally produced SDF-1alpha and CXCR4 expressed on the BMSC surface plays an important role in the migration of transplanted cells, suggesting that it might be a potential approach to modulate the expression of the two molecules in order to further facilitate the therapeutic effects using BMSCs.
Collapse
|
300
|
Transplantation of NGF-gene-modified bone marrow stromal cells into a rat model of Alzheimer' disease. J Mol Neurosci 2007; 34:157-63. [PMID: 18074108 DOI: 10.1007/s12031-007-9022-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Accepted: 11/02/2007] [Indexed: 12/26/2022]
Abstract
It is well known that bone marrow stromal cells (BMSC) grafted into the hippocampus of the rat model of Alzheimer's disease (AD) could survive and differentiate into cholinergic neurons as well as contribute towards functional restoration. The present study evaluated the effects of BMSC as a seed cell modified by nerve growth factor (NGF) gene into the hippocampus of AD rats. The beta-amyloid protein was injected bilaterally into the rat hippocampus to reproduce the AD model. After the human total RNA was extracted, the NGF gene was amplified by reverse transcription-polymerase chain reaction, then cloned into the pcDNA3. BMSC derived from a green fluorescence protein transgenic mouse were isolated, cultured, identified, and transfected by the NGF recombinant. The NGF-gene-modified BMSC were then transplanted into the hippocampus of AD rats. The results showed that implanted BMSC survived, migrated and expressed NGF as well as differentiated into ChAT-positive neurons. A significant improvement in learning and memory in AD rats was also seen in NGF-gene-modified BMSC group, when compared with the BMSC group. The present findings suggested that BMSC provided an effective carrier for delivery of NGF into AD rats, and the administration of NGF-gene-modified BMSC may be considered as a potential strategy for the development of effective therapies for the treatment of AD.
Collapse
|