151
|
Zhang F, Tsai S, Kato K, Yamanouchi D, Wang C, Rafii S, Liu B, Kent KC. Transforming growth factor-beta promotes recruitment of bone marrow cells and bone marrow-derived mesenchymal stem cells through stimulation of MCP-1 production in vascular smooth muscle cells. J Biol Chem 2009; 284:17564-74. [PMID: 19406748 PMCID: PMC2719395 DOI: 10.1074/jbc.m109.013987] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2009] [Revised: 04/28/2009] [Indexed: 12/22/2022] Open
Abstract
Bone marrow-derived progenitor cells have recently been shown to be involved in the development of intimal hyperplasia after vascular injury. Transforming growth factor-beta (TGF-beta) has profound stimulatory effects on intimal hyperplasia, but it is unknown whether these effects involve progenitor cell recruitment. In this study we found that although TGF-beta had no direct effect on progenitor cell recruitment, conditioned media derived from vascular smooth muscle cells (VSMC) stimulated with TGF-beta induced migration of both total bone marrow (BM) cells and BM-mesenchymal stem cells (MSC) and also induced MSC differentiation into smooth muscle like cells. Furthermore, overexpression of the signaling molecule Smad3 in VSMC via adenovirus-mediated gene transfer (AdSmad3) enhanced the TGF-beta's chemotactic effect. Microarray analysis of VSMC stimulated by TGF-beta/AdSmad3 revealed monocyte chemoattractant protein-1 (MCP-1) as a likely factor responsible for progenitor cell recruitment. We then demonstrated that TGF-beta through Smad3 phosphorylation induced a robust expression of MCP-1 in VSMC. Recombinant MCP-1 mimicked the stimulatory effect of conditioned media on BM and MSC migration. In the rat carotid injury model, Smad3 overexpression significantly increased MCP-1 expression after vascular injury, consistent with our in vitro results. Interestingly, TGF-beta/Smad3-induced MCP-1 was completely blocked by both Ro-32-0432 and rotterlin, suggesting protein kinase C-delta (PKCdelta) may play a role in TGF-beta/Smad3-induced MCP-1 expression. In summary, our data demonstrate that TGF-beta, through Smad3 and PKCdelta, stimulates VSMC production of MCP-1, which is a chemoattractant for bone marrow-derived cells, specifically MSC. Manipulation of this signaling system may provide a novel approach to inhibition of intimal hyperplasia.
Collapse
MESH Headings
- Angioplasty, Balloon
- Animals
- Aorta, Thoracic/cytology
- Aorta, Thoracic/metabolism
- Biomarkers/metabolism
- Blotting, Western
- Bone Marrow Cells/metabolism
- Cell Communication
- Cell Differentiation
- Cell Movement
- Cells, Cultured
- Chemokine CCL2/antagonists & inhibitors
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Chemotaxis
- Gene Expression Profiling
- Hyperplasia/metabolism
- Hyperplasia/pathology
- Immunoenzyme Techniques
- Male
- Mesenchymal Stem Cells/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Oligonucleotide Array Sequence Analysis
- Protein Kinase C-delta/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Smad3 Protein/antagonists & inhibitors
- Smad3 Protein/genetics
- Smad3 Protein/metabolism
- Stem Cells
- Transforming Growth Factor beta/pharmacology
- Tunica Intima/metabolism
- Tunica Intima/pathology
Collapse
Affiliation(s)
- Fan Zhang
- From the Department of Surgery, University of Wisconsin School of Medicine, Madison, Wisconsin, 53705
| | - Shirling Tsai
- Department of Surgery, Division of Vascular Surgery, Weill Medical College of Cornell University, New York, New York 10065, and
| | - Kaori Kato
- From the Department of Surgery, University of Wisconsin School of Medicine, Madison, Wisconsin, 53705
| | - Dai Yamanouchi
- From the Department of Surgery, University of Wisconsin School of Medicine, Madison, Wisconsin, 53705
| | - Chunjie Wang
- Department of Surgery, Division of Vascular Surgery, Weill Medical College of Cornell University, New York, New York 10065, and
| | - Shahin Rafii
- Howard Hughes Medical Institute, Weill Cornell Medical College, New York, New York 10065
| | - Bo Liu
- From the Department of Surgery, University of Wisconsin School of Medicine, Madison, Wisconsin, 53705
| | - K. Craig Kent
- From the Department of Surgery, University of Wisconsin School of Medicine, Madison, Wisconsin, 53705
| |
Collapse
|
152
|
Li L, Bai X, Gong X, Liu H, Chen L, Guan W, Ma Y. Differentiation potential of bone marrow mesenchymal stem cells in duck. J Genet Genomics 2009; 36:133-40. [PMID: 19302969 DOI: 10.1016/s1673-8527(08)60100-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2008] [Revised: 01/21/2009] [Accepted: 01/23/2009] [Indexed: 11/18/2022]
Abstract
The bone marrow mesenchymal stem cells (MSCs) are multipotent stem cells which can differentiate into mesenchymal cells in vitro. In this study, MSCs in duck were isolated from bone marrow by density gradient centrifuge separation, purified and expanded in the medium. The primary MSCs were expanded for passages. The different-passage MSCs were induced to differentiate into osteoblasts and neuron-like cells. Karyotype analysis indicated that MSCs kept diploid condition and the hereditary feature was stable. The different-passage MSCs expressed CD44, ICAM- and SSEA-4, but not CD34, CD45 and SSEA-when detected by immunofluorescence staining. There was no significant difference among the positive rates of passages 2, 6 and 8 (P > 0.05), but a significant difference existed among those of passages 2, 6, 8 and 11 (P < 0.05). After the osteogenic inducement was added, the induced different-passage MSCs expressed high-level alkaline phosphatase (ALP), and are positive for tetracycline staining, Alizarin Red staining and Von Kossa staining. After the neural inducement was added, about 70% cells exhibited typical neuron-like phenotype, the induced different-passage MSCs expressed Nestin, neuron-specific enolase (NSE) and glial fibrillary acidic protein (GFAP) when detected by immunofluorescence staining. There was no significant difference among the positive rates of passages 3, 4 and 6 (P>0.05), but a significant difference existed among those of passages 3, 4, 6 and 8 (P<0.05). These results suggest that MSCs in duck were capable of differentiating into osteoblasts and neuron-like cells in vitro.
Collapse
Affiliation(s)
- Linfeng Li
- Institute of Animal Science, Chinese Academy of Agricultural Science, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
153
|
Eslaminejad MB, Nadri S. Murine mesenchymal stem cell isolated and expanded in low and high density culture system: surface antigen expression and osteogenic culture mineralization. In Vitro Cell Dev Biol Anim 2009; 45:451-9. [DOI: 10.1007/s11626-009-9198-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2008] [Accepted: 03/12/2009] [Indexed: 11/29/2022]
|
154
|
Abstract
Mesenchymal stem cells (MSC) are a type of multipotent progenitor cell, originally isolated from the bone marrow. In addition to multilineage differentiation and participation in the hematopoietic niche, they exert powerful immunomodulatory effects, which include inhibition of proliferation and function of T cells, B cells, and natural killer cells. These unique properties make MSC of great interest for clinical applications in tissue engineering and immunosuppression. Underlying the MSC-mediated immunomodulatory mechanisms is a nonspecific antiproliferative effect, which is the consequence of cyclin D2 inhibition. Of special interest are the molecular mechanisms, by which MSC influence their target cells. Several studies have been conducted in this field, and the current data suggest roles for indoleamine 2,3-dioxygenase, prostaglandin E2, nitric oxide, histocompatibility locus antigen-G, insulin-like growth factor-binding proteins, and tolerogenic antigen-presenting cells. Understanding these mechanisms is crucial for future use of MSC in research and clinical applications.
Collapse
Affiliation(s)
- Georg Siegel
- Institute of Clinical and Experimental Transfusion Medicine (IKET), Eberhard Karls University Tübingen, Germany
| | | | | |
Collapse
|
155
|
Abstract
The study of MSC trafficking is clinically relevant for minimally invasive cell therapy to promote regeneration of damaged tissue, to treat inflammation, and to promote angiogenesis. However, these studies are complicated by the diverse methods used to culture, characterize, and deliver MSCs and by the variety of methods used to assess homing events. This review provides a critical analysis of the methods used to track homing of exogenously infused MSCs and discusses strategies for enhancing their trafficking to particular tissues.
Collapse
Affiliation(s)
- Jeffrey M Karp
- Harvard-MIT Division of Health Science and Technology, 77 Massachusetts Avenue, E25-519, Cambridge, MA 02139, USA.
| | | |
Collapse
|
156
|
Wang H, Ge W, Arp J, Zassoko R, Liu W, Ichim TE, Jiang J, Jevnikar AM, Garcia B. Free Bone Graft Attenuates Acute Rejection and in Combination with Cyclosporin A Leads to Indefinite Cardiac Allograft Survival. THE JOURNAL OF IMMUNOLOGY 2009; 182:5970-81. [DOI: 10.4049/jimmunol.0801037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
157
|
[Regenerative potential of human adult precursor cells: cell therapy--an option for treating cartilage defects?]. Z Rheumatol 2009; 68:234-8. [PMID: 19384550 DOI: 10.1007/s00393-008-0396-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Cell-based therapeutical approaches are already in clinical use and are attracting growing interest for the treatment of joint defects. Mesenchymal stem and precursor cells (MSC) cover a wide range of properties that are useful for the regeneration process of bone and cartilage defects. The following article is an overview of the regenerative potential of MSC and discusses how the properties of these cells can be used for the development of new strategies in regenerative medicine.
Collapse
|
158
|
Xu YL, Gao YH, Liu Z, Tan KB, Hua X, Fang ZQ, Wang YL, Wang YJ, Xia HM, Zhuo ZX. Myocardium-targeted transplantation of mesenchymal stem cells by diagnostic ultrasound-mediated microbubble destruction improves cardiac function in myocardial infarction of New Zealand rabbits. Int J Cardiol 2009; 138:182-95. [PMID: 19383567 DOI: 10.1016/j.ijcard.2009.03.071] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2008] [Revised: 12/31/2008] [Accepted: 03/10/2009] [Indexed: 11/17/2022]
Abstract
BACKGROUND Therapeutic ultrasound-mediated microbubble destruction has been applied in the targeted delivery of genes, drugs and stem cells. We intended to study whether diagnostic US irradiating lipid-coated microbubble destruction combined with bone-marrow derived MSC infusion could enable the targeted delivery of MSCs into the myocardium and improve cardiac function of the myocardial infarction of New Zealand rabbits. METHODS Diagnostic ultrasound was applied to the anterior chest for 10 min after intravenous injection of lipid-coated microbubble followed by infusion of BM-MSCs. Echocardiography, histological examination, and western blotting were performed 4 weeks after cell transplantation. RESULTS The cardiac function (assessed by fractional shortening and ejection fraction) was markedly improved by US+Microbubble+MSC treatment. The number of capillaries stained by HE in US+Microbubble+MSC group (47+/-23) was much greater than that of the MSCs infusion group (26+/-7), US+Microbubble group(22+/-5) and PBS infusion group (19+/-10), P<0.01. US+Microbubble stimulation induced the expression of adhesion molecule (VCAM-1) in capillaries and enhanced the myocardial permeability of microvessels. US+Microbubble-mediated supply of MSCs increased the level of VEGF in ischemic myocardium. Area of cardiac fibrosis in the US+Microbubble+MSC group was significantly decreased by 25.6%,40.1% and 46.8% when compared with MSC infusion group, US+Microbubble group and PBS infusion group, respectively. CONCLUSIONS This non-invasive cell delivery system may be useful as a novel and efficient approach for angiogenic cell therapy to the infarcted myocardium.
Collapse
Affiliation(s)
- Ya-Li Xu
- Department of Ultrasound, Second affiliated Hospital, Third Military Medical University, Chongqing 400037, PR China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
159
|
Wolf D, Reinhard A, Seckinger A, Katus HA, Kuecherer H, Hansen A. Dose-dependent effects of intravenous allogeneic mesenchymal stem cells in the infarcted porcine heart. Stem Cells Dev 2009; 18:321-9. [PMID: 18435573 DOI: 10.1089/scd.2008.0019] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Intravenous delivery of mesenchymal stem cells (MSCs) preserves myocardial function after infarction. This dose-escalating study was performed to examine pathologic remodeling and scar formation in a pig model of permanent coronary occlusion without restoration of reperfusion. MSCs labeled with fluorescent dye 48 h or saline (negative control, n = 8) were given intravenously 48 h post proximal left anterior descending artery occlusion. Animals received either autologous or allogeneic MSCs in doses from 1 x 10(3) up to 1 x 10(6) per kg bodyweight from an unrelated donor pig. Infarct size and myocardial function were assessed after 1 month. Morphologic analysis revealed that labeled autologous MSCs migrated in the peri-infarct region resulting in smaller infarct size (19 +/- 7% vs. 32 +/- 7%, p < 0.008) and higher fractional area shortening (33 +/- 7% vs. 21 +/- 3%, p < 0.001). Similarly, allogeneic MSCs had dose-dependent beneficial effects on cardiac function, statistically significant at 1 x 10(5) and 1 x 10(6) cells per kg bodyweight. Autologous as well as allogeneic MSCs specifically "home" to the heart after systemic delivery, leading to limited myocardial infarct size and improved functional outcome, even without coronary reperfusion. Therefore, intravenously administration of MSCs is an attractive minimal-invasive approach for cardiac tissue repair.
Collapse
Affiliation(s)
- David Wolf
- Department of Cardiology, University of Heidelberg, Koesching, Germany
| | | | | | | | | | | |
Collapse
|
160
|
García-Castro J, Trigueros C, Madrenas J, Pérez-Simón JA, Rodriguez R, Menendez P. Mesenchymal stem cells and their use as cell replacement therapy and disease modelling tool. J Cell Mol Med 2009; 12:2552-65. [PMID: 19210755 PMCID: PMC3828873 DOI: 10.1111/j.1582-4934.2008.00516.x] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) from adult somatic tissues may differentiate in vitro and in vivo into multiple mesodermal tissues including bone, cartilage, adipose tissue, tendon, ligament or even muscle. MSCs preferentially home to damaged tissues where they exert their therapeutic potential. A striking feature of the MSCs is their low inherent immunogenicity as they induce little, if any, proliferation of allogeneic lymphocytes and antigen-presenting cells. Instead, MSCs appear to be immunosuppressive in vitro. Their multi-lineage differentiation potential coupled to their immuno-privileged properties is being exploited worldwide for both autologous and allo-geneic cell replacement strategies. Here, we introduce the readers to the biology of MSCs and the mechanisms underlying immune tolerance. We then outline potential cell replacement strategies and clinical applications based on the MSCs immunological properties. Ongoing clinical trials for graft-versus-host-disease, haematopoietic recovery after co-transplantation of MSCs along with haematopoietic stem cells and tissue repair are discussed. Finally, we review the emerging area based on the use of MSCs as a target cell subset for either spontaneous or induced neoplastic transformation and, for modelling non-haematological mesenchymal cancers such as sarcomas.
Collapse
Affiliation(s)
- J García-Castro
- Andalusian Stem Cell Bank (BACM), University of Granada, Granada, Spain
| | | | | | | | | | | |
Collapse
|
161
|
Stich S, Haag M, Häupl T, Sezer O, Notter M, Kaps C, Sittinger M, Ringe J. Gene expression profiling of human mesenchymal stem cells chemotactically induced with CXCL12. Cell Tissue Res 2009; 336:225-36. [PMID: 19296133 DOI: 10.1007/s00441-009-0768-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Accepted: 01/21/2009] [Indexed: 12/22/2022]
Abstract
In situ tissue engineering is a promising approach in regenerative medicine, with the possibility that adult stem or progenitor cells will be guided chemotactically to a tissue defect and subsequently differentiate into the surrounding tissue type. Mesenchymal stem cells (MSC) represent attractive candidate cells. Chemokines such as CXCL12 (SDF-1alpha) chemoattract MSC, but little is known about the molecular processes involved in the chemotaxis and migration of MSC. In this study, MSC recruitment by CXCL12 was investigated by genome-wide microarray analysis. The dose-dependent migration potential of bone-marrow-derived MSC toward CXCL12 was measured in an in vitro assay, with a maximum being recorded at a concentration of 1,000 nM CXCL12. Microarray analysis of MSC stimulated with CXCL12 and non-stimulated controls showed 30 differentially expressed genes (24 induced and six repressed). Pathway analysis revealed 11 differentially expressed genes involved in cellular movement and cytokine-cytokine receptor interaction, including those for migratory inducers such as the chemokines CXCL8 and CCL26, the leukocyte inhibitory factor, secretogranin II, and prostaglandin endoperoxide synthase 2. These results were confirmed by real-time polymerase chain reaction for selected genes. The obtained data provide further insights into the molecular mechanisms involved in chemotactic processes in cell migration and designate CXCL12 as a promising candidate for in situ recruitment in regenerative therapies.
Collapse
Affiliation(s)
- Stefan Stich
- Tissue Engineering Laboratory and Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology and Clinical Immunology, Charité-University Medicine Berlin, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
162
|
Wolf D, Reinhard A, Seckinger A, Gross L, Katus HA, Hansen A. Regenerative capacity of intravenous autologous, allogeneic and human mesenchymal stem cells in the infarcted pig myocardium-complicated by myocardial tumor formation. SCAND CARDIOVASC J 2009; 43:39-45. [PMID: 18609048 DOI: 10.1080/14017430802100280] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVES Intravenous delivery of mesenchymal stem cells (MSCs) is an attractive approach for regeneration of infarcted myocardium. However, its efficacy is not well-defined in large animals. METHODS Pigs (n =8) received intravenously autologous, allogeneic porcine or human MSCs (1 x 10(6) per kg bodyweight) labeled with fluorescent dye 48 hours post proximal LAD occlusion. Infarct size, histology and myocardial function were assessed 4 weeks later. RESULTS Labeled MSCs migrated in the peri-infarct region resulting in improved myocardial function. Infarct size was larger in the control group (32+/-7%) compared to autologous (19+/-7%, p =0.008), allogeneic (24+/-4%, p =0.01) and human MSCs (26+/-5%, p =0.03). Fractional area shortening significantly increased after 4 weeks in pigs receiving autologous MSCs (34+/-7%, p =0.001), allogeneic MSCs (28+/-2%, p =0.004) and human MSCs (24+/-5%, p =0.027), but was lower in the control group (23+/-3%, n.s.). However, substantial callus formation and a non-malignant cardiac "tumor" containing mesenchymal tissue was observed in one animal treated with human MSCs. CONCLUSIONS Intravenously administered MSCs prevent pathologic remodeling and scar formation but bare potential risks from inflammatory-related products.
Collapse
Affiliation(s)
- David Wolf
- Department of Cardiology, University of Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
163
|
Cellules souches mésenchymateuses et immunomodulation : vers de nouvelles stratégies immunosuppressives pour le traitement des maladies auto-immunes ? Rev Med Interne 2009; 30:287-99. [DOI: 10.1016/j.revmed.2008.08.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2008] [Revised: 08/27/2008] [Accepted: 08/29/2008] [Indexed: 12/29/2022]
|
164
|
Lee SW, Padmanabhan P, Ray P, Gambhir SS, Doyle T, Contag C, Goodman SB, Biswal S. Stem cell-mediated accelerated bone healing observed with in vivo molecular and small animal imaging technologies in a model of skeletal injury. J Orthop Res 2009; 27:295-302. [PMID: 18752273 PMCID: PMC4154812 DOI: 10.1002/jor.20736] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2007] [Accepted: 06/05/2008] [Indexed: 02/04/2023]
Abstract
Adult stem cells are promising therapeutic reagents for skeletal regeneration. We hope to validate by molecular imaging technologies the in vivo life cycle of adipose-derived multipotent cells (ADMCs) in an animal model of skeletal injury. Primary ADMCs were lentivirally transfected with a fusion reporter gene and injected intravenously into mice with bone injury or sham operation. Bioluminescence imaging (BLI), [(18)F]FHBG (9-(fluoro-hydroxy-methyl-butyl-guanine)-micro-PET, [(18)F]Fluoride ion micro-PET and micro-CT were performed to monitor stem cells and their effect. Bioluminescence microscopy and immunohistochemistry were done for histological confirmation. BLI showed ADMC's traffic from the lungs then to the injury site. BLI microscopy and immunohistochemistry confirmed the ADMCs in the bone defect. Micro-CT measurements showed increased bone healing in the cell-injected group compared to the noninjected group at postoperative day 7 (p < 0.05). Systemically administered ADMC's traffic to the site of skeletal injury and facilitate bone healing, as demonstrated by molecular and small animal imaging. Molecular imaging technologies can validate the usage of adult adipose tissue-derived multipotent cells to promote fracture healing. Imaging can in the future help establish therapeutic strategies including dosage and administration route.
Collapse
Affiliation(s)
- Sheen-Woo Lee
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, 300 Pasteur Drive S-062B, Stanford, California 94304
| | - Parasuraman Padmanabhan
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, 300 Pasteur Drive S-062B, Stanford, California 94304
| | - Pritha Ray
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, 300 Pasteur Drive S-062B, Stanford, California 94304
| | - Sanjiv Sam Gambhir
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, 300 Pasteur Drive S-062B, Stanford, California 94304
| | - Timothy Doyle
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, 300 Pasteur Drive S-062B, Stanford, California 94304
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Christopher Contag
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, 300 Pasteur Drive S-062B, Stanford, California 94304
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - Sandip Biswal
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, 300 Pasteur Drive S-062B, Stanford, California 94304
| |
Collapse
|
165
|
Dumas A, Brigitte M, Moreau MF, Chrétien F, Baslé MF, Chappard D. Bone mass and microarchitecture of irradiated and bone marrow-transplanted mice: influences of the donor strain. Osteoporos Int 2009; 20:435-43. [PMID: 18548305 DOI: 10.1007/s00198-008-0658-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Accepted: 04/24/2008] [Indexed: 10/22/2022]
Abstract
UNLABELLED Total body irradiation and bone marrow transplantation induced dramatic trabecular bone loss and cortical thickening in mice. Transplanted cells were engrafted in bone marrow, along trabeculae, and in periosteal and endosteal envelopes. None of the osteocytes were of donor origin. Bone microarchitecture of transplanted mice changed to tend toward the donor phenotype. INTRODUCTION Osteopenia and osteoporosis are complications of bone marrow transplants (BMT) attributed to related chemotherapy. However, the specific influence of total body irradiation (TBI) is unknown. METHODS We investigated the effects of TBI and BMT on bone mass and microarchitecture by micro-CT. Eighteen C57Bl/6 (B6) mice receiving lethal TBI had a BMT with marrow cells from green fluorescent protein--transgenic-C57Bl/6 (GFP) mice. Transplanted (T(GFP)B6), B6, and GFP mice were euthanized 1, 3, and 6 months after BMT or at a related age. RESULTS T(GFP)B6 presented a dramatic bone loss compared with B6 and did not restore their trabecular bone mass over time, despite a cortical thickening 6 months after BMT. Serum testosterone levels were not significantly reduced after BMT. During aging, GFP mice have less trabeculae, thicker cortices, but a narrower femoral shaft than B6 mice. From 3 months after BMT, cortical characteristics of T(GFP)B6 mice differed statistically from B6 mice and were identical to those of GFP mice. GFP(+) cells were located along trabecular surfaces and in periosteal and endosteal envelopes, but none of the osteocytes expressed GFP. CONCLUSION Our findings suggest that engrafted cells did not restore the irradiation-induced trabecular bone loss, but reconstituted a marrow microenvironment and bone remodeling similar to those of the donor. The effects of irradiation and graft on bone remodeling differed between cortical and trabecular bone.
Collapse
Affiliation(s)
- A Dumas
- INSERM, U922, "Remodelage osseux et biomatériaux", LHEA-Faculté de Médecine, 49045 Angers Cedex, France
| | | | | | | | | | | |
Collapse
|
166
|
Solari MG, Srinivasan S, Boumaza I, Unadkat J, Harb G, Garcia-Ocana A, Feili-Hariri M. Marginal mass islet transplantation with autologous mesenchymal stem cells promotes long-term islet allograft survival and sustained normoglycemia. J Autoimmun 2009; 32:116-24. [PMID: 19217258 DOI: 10.1016/j.jaut.2009.01.003] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Revised: 01/06/2009] [Accepted: 01/12/2009] [Indexed: 12/25/2022]
Abstract
Allogeneic islet transplantation is an option to treat diabetes however there are obstacles that are limiting its clinical use. We have examined whether mesenchymal stem cells (MSC) improve islet graft survival and whether such therapy allows for better graft acceptance with reduced requirement for immunosuppression. In vitro-expanded syngeneic bone marrow-derived MSC were co-transplanted with islets into omental pouch in a rat model of streptozotocin-induced diabetes. Marginal mass syngeneic islet transplantation into the omentum with MSC promoted sustained normoglycemia. Interestingly, allogeneic islets +MSC, but not islets alone, with short-term use of immunosuppression enhanced long-term islet graft survival, insulin expression in the grafts and induced normal serum insulin levels and normoglycemia. T cells from recipients transplanted with allogeneic islets +MSC produced low levels of IFN-gamma and TNF-alpha upon ex-vivo activation, and this transplantation protocol promoted the generation of IL-10-secreting CD4(+) T cells. These data encourage further preclinical and eventually, clinical MSC-based islet transplantation to improve the outcome of allogeneic islet transplantation in the treatment of diabetes.
Collapse
Affiliation(s)
- Mario G Solari
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | |
Collapse
|
167
|
Pekkanen-Mattila M, Kerkelä E, Tanskanen JMA, Pietilä M, Pelto-Huikko M, Hyttinen J, Skottman H, Suuronen R, Aalto-Setälä K. Substantial variation in the cardiac differentiation of human embryonic stem cell lines derived and propagated under the same conditions--a comparison of multiple cell lines. Ann Med 2009; 41:360-70. [PMID: 19165643 DOI: 10.1080/07853890802609542] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
AIM The differentiation efficiencies of human embryonic stem cell (hESC) lines differ from each other. To assess this in more detail we studied the cardiac differentiation of eight hESC lines derived in the same laboratory. RESULTS Substantial variation in growth and in the ability to form beating areas was seen between the different hESC lines; line HS346 gave the best efficiency (9.4%), while HS293 did not differentiate into beating colonies at all. Nine germ layer and differentiation markers were quantified during early differentiation in four hESC lines. The expression levels of Brachyury T, MESP1 and NKX2.5 were highest in the most efficient cardiac line (HS346). A systematic characterization of the beating cells revealed proper cardiac marker expression, electrophysiological activity, and pharmacological response. CONCLUSIONS The hESC lines derived in the same laboratory varied considerably in their potential to differentiate into beating cardiomyocytes. None of the expression markers could clearly predict cardiac differentiation potential, although the expression of early cardiomyogenic genes was upregulated in the best cardiac line. The proper cardiomyocyte characteristics and pharmacological response indicate that these cells could be used as a model for human cardiomyocytes in pharmacological and toxicological analyses when investigating new heart medications or cardiac side-effects.
Collapse
Affiliation(s)
- Mari Pekkanen-Mattila
- REGEA, Institute for Regenerative Medicine, University of Tampere and Tampere University Hospital, Biokatu 12, Tampere, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
168
|
Qi CM, Ma GS, Liu NF, Shen CX, Chen Z, Liu XJ, Hu YP, Zhang XL, Chen JY, Ju,Yao-liang Tang SH. Identification and differentiation of magnetically labeled mesenchymal stem cells in vivo in swines with myocardial infarction. Int J Cardiol 2009; 131:417-9. [DOI: 10.1016/j.ijcard.2007.07.164] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2007] [Accepted: 07/06/2007] [Indexed: 01/31/2023]
|
169
|
Duan X, Guan H, Cao Y, Kleinerman ES. Murine bone marrow-derived mesenchymal stem cells as vehicles for interleukin-12 gene delivery into Ewing sarcoma tumors. Cancer 2009; 115:13-22. [PMID: 19051291 PMCID: PMC2652505 DOI: 10.1002/cncr.24013] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND This study evaluated the therapeutic efficacy of interleukin 12 (IL-12) gene therapy in Ewing sarcoma and whether murine mesenchymal stem cells (MSCs) could serve as vehicles for IL-12 gene delivery. METHODS MSCs were isolated from murine bone marrow cells. Cells were phenotyped using flow cytometry. Cultured MSCs differentiated into osteocytes and adipocytes using the appropriate media. Freshly isolated MSCs were transfected with adenoviral vectors containing either the beta-galactosidase (Ad:beta-gal) or the IL-12 (Ad:IL-12) gene. Expression of IL-12 was confirmed using reverse transcription polymerase chain reaction. Mice with TC71 Ewing sarcoma tumors were then treated intravenously with MSCs transfected with Ad:beta-gal or Ad:IL-12. Tumors were measured and analyzed by immunohistochemical analysis for expression of IL-12 protein. RESULTS Expression of both p35 and p40 IL-12 subunits was demonstrated in MSCs transfected in vitro with Ad:IL-12. IL-12 expression was seen in tumors from mice treated with MSCs transfected with Ad:IL-12. Tumor growth was also significantly inhibited compared with that in mice treated with MSCs transfected with Ad:beta-gal. CONCLUSIONS MSCs can be transfected with the IL-12 gene. These transfected cells localize to tumors after intravenous injection and induce local IL-12 protein production and the regression of established tumors.
Collapse
Affiliation(s)
- Xiaoping Duan
- Division of Pediatrics, The University of Texas M. D. Anderson Cancer, Houston, Texas
| | - Hui Guan
- Division of Pediatrics, The University of Texas M. D. Anderson Cancer, Houston, Texas
| | - Ying Cao
- Division of Pediatrics, The University of Texas M. D. Anderson Cancer, Houston, Texas
| | - Eugenie S. Kleinerman
- Division of Pediatrics, The University of Texas M. D. Anderson Cancer, Houston, Texas
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer, Houston, Texas
| |
Collapse
|
170
|
Abstract
Adult marrow-derived mesenchymal stem cells (MSCs) are able to differentiate into bone, cartilage, muscle, marrow stroma, tendon-ligament, fat and other connective tissues. The questions can be asked, what do MSCs do naturally and where is the MSC niche? New insight and clinical experience suggest that MSCs are naturally found as perivascular cells, summarily referred to as pericytes, which are released at sites of injury, where they secrete large quantities of bioactive factors that are both immunomodulatory and trophic. The trophic activity inhibits ischaemia-caused apoptosis and scarring while stimulating angiogenesis and the mitosis of tissue intrinsic progenitor cells. The immunomodulation inhibits lymphocyte surveillance of the injured tissue, thus preventing autoimmunity, and allows allogeneic MSCs to be used in a variety of clinical situations. Thus, a new, enlightened era of experimentation and clinical trials has been initiated with xenogenic and allogeneic MSCs.
Collapse
Affiliation(s)
- A I Caplan
- Skeletal Research Center, Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
171
|
Brooke G, Tong H, Levesque JP, Atkinson K. Molecular trafficking mechanisms of multipotent mesenchymal stem cells derived from human bone marrow and placenta. Stem Cells Dev 2008; 17:929-40. [PMID: 18564033 DOI: 10.1089/scd.2007.0156] [Citation(s) in RCA: 160] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
We compared potential trafficking mechanisms used by human (h) multipotent mesenchymal stem cells (MSC) derived from bone marrow (bm) or placenta (p). Both hbmMSC and hpMSC expressed a broad range of cell surface adhesion molecules including beta1-integrins (CD29) and CD44. Array data showed that both hbmMSC and hpMSC expressed mRNA for the cell adhesion molecules CD54 (ICAM-1), E-cadherin, CD166 (ALCAM), CD56 (NCAM), CD106 (VCAM-1), CD49a, b, c, e and f (integrins alpha1, 2, 3, 4 and 6), integrin alpha11, CD51 (integrin alphaV), and CD29 (integrins beta1). Functional binding of hpMSC, but not hbmMSC to VCAM-1 was demonstrated using recombinant chimeric constructs. Neither bone marrow nor placental MSC expressed ligands to endothelial selectins such as PSGL-1 or sialyl Lewis X (sLe(x)) carbohydrates and neither were able to bind functionally to chimeric constructs of the endothelial selectins CD62E (E-selectin) and CD62P (P-selectin). Furthermore, MSC expressed a restricted range of transferases necessary for expression of sLe(x), with no detectable expression of fucosyl transferases IV or VII. Placental MSC, but not hbmMSC, expressed mRNA for the chemokine receptors CCR1 and CCR3, and both hbmMSC and hpMSC expressed mRNA for CCR7, CCR8, CCR10, CCR11, CXCR4 and CXCR6. Intracellular chemokine receptor protein expression of CCR1, CCR3, CXCR3, CXCR4 and CXCR6 was detected in both hbmMSC and hpMSC. Cell surface expression of chemokine receptors was much more restricted with only CXCR6 displaying a strong signal on hbmMSC and hpMSC. Although cell surface expression of CXCR4 was not detected, MSC migrated in response to its ligand, CXCL12 (SDF-1). Thus, hbmMSC and hpMSC have an almost identical profile for cell surface adhesion and chemokine receptor molecules at the mRNA and protein levels. However, at the functional level, hpMSC likely utilise VLA-4-mediated binding in a superior manner to hbmMSC and thus may have superior engraftment properties to hbmMSC in vivo.
Collapse
Affiliation(s)
- Gary Brooke
- Adult Stem Cell and Haematopoietic Stem Cell Laboratories, Mater Medical Research Institute, Brisbane, Queensland, Australia
| | | | | | | |
Collapse
|
172
|
Boumaza I, Srinivasan S, Witt WT, Feghali-Bostwick C, Dai Y, Garcia-Ocana A, Feili-Hariri M. Autologous bone marrow-derived rat mesenchymal stem cells promote PDX-1 and insulin expression in the islets, alter T cell cytokine pattern and preserve regulatory T cells in the periphery and induce sustained normoglycemia. J Autoimmun 2008; 32:33-42. [PMID: 19062254 DOI: 10.1016/j.jaut.2008.10.004] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2008] [Revised: 10/28/2008] [Accepted: 10/31/2008] [Indexed: 12/11/2022]
Abstract
Cell-based therapies offer considerable promise for prevention or cure of diabetes. We explored the potential of autologous, self-renewing, mesenchymal stem cells (MSC) as a clinically-applicable approach to promote glucose homeostasis. In vitro-expanded syngeneic bone marrow-derived MSC were administered following or prior to diabetes induction into a rat model of streptozotocin-induced beta cell injury. MSC were CD45(-)/CD44(+)/CD54(+)/CD90(+)/CD106(+). MSC spontaneously secreted IL-6, HGF, TGF-beta1 and expressed high levels of SDF-1 and low levels of VEGF, IL-1beta and PGE(2), but no EGF, insulin or glucagon. MSC homed to the pancreas and this therapy allowed for enhanced insulin secretion and sustained normoglycemia. Interestingly, immunohistochemistry demonstrated that, the islets from MSC-treated rats expressed high levels of PDX-1 and that these cells were also positive for insulin staining. In addition, peripheral T cells from MSC-treated rats exhibited a shift toward IL-10/IL-13 production and higher frequencies of CD4(+)/CD8(+) Foxp3(+) T cells compared to the PBS-treated rats. These data suggest that the bioactive factors secreted by MSC establish a tissue microenvironment that supports beta cell activation/survival in the pancreas. In addition, because of anti-inflammatory and immunoregulatory effects of MSC on T cells, this work can lead to clinical trial of autologous MSC to prevent/cure type-1 diabetes.
Collapse
Affiliation(s)
- Imene Boumaza
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | |
Collapse
|
173
|
Yue WM, Liu W, Bi YW, He XP, Sun WY, Pang XY, Gu XH, Wang XP. Mesenchymal stem cells differentiate into an endothelial phenotype, reduce neointimal formation, and enhance endothelial function in a rat vein grafting model. Stem Cells Dev 2008; 17:785-93. [PMID: 18522495 DOI: 10.1089/scd.2007.0243] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Autologous vein grafts is still commonly used for arterial reconstructive procedures. Their success is limited by the development of neointimal hyperplasia. Clinical and experimental evidence suggest that the bone marrow derived mesenchymal stem cells (MSCs) participate in the neovascularization. The current study used a direct approach to test the hypothesis that, after vein grafting in a rat model, MSCs have potential effects on reendothelialization and neointimal formation. MSCs were isolated by bone marrow cell adherence. Autologously interpositioning left external jugular vein (LEJV) to left common carotid artery-induced vein grafting model of r at w as utilized. Vascular lesion formation after transplantation of MSCs labeled with 4',6-diamidino-2-phenylindole (DAPI) was investigated. Two weeks after implantation, immunofluorescence studies revealed that engrafted cells acquired an endothelial phenotype, and some expressed endothelial nitric oxide synthase (eNOS). Furthermore, proliferation of cells and neointimal formation decreased significantly after MSC implantation. Real-time reverse transcription-PCR and western blotting analysis showed a rise of eNOS expression in the MSC group compared with the vein grafting group. Therefore, engrafted MSCs appeared to differentiate into endothelial cells, diminish the neointima formation and contribute to the improvement on endothelial function, which indicates that MSCs may exert an important function as repair mechanism in vascular injury after vein grafting.
Collapse
Affiliation(s)
- Wei-Ming Yue
- Department of Cardiovascular Surgery, Qilu Hospital, Shandong University, Jinan, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
174
|
Potential role of mesenchymal stromal cells in pediatric hematopoietic SCT. Bone Marrow Transplant 2008; 42 Suppl 2:S60-6. [DOI: 10.1038/bmt.2008.286] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
175
|
Jun L, Minh D, Calvin W, Carolyn J T, Ray C J C, Dominique ST. The immature heart: the roles of bone marrow stromal stem cells in growth and myocardial repair. Open Cardiovasc Med J 2008; 1:27-33. [PMID: 18949088 PMCID: PMC2570564 DOI: 10.2174/1874192400701010027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2007] [Revised: 11/06/2007] [Accepted: 11/07/2007] [Indexed: 01/14/2023] Open
Abstract
Studies have shown that adult bone marrow derived stem cells (MSCs) can participate in repair of myocardial injury in adult hearts, as well as in cardiac growth during fetal development in utero. Yet, no studies have evaluated the role of MSCs with respect to normal growth or tissue repair in immature hearts after birth. The present study examines whether MSCs may participate in the myocardial growth and injury in the post-natal immature hearts. MSCs were isolated from adult Lewis rats and labeled with Lac-Z gene using retroviral vectors. These MSCs were injected systemically into groups of neonatal (NB=2days-old), immature (B=30days-old) and adult (A=>3months-old) isogeneic Lewis rats. Additionally, left coronary artery ligation was carried out in subgroups of immature (BL) and adult (AL) rats one week after MSCs injection. The hearts were harvested serially from 2-days to 6-weeks, stained with X-Gal for labeled MSCs. Cardiomyocyte phenotypic expression was evaluated by immunohistological staining for Troponin I-C and Connexin-43. Labeled MSCs were found to home into the bone marrow in all rats of different developmental stages. They could be recruited from bone marrow into the infarcted site of myocardium only in groups AL and BL. They were also capable of differentiating into cardiomyocyte phenotype after myocardial injury. In contrast to that reported in the developing fetus, MSCs did not appear to contribute to the growth of non-injured hearts after birth. However, they can be recruited from the bone marrow and regenerate damaged myocardium both in the adult and in the immature hearts.
Collapse
Affiliation(s)
- Luo Jun
- Division of Cardiothoracic Surgery, the Montreal General Hospital, MUHC, Canada
| | | | | | | | | | | |
Collapse
|
176
|
Bonora-Centelles A, Castell JV, Gómez-Lechón MJ. [Adipose tissue-derived stem cells: hepatic plasticity]. GASTROENTEROLOGIA Y HEPATOLOGIA 2008; 31:299-309. [PMID: 18448061 DOI: 10.1157/13119884] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Currently, the only effective treatment for end-stage liver disease is liver transplantation. The number of patients on the waiting list increases considerably each year, giving rise to a wide imbalance between supply and demand for healthy livers. Knowledge of stem cells and their possible use have awakened great interest in the field of hepatology, these cells being one of the most promising short-term alternatives. Hepatic stem cell therapy consists of the implantation of healthy cells capable of performing the functions that damaged cells are unable to carry out. Recent observations indicate that several stem cells can differentiate into distinct cell lineages. Hepatic differentiation of adult stem cells from several origins has yielded highly promising results. Adipose tissue in adults contains a reservoir of stem cells that can be induced and differentiated into different types of cells, showing a high degree of plasticity. Because of its abundance and easy access, adipose tissue is a promising source of adult stem cells for hepatic stem cell therapy. The present article reviews the progress made in the differentiation of adult stem cells from adipose tissue into cells with hepatic phenotype. We also discuss the potential application of this technique as a therapy for temporary metabolic support in patients with end-stage liver failure awaiting whole organ transplantation, as a method to support liver function and facilitate regeneration of the native liver in cases of fulminant hepatic failure, and as a treatment in patients with genetic metabolic defects in vital liver functions.
Collapse
Affiliation(s)
- Ana Bonora-Centelles
- Unidad de Hepatología Experimental, Centro de Investigación, Hospital Universitario La Fe, Valencia, España
| | | | | |
Collapse
|
177
|
Kato J, Hisha H, Wang XL, Mizokami T, Okazaki S, Li Q, Song CY, Maki M, Hosaka N, Adachi Y, Inaba M, Ikehara S. Contribution of neural cell adhesion molecule (NCAM) to hemopoietic system in monkeys. Ann Hematol 2008; 87:797-807. [PMID: 18575864 DOI: 10.1007/s00277-008-0513-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2007] [Accepted: 05/21/2008] [Indexed: 11/24/2022]
Abstract
Neural cell adhesion molecules (CD56) are important adhesion molecules that are mainly expressed on neural cells and natural killer cells. Although freshly isolated cynomolgus monkey bone marrow cells (BMCs) contained only a few CD56-positive cells, almost all the BM adherent cells (obtained after a 2- to 3-week culture of the BMCs) were stained positively with anti-CD56 monoclonal antibody (mAb). The BM adherent cells showed uniformly fibroblastic morphology and were negative for hematolymphoid markers (CD4, CD8, CD11b, CD14, CD34, and CD45). Adipogenesis and osteogenesis were observed under inductive culture conditions. The BM adherent cells had the ability to support hemopoiesis of hemopoietic stem cells (HSCs) in vitro, and the proliferation of HSCs was significantly inhibited by the addition of anti-CD56 mAb to the coculture system. CD56 molecules were also expressed on HSCs because about 20% of an HSC-enriched population (lineage-negative and blast-gated cells) was positive for CD56. In addition, the immunostaining of monkey BM sections revealed that many stromal cells were CD56-positive, and some CD56-positive stromal cells came into direct contact with CD56-positive hemopoietic cells. These results indicate that the CD56 molecule is expressed on both HSCs and BM stromal cells (containing MSCs) in monkeys, and therefore it can be speculated that CD56 also contributes to the human hematopoietic system.
Collapse
Affiliation(s)
- Junko Kato
- 1st Department of Pathology, Kansai Medical University, Moriguchi, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
178
|
Chen J, Park HC, Addabbo F, Ni J, Pelger E, Li H, Plotkin M, Goligorsky MS. Kidney-derived mesenchymal stem cells contribute to vasculogenesis, angiogenesis and endothelial repair. Kidney Int 2008; 74:879-89. [PMID: 18596729 PMCID: PMC2782525 DOI: 10.1038/ki.2008.304] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We isolated a clonal cell line (4E) from kidneys of mice expressing green fluorescent protein controlled by the endothelial-specific Tie2 promoter. When grown in a three-dimensional matrigel matrix they formed a fluorescent capillary network. In vivo angiogenesis assays using growth factor-depleted matrigel implanted plugs promoted a moderate angiogenesis of host endothelial cells. Using vascular endothelial growth factor (VEGF)-A and fibroblast growth factor-2 in the plugs containing 4E-cells resulted in a robust vasculogenesis. Transplantation of 4E cells into mice with acute renal ischemia showed selective engraftment in the ischemic kidney which promoted tubular regeneration by increasing epithelial proliferation and inhibiting apoptosis. This resulted in an accelerated functional recovery 3 days after ischemia. These mice showed a 5-fold increase in tissue VEGF expression compared to controls, but no difference in plasma VEGF level corresponding with better preservation of peritubular capillaries, perhaps due to a local paracrine effect following systemic 4E infusion. One month after ischemia, 9% of engrafted 4E cells expressed green fluorescent protein in the peritubular region while half of them expressed alpha-smooth muscle actin. Our study shows that kidney mesenchymal stem cells are capable of differentiation toward endothelial and smooth muscle cell lineages in vitro and in vivo, support new blood vessel formation in favorable conditions and promote functional recovery of an ischemic kidney.
Collapse
Affiliation(s)
- Jun Chen
- Department of Medicine, New York Medical College, Valhalla, New York 10595, USA.
| | | | | | | | | | | | | | | |
Collapse
|
179
|
Abstract
Mesenchymal stromal cells (MSCs) originally isolated from bone marrow have been derived from almost every tissue in the body. These multipotent cells can be differentiated in vitro and in vivo into various cell types of mesenchymal origin, such as bone, fat, and cartilage. Furthermore, under some experimental conditions, these cells can differentiate into a wider variety of cell types. Upon systemic administration, ex vivo expanded MSCs preferentially home to damaged tissues and participate in regeneration processes through their diverse biological properties. In vitro and in vivo data suggest that MSCs have low inherent immunogenicity and modulate/suppress immunologic responses through interactions with different immune cells. Ease of isolation and ex vivo expansion of MSCs, combined with their intriguing differentiation and immunomodulatory potential, and their impressive record of safety in clinical trials make these cells prime candidates for cellular therapy. Mesenchymal stromal cells derived from bone marrow are currently being evaluated for a wide range of clinical applications including for treatment of immune dysregulation disorders such as acute graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. In the future, MSCs might potentially provide novel therapeutic options for treatment/prevention of rejection and/or repair of organ allografts through their multifaceted properties.
Collapse
Affiliation(s)
- Peiman Hematti
- Department of Medicine, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI 53792, USA.
| |
Collapse
|
180
|
Bajada S, Mazakova I, Richardson JB, Ashammakhi N. Updates on stem cells and their applications in regenerative medicine. J Tissue Eng Regen Med 2008; 2:169-83. [PMID: 18493906 DOI: 10.1002/term.83] [Citation(s) in RCA: 215] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Stem cells have the capacity for self-renewal and capability of differentiation to various cell lineages. Thus, they represent an important building block for regenerative medicine and tissue engineering. These cells can be broadly classified into embryonic stem cells (ESCs) and non-embryonic or adult stem cells. ESCs have great potential but their use is still limited by several ethical and scientific considerations. The use of bone marrow-, umbilical cord-, adipose tissue-, skin- and amniotic fluid-derived mesenchymal stem cells might be an adequate alternative for translational practice. In particular, bone marrow-derived stem cells have been used successfully in the clinic for bone, cartilage, spinal cord, cardiac and bladder regeneration. Several preclinical experimental studies are under way for the application of stem cells in other conditions where current treatment options are inadequate. Stem cells can be used to improve healthcare by either augmenting the body's own regenerative potential or developing new therapies. This review is not meant to be exhaustive but gives a brief outlook on the past, present and the future of stem cell-based therapies in clinical practice.
Collapse
Affiliation(s)
- Stefan Bajada
- Institute for Science and Technology in Medicine, Keele University, Staffordshire, UK.
| | | | | | | |
Collapse
|
181
|
Fritz V, Noël D, Bouquet C, Opolon P, Voide R, Apparailly F, Louis-Plence P, Bouffi C, Drissi H, Xie C, Perricaudet M, Müller R, Schwarz E, Jorgensen C. Antitumoral activity and osteogenic potential of mesenchymal stem cells expressing the urokinase-type plasminogen antagonist amino-terminal fragment in a murine model of osteolytic tumor. Stem Cells 2008; 26:2981-90. [PMID: 18757301 DOI: 10.1634/stemcells.2008-0139] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Prostate cancer metastasis to bone results in mixed osteolytic and osteoblastic lesions associated with high morbidity, and there is mounting evidence that the urokinase-type plasminogen system is causatively involved in the progression of prostate cancer. Adult mesenchymal stem cells (MSCs) are promising tools for cell-mediated gene therapy with the advantage of osteogenic potential, a critical issue in the case of osteolytic metastases. In this study, we evaluated the therapeutic use of engineered murine MSCs for in vivo delivery of the urokinase-type plasminogen antagonist amino-terminal fragment (hATF) to impair osteolytic prostate cancer cell progression in bone and to repair bone lesions. Bioluminescence imaging revealed that both primary MSCs and the MSC line C3H10T1/2 (C3) expressing hATF (MSC-hATF) significantly inhibited intratibial PC-3 Luciferase (Luc) growth following coinjection in SCID mice. Furthermore, microcomputed tomography imaging of vascular network clearly demonstrated a significant decrease in tumor-associated angiogenesis and a protection from tumor-induced osteolysis in MSC-hATF-treated mice. Importantly, the osteogenic potential of MSC-hATF cells was unaffected, and an area of new bone formation was evidenced in 60% of animals. Together, these data support the concept of MSC-based therapy of tumor osteolysis disease, indicating that MSCs may combine properties of vehicle for angiostatic agent with osteogenic potential. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Vanessa Fritz
- Institut National de la Santé et de la Recherche Médicale U844, Montpellier, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
182
|
Weiss ML, Anderson C, Medicetty S, Seshareddy KB, Weiss RJ, VanderWerff I, Troyer D, McIntosh KR. Immune properties of human umbilical cord Wharton's jelly-derived cells. Stem Cells 2008; 26:2865-74. [PMID: 18703664 DOI: 10.1634/stemcells.2007-1028] [Citation(s) in RCA: 424] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cells isolated from Wharton's jelly, referred to as umbilical cord matrix stromal (UCMS) cells, adhere to a tissue-culture plastic substrate, express mesenchymal stromal cell (MSC) surface markers, self-renew, and are multipotent (differentiate into bone, fat, cartilage, etc.) in vitro. These properties support the notion that UCMS cells are a member of the MSC family. Here, the immune properties of UCMS cells are characterized in vitro. The overall hypothesis is that UCMS cells possess immune properties that would be permissive to allogeneic transplantation. For example, UCMS cells will suppress of the proliferation of "stimulated" lymphocytes (immune suppression) and have reduced immunogenicity (e.g., would be poor stimulators of allogeneic lymphocyte proliferation). Hypothesis testing was as follows: first, the effect on proliferation of coculture of mitotically inactivated human UCMS cells with concanavalin-A-stimulated rat splenocytes was assessed in three different assays. Second, the effect of human UCMS cells on one-way and two-way mixed lymphocyte reaction (MLR) assays was determined. Third, the expression of human leukocyte antigen (HLA)-G was examined in human UCMS cells using reverse transcription-polymerase chain reaction, since HLA-G expression conveys immune regulatory properties at the maternal-fetal interface. Fourth, the expression of CD40, CD80, and CD86 was determined by flow cytometry. Fifth, the cytokine expression of UCMS cells was evaluated by focused gene array. The results indicate that human UCMS cells inhibit splenocyte proliferation response to concanavalin A stimulation, that they do not stimulate T-cell proliferation in a one-way MLR, and that they inhibit the proliferation of stimulated T cells in a two-way MLR. Human UCMS cells do not inhibit nonstimulated splenocyte proliferation, suggesting specificity of the response. UCMS cells express mRNA for pan-HLA-G. UCMS cells do not express the costimulatory surface antigens CD40, CD80, and CD86. UCMS cells express vascular endothelial growth factor and interleukin-6, molecules previously implicated in the immune modulation observed in MSCs. In addition, the array data indicate that UCMS cells make a cytokine and other factors that may support hematopoiesis. Together, these results support previous observations made following xenotransplantation; for example, there was no evidence of frank immune rejection of undifferentiated UCMS cells. The results suggest that human UCMS will be tolerated in allogeneic transplantation. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Mark L Weiss
- Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas 66506, USA.
| | | | | | | | | | | | | | | |
Collapse
|
183
|
Spitkovsky D, Hescheler J. Adult mesenchymal stromal stem cells for therapeutic applications. MINIM INVASIV THER 2008; 17:79-90. [PMID: 18465443 DOI: 10.1080/13645700801969758] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stromal stem cells (MSC) can be found in almost any adult organ. They can be isolated and expanded within several weeks up to hundreds of millions of cells. The cell isolation based on the surface antigen expression may significantly enrich for the desired cell population and reduce the time required for cell expansion. MSC display a unique molecular signature which clearly discriminates them from other stem cell types. MSC can be differentiated into the cells of several lineages. Additionally, the unique biological properties of MSC are mediated by strong immunomodulatory activity and by paracrine mechanisms. Potential therapeutic applications of the cells require clinically compliant protocols for cell isolation and expansion. The therapeutic utility of MSC has been evaluated and found to be useful in several pre-clinical animal models as well as in clinical trials.
Collapse
Affiliation(s)
- D Spitkovsky
- Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | | |
Collapse
|
184
|
Swart J, Martens A, Wulffraat N. Mesenchymal stem cells: a future for the treatment of arthritis? Joint Bone Spine 2008; 75:379-82. [PMID: 18558507 DOI: 10.1016/j.jbspin.2008.04.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Accepted: 04/16/2008] [Indexed: 12/20/2022]
|
185
|
Microchimeric fetal cells cluster at sites of tissue injury in lung decades after pregnancy. Reprod Biomed Online 2008; 16:382-90. [PMID: 18339261 DOI: 10.1016/s1472-6483(10)60600-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Fetal cells trafficking into maternal blood during pregnancy engraft tissues and persist for decades in marrow and bone. While persistent fetal cells were initially implicated in autoimmune disease, animal studies suggest that microchimeric fetal cells play a broader role in response to tissue injury. This study investigated whether fetal cells participate in tissue repair after human pregnancy. Specimens were obtained from women undergoing surgery for suspected lung cancer. Y-fluorescence in-situ hybridization was performed on paraffin-embedded sections, with the investigator blinded to medical histories. Male cells were identified in lung/thymus tissue from all women with known male pregnancies, but not in those without sons. The frequency of male microchimeric cells was seven-fold greater in lung/thymus tissues than marrow and was two-fold greater than normal bone from the same women. Nested-polymerase chain reaction for sex determining region Y confirmed male DNA in tissues. Male cells in lung were clustered in tumour rather than surrounding healthy tissues. In conclusion, male presumed-fetal cells were identified in pathological post-reproductive tissues, where they were more likely to be located in diseased tissues at several-fold higher frequency than normal tissues. It is suggested that fetal cells are present at sites of tissue injury and may be stem cells, either recruited from marrow or having proliferated locally.
Collapse
|
186
|
Stem cells as potential novel therapeutic strategy for inflammatory bowel disease. J Crohns Colitis 2008; 2:99-106. [PMID: 21172199 DOI: 10.1016/j.crohns.2007.12.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Accepted: 12/21/2007] [Indexed: 02/08/2023]
Abstract
Hematopoietic stem cell transplantation and mesenchymal stromal cell therapy are currently under investigation as novel therapies for inflammatory bowel diseases. Hematopoietic stem cells (HSC) are thought to repopulate the immune system and reset the immunological response to luminal antigens. Mesenchymal stromal cells (MSC) are cells that have the capacity to differentiate into wide variety of distinct cell lineages and suppress immune responses in vitro and in vivo. Recent results from animal models and early human experience in graft-versus-host disease but also Crohn's Disease suggest that ex vivo expanded MSCs may have clinically useful immunomodulatory effects.
Collapse
|
187
|
Feng SW, Lu XL, Liu ZS, Zhang YN, Liu TY, Li JL, Yu MJ, Zeng Y, Zhang C. Dynamic distribution of bone marrow-derived mesenchymal stromal cells and change of pathology after infusing into mdx mice. Cytotherapy 2008; 10:254-64. [PMID: 18418771 DOI: 10.1080/14653240802020381] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND Mesenchymal stromal cells (MSC) are attractive candidates for the treatment of Duchenne muscular dystrophy (DMD) but how the donor MSC distribute in multiple organs and whether the increased dystrophin leads to a change in the pathology of mdx mice is still uncertain. In this research we detected the distribution of MSC and the pathology of mdx mice after MSC infusion. METHODS MSC were isolated from rat bone marrow (BM) and expanded in proliferation medium. MSC of the fifth passage were delivered intravenously into irradiated mdx mice. The distribution of MSC labeled by [3H]TdR into a recipient's organs was calculated by radioactivity. The expression of dystrophin was detected at weeks 4, 8, 12 and 16 after MSC transplantation by immunofluorescence staining, RT-PCR and Western blot. Serum creatine kinase (CK) and centrally nucleated fiber (CNF) were also detected to assess the change in pathology. RESULTS 24-48 h after transplantation, MSC were mainly found in the BM, liver and lung. The radioactivity in these organs decreased, whereas skeletal and myocardial muscle radioactivity increased gradually over time. In accordance with the increased radioactivity in skeletal muscle, the amount of dystrophin-positive myofibers increased. Furthermore, serum CK and CNF decreased slightly, suggesting specific pathophysiologic features of the dystrophic muscle were partially restored. DISCUSSION Upon certification of the distribution of transplanted MSC in irradiated mdx mice, we found evidence of myogenic differentiation of MSC in skeletal muscle. This research may help us understand the mechanism of therapy of MSC transplantation.
Collapse
Affiliation(s)
- S-W Feng
- Department of Neurology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
188
|
Immunological properties of umbilical cord blood-derived mesenchymal stromal cells. Cell Immunol 2008; 251:116-23. [PMID: 18495100 DOI: 10.1016/j.cellimm.2008.04.003] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2008] [Revised: 03/29/2008] [Accepted: 04/04/2008] [Indexed: 12/12/2022]
Abstract
Mesenchymal stromal cells (MSCs) are promising candidates for developing cell therapies for intractable diseases. To assess the feasibility of transplantation with human umbilical cord blood (hUCB)-derived MSCs, we analyzed the ability of these cells to function as alloantigen-presenting cells (APC) in vitro. hUCB-MSCs were strongly positive for MSC-related antigens and stained positively for human leukocyte antigen (HLA)-AB and negatively for HLA-DR. When treated with interferon (IFN)-gamma, the expression of HLA-AB and HLA-DR, but not the co-stimulatory molecules CD80 and CD86, was increased. hUCB-MSCs did not provoke allogeneic PBMC (peripheral blood mononuclear cell) proliferation, even when their HLA-molecule expression was up-regulated by IFN-gamma pretreatment. When added to a mixed lymphocyte reaction (MLR), hUCB-MSCs actively suppressed the allogeneic proliferation of the responder lymphocytes. This suppressive effect was mediated by soluble factors. We conclude that hUCB-MSCs can suppress the allogeneic response of lymphocytes and may thus be useful in allogeneic cell therapies.
Collapse
|
189
|
Bifari F, Lisi V, Mimiola E, Pasini A, Krampera M. Immune Modulation by Mesenchymal Stem Cells. ACTA ACUST UNITED AC 2008; 35:194-204. [PMID: 21547117 DOI: 10.1159/000128968] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Accepted: 03/05/2008] [Indexed: 12/27/2022]
Abstract
SUMMARY: Mesenchymal stem cells (MSCs) and their stromal progeny may be considered powerful regulatory cells, a sort of dendritic cell counterpart, which influence all the main immune effectors and functional roles in vivo, as well as potential applications in the treatment of a number of human immunological diseases. By choosing MSC tissue origin, cell dose, administration route, and treatment schedule, all the potential side effects related to MSC use, including tumor growth enhancement, have to be well considered to maximize the benefits of MSC-depen-dent immune regulation without significant risks for the patients.
Collapse
Affiliation(s)
- Francesco Bifari
- Stem Cell Research Laboratory, Section of Hematology, Department of Clinical and Experimental Medicine, University of Verona, Italy
| | | | | | | | | |
Collapse
|
190
|
Cheng Z, Liu X, Ou L, Zhou X, Liu Y, Jia X, Zhang J, Li Y, Kong D. Mobilization of mesenchymal stem cells by granulocyte colony-stimulating factor in rats with acute myocardial infarction. Cardiovasc Drugs Ther 2008; 22:363-71. [PMID: 18461431 DOI: 10.1007/s10557-008-6110-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Accepted: 03/27/2008] [Indexed: 01/16/2023]
Abstract
PURPOSE Intravenous delivery of mesenchymal stem cells (MSCs), a noninvasive strategy for myocardial repair after acute myocardial infarction (MI), is limited by the low percentage of MSCs migration to the heart. The purpose of this study was to test whether granulocyte colony-stimulating factor (G-CSF) would enhance the colonization of intravenously infused MSCs in damaged heart in a rat model of acute MI. METHODS After induction of anterior MI, Sprague-Dawley rats were randomized to receive: (1) saline (n=9); (2) MSCs (n=15); and (3) MSCs plus G-CSF (50 microg/kg/day for 5 consecutive days, n=13). RESULTS Flow cytometry revealed that G-CSF slightly increased surface CXCR4 expression on MSCs in vitro. After completion of G-CSF administration, MSCs showed a significantly lower colonization in bone marrow and a trend toward higher localization in the infarcted myocardium. At 3 months, vessel density in the infarct region of heart was significantly increased in MSCs group and trended to increase in MSCs+G-CSF group. However, echocardiographic and hemodynamic parameters, including left ventricular (LV) end-diastolic diameters, ejection fraction, and +/-dP/dtmax, were not statistically different. Morphological analysis showed that infarct size and collagen content were similar in the three groups. Immunohistochemistry revealed that the combined therapy accelerated endothelial recovery of the blood vessels in the ischemic myocardium. However, myocardial regeneration resulting from MSCs differentiation was not observed. CONCLUSIONS G-CSF enhanced the migration of systemically delivered MSCs from bone marrow to infarcted heart. However, the beneficial effect of this kind of migration is limited, as cardiac function did not improve.
Collapse
Affiliation(s)
- Zhaokang Cheng
- Key Laboratory of Bioactive Materials of Education of Ministry, College of Life Science, Nankai University, and Institute of Cardiovascular Disease, Pingjin Hospital, Tianjin, 300071, China
| | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Marrow stromal cells as universal donor cells for myocardial regenerative therapy: their unique immune tolerance. Ann Thorac Surg 2008; 85:571-9. [PMID: 18222266 DOI: 10.1016/j.athoracsur.2007.10.034] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Revised: 10/06/2007] [Accepted: 10/08/2007] [Indexed: 01/14/2023]
Abstract
BACKGROUND Recently rodent and porcine bone marrow stromal cells (MSCs) have been reported to be uniquely immune tolerant. To confirm these findings in human cells, we tested whether human MSCs are also immune tolerant, such that they can be used as universal donor cells for myocardial regenerative therapy. METHODS Immunocompetent female rats underwent coronary ligations (n = 90). In group I, lacZ-labeled male human MSCs were implanted into the peri-infarcted area. In groups II, III, and IV, isogeneic rat MSCs, culture medium, or human fibroblasts were injected, respectively. Echocardiography was carried out to assess cardiac function, and the specimens were examined serially for up to 8 weeks with immunohistochemistry, fluorescent in situ hybridization, and polymerase chain reaction to examine MSCs survival and differentiation. RESULTS Human MSCs survived within the rat myocardium for more than 8 weeks without immunosuppression. Furthermore, the implanted MSCs significantly contributed to the improvement in ventricular function and attenuated left ventricular remodeling. No cellular infiltration characteristic of immune rejection was noted in contrast to group IV. CONCLUSIONS Human MSCs survived within this xenogeneic environment, and contributed to the improvement in cardiac function. Our findings support the feasibility of using these cells as universal donor cells for xenogeneic or allogeneic cell therapy, as they can be prepared and stored well in advance for urgent use. Allogeneic MSCs from healthy donors may be particularly useful for severely ill or elderly patients whose own MSCs could be dysfunctional.
Collapse
|
192
|
Transplantation of magnetically labeled mesenchymal stem cells improves cardiac function in a swine myocardial infarction model. Chin Med J (Engl) 2008. [DOI: 10.1097/00029330-200803020-00016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
193
|
Effect of mesenchymal stem cell transplantation on the engraftment of human hematopoietic stem cells and leukemic cells in mice model. Int J Hematol 2008; 87:327-37. [PMID: 18293059 DOI: 10.1007/s12185-008-0041-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2007] [Revised: 01/24/2008] [Accepted: 01/26/2008] [Indexed: 02/07/2023]
Abstract
We investigated the effect of human bone marrow-derived mesenchymal stem cells on engraftment of human umbilical cord blood CD34+ cells and acute myelogenous leukemia cells and also assessed the homing capability of MSCs. Forty-two NOD/SCID mice were administered sublethal irradiation followed by various cell doses of intravenous UCB CD34+ cells with or without MSCs. Another 12 NOD/SCID mice were also sublethally irradiated followed by intravenous injection of AML cells with or without MSCs. In ten of these mice, MSCs were genetically modified with an adenoviral vector encoding eGFP gene for tracking purpose. Cotransplantation of UCB CD34+)cells and MSCs resulted in a significant increase in bone marrow engraftment after 6 weeks, and the engraftment promoting effect of MSCs was proportional to the dose of MSCs and obvious when low doses of UCB CD34+ cells were given. There was no effect of MSCs on AML cells engraftment. All of the ten mice transplanted with eGFP-transfected MSCs showed positive for eGFP in their major organs. These data demonstrate that MSCs promote engraftment of UCB CD34+ cells in bone marrow, but exert no effect on engraftment of AML cells, and are capable of homing to the major organs including bone marrow following intravenous infusion.
Collapse
|
194
|
Parameswaran R, Morad V, Laronne A, Rousso-Noori L, Shani N, Naffar-Abu-Amara S, Zipori D. Targeting the Bone Marrow with Activin A-Overexpressing Embryonic Multipotent Stromal Cells Specifically Modifies B Lymphopoiesis. Stem Cells Dev 2008; 17:93-106. [DOI: 10.1089/scd.2007.0099] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Reshmi Parameswaran
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Vered Morad
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ayelet Laronne
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Liat Rousso-Noori
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Nir Shani
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Suha Naffar-Abu-Amara
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Dov Zipori
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
195
|
Kim BS, Lee CCI, Christensen JE, Huser TR, Chan JW, Tarantal AF. Growth, Differentiation, and Biochemical Signatures of Rhesus Monkey Mesenchymal Stem Cells. Stem Cells Dev 2008; 17:185-98. [DOI: 10.1089/scd.2007.0076] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- Brandon S. Kim
- California National Primate Research Center, University of California, Davis, CA 95616-8542
| | - C. Chang I. Lee
- California National Primate Research Center, University of California, Davis, CA 95616-8542
| | - Jared E. Christensen
- California National Primate Research Center, University of California, Davis, CA 95616-8542
| | - Thomas R. Huser
- NSF Center for Biophotonics Science and Technology, University of California, Davis, CA 95616-8542
- Department of Internal Medicine, University of California, Davis, CA 95616-8542
| | - James W. Chan
- NSF Center for Biophotonics Science and Technology, University of California, Davis, CA 95616-8542
| | - Alice F. Tarantal
- California National Primate Research Center, University of California, Davis, CA 95616-8542
- Department of Pediatrics, University of California, Davis, CA 95616-8542
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California, Davis, CA 95616-8542
| |
Collapse
|
196
|
Caplan AI. Adult mesenchymal stem cells for tissue engineering versus regenerative medicine. J Cell Physiol 2008; 213:341-7. [PMID: 17620285 DOI: 10.1002/jcp.21200] [Citation(s) in RCA: 1341] [Impact Index Per Article: 78.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Adult mesenchymal stem cells (MSCs) can be isolated from bone marrow or marrow aspirates and because they are culture-dish adherent, they can be expanded in culture while maintaining their multipotency. The MSCs have been used in preclinical models for tissue engineering of bone, cartilage, muscle, marrow stroma, tendon, fat, and other connective tissues. These tissue-engineered materials show considerable promise for use in rebuilding damaged or diseased mesenchymal tissues. Unanticipated is the realization that the MSCs secrete a large spectrum of bioactive molecules. These molecules are immunosuppressive, especially for T-cells and, thus, allogeneic MSCs can be considered for therapeutic use. In this context, the secreted bioactive molecules provide a regenerative microenvironment for a variety of injured adult tissues to limit the area of damage and to mount a self-regulated regenerative response. This regenerative microenvironment is referred to as trophic activity and, therefore, MSCs appear to be valuable mediators for tissue repair and regeneration. The natural titers of MSCs that are drawn to sites of tissue injury can be augmented by allogeneic MSCs delivered via the bloodstream. Indeed, human clinical trials are now under way to use allogeneic MSCs for treatment of myocardial infarcts, graft-versus-host disease, Crohn's Disease, cartilage and meniscus repair, stroke, and spinal cord injury. This review summarizes the biological basis for the in vivo functioning of MSCs through development and aging.
Collapse
Affiliation(s)
- Arnold I Caplan
- Skeletal Research Center, Department of Biology, Case Western Reserve University, Cleveland, Ohio, USA.
| |
Collapse
|
197
|
Slatter MA, Brigham K, Dickinson AM, Harvey HL, Barge D, Jackson A, Bown N, Flood TJ, Cant AJ, Abinun M, Gennery AR. Long-term immune reconstitution after anti-CD52-treated or anti-CD34-treated hematopoietic stem cell transplantation for severe T-lymphocyte immunodeficiency. J Allergy Clin Immunol 2007; 121:361-7. [PMID: 18086494 DOI: 10.1016/j.jaci.2007.10.035] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2007] [Revised: 10/25/2007] [Accepted: 10/26/2007] [Indexed: 10/22/2022]
Abstract
BACKGROUND Results of treatment of severe T-lymphocyte immunodeficiencies by means of hematopoietic stem cell (HSC) transplantation have improved. T cell-depleted haploidentical transplantations are successful if there is no HLA-identical donor. Methods to remove T lymphocytes include addition of anti-CD52 antibodies and CD34(+) HSC selection. OBJECTIVE Assessment of long-term immune function is important after these treatments. We looked at immune reconstitution in 36 survivors for more than 2 years after HSC transplantation for severe T-lymphocyte immunodeficiencies and compared engraftment quality between the 2 T-lymphocyte depletion methods. METHODS Chimerism, T- and B-lymphocyte subsets, immunoglobulin levels, and specific antibody production at last follow-up were examined. The chi(2) (Fisher exact test) and Wilcoxon rank sum analyses were used to compare the groups. RESULTS Nineteen patients received anti-CD52-treated and 19 anti-CD34-treated HSCs. More anti-CD52-treated patients had full donor myeloid chimerism (P = .025). All patients had full donor T-lymphocyte chimerism. There was no difference in donor B-lymphocyte chimerism, but significantly more anti-CD52-treated patients had class-switched memory B lymphocytes (P = .024), normal IgG levels, and normal responses to tetanus and Haemophilus influenzae type B vaccination. More anti-CD52-treated patients with common gamma chain or Janus-associated kinase 3 severe combined immunodeficiency had donor B lymphocytes. CONCLUSION Long-term T-lymphocyte function is good with either treatment method, with a low incidence of graft-versus-host disease. The results imply more incomplete donor chimerism in anti-CD34-treated patients with less B-lymphocyte function.
Collapse
Affiliation(s)
- Mary A Slatter
- Department of Paediatric Immunology, Newcastle upon Tyne Hospitals Foundation Trust, Newcastle upon Tyne, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
198
|
Chamberlain J, Yamagami T, Colletti E, Theise ND, Desai J, Frias A, Pixley J, Zanjani ED, Porada CD, Almeida-Porada G. Efficient generation of human hepatocytes by the intrahepatic delivery of clonal human mesenchymal stem cells in fetal sheep. Hepatology 2007; 46:1935-45. [PMID: 17705296 DOI: 10.1002/hep.21899] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
UNLABELLED Alternative methods to whole liver transplantation require a suitable cell that can be expanded to obtain sufficient numbers required for successful transplantation while maintaining the ability to differentiate into hepatocytes. Mesenchymal stem cells (MSCs) possess several advantageous characteristics for cell-based therapy and have been shown to be able to differentiate into hepatocytes. Thus, we investigated whether the intrahepatic delivery of human MSCs is a safe and effective method for generating human hepatocytes and whether the route of administration influences the levels of donor-derived hepatocytes and their pattern of distribution throughout the parenchyma of the recipient's liver. Human clonally derived MSCs were transplanted by an intraperitoneal (n = 6) or intrahepatic (n = 6) route into preimmune fetal sheep. The animals were analyzed 56-70 days after transplantation by immunohistochemistry, enzyme-linked immunosorbent assay, and flow cytometry. The intrahepatic injection of human MSCs was safe and resulted in more efficient generation of hepatocytes (12.5% +/- 3.5% versus 2.6% +/- 0.4%). The animals that received an intrahepatic injection exhibited a widespread distribution of hepatocytes throughout the liver parenchyma, whereas an intraperitoneal injection resulted in a preferential periportal distribution of human hepatocytes that produced higher amounts of albumin. Furthermore, hepatocytes were generated from MSCs without the need to first migrate/lodge to the bone marrow and give rise to hematopoietic cells. CONCLUSION Our studies provide evidence that MSCs are a valuable source of cells for liver repair and regeneration and that, by the alteration of the site of injection, the generation of hepatocytes occurs in different hepatic zones, suggesting that a combined transplantation approach may be necessary to successfully repopulate the liver with these cells.
Collapse
Affiliation(s)
- Jason Chamberlain
- Department of Animal Biotechnology, University of Nevada at Reno, Reno, NV 89557-0104, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
199
|
Abstract
Mesenchymal stem cells (MSCs) from adult marrow can differentiate in vitro and in vivo into various cell types, such as bone, fat and cartilage. MSCs preferentially home to damaged tissue and may have therapeutic potential. In vitro data suggest that MSCs have low inherent immunogenicity as they induce little, if any, proliferation of allogeneic lymphocytes. Instead, MSCs appear to be immunosuppressive in vitro. They inhibit T-cell proliferation to alloantigens and mitogens and prevent the development of cytotoxic T-cells. In vivo, MSCs prolong skin allograft survival and have several immunomodulatory effects, which are presented and discussed in the present study. Possible clinical applications include therapy-resistant severe acute graft-versus-host disease, tissue repair, treatment of rejection of organ allografts and autoimmune disorders.
Collapse
Affiliation(s)
- K Le Blanc
- Division of Clinical Immunology, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden.
| | | |
Collapse
|
200
|
Christopeit M, Schendel M, Föll J, Müller LP, Keysser G, Behre G. Marked improvement of severe progressive systemic sclerosis after transplantation of mesenchymal stem cells from an allogeneic haploidentical-related donor mediated by ligation of CD137L. Leukemia 2007; 22:1062-4. [PMID: 17972956 DOI: 10.1038/sj.leu.2404996] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|