1
|
Kihara Y, Tanaka Y, Ikeda M, Homma J, Takagi R, Ishigaki K, Yamanouchi K, Honda H, Nagata S, Yamato M. In utero transplantation of myoblasts and adipose-derived mesenchymal stem cells to murine models of Duchenne muscular dystrophy does not lead to engraftment and frequently results in fetal death. Regen Ther 2022; 21:486-493. [PMID: 36313392 PMCID: PMC9596598 DOI: 10.1016/j.reth.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/19/2022] [Accepted: 10/09/2022] [Indexed: 11/07/2022] Open
Abstract
Introduction Duchenne muscular dystrophy (DMD) is a progressive disease that leads to damage of muscle and myocardium due to genetic abnormalities in the dystrophin gene. In utero cell transplantation that might facilitate allogenic transplantation is worth considering to treat this disease. Methods We performed allogeneic in utero transplantation of GFP-positive myoblasts and adipose-derived mesenchymal stem cells into murine DMD model animals. The transplantation route in this study was fetal intraperitoneal transplantation and transplacental transplantation. Transplanted animals were examined at 4-weeks old by immunofluorescence staining and RT-qPCR. Results No GFP-positive cells were found by immunofluorescence staining of skeletal muscle and no GFP mRNA was detected by RT-qPCR in any animal, transplantation method and cell type. Compared with previous reports, myoblast transplantation exhibited an equivalent mortality rate, but adipose-derived stem cell (ASC) transplantation produced a higher mortality rate. Conclusions In utero transplantation of myoblasts or ASCs to murine models of DMD does not lead to engraftment and, in ASC transplantation primarily, frequently results in fetal death.
Collapse
Affiliation(s)
- Yuki Kihara
- Department of Pediatrics, Tokyo Women’s Medical University, School of Medicine, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan,Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Yukie Tanaka
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Japan
| | - Masanari Ikeda
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Japan
| | - Jun Homma
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Ryo Takagi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Keiko Ishigaki
- Department of Pediatrics, Tokyo Women’s Medical University, School of Medicine, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Keitaro Yamanouchi
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Japan
| | - Hiroaki Honda
- Human Disease Models, Institute of Laboratory Animals, Tokyo Women's Medical University, Tokyo, Japan
| | - Satoru Nagata
- Department of Pediatrics, Tokyo Women’s Medical University, School of Medicine, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Masayuki Yamato
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan,Corresponding author. Fax: +81 3-3359-6046.
| |
Collapse
|
2
|
Genetic neuromuscular disorders: living the era of a therapeutic revolution. Part 1: peripheral neuropathies. Neurol Sci 2019; 40:661-669. [DOI: 10.1007/s10072-019-03778-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/16/2019] [Indexed: 12/17/2022]
|
3
|
Chan DC, Chiu CY, Lan KC, Weng TI, Yang RS, Liu SH. Transplantation of human skeletal muscle-derived progenitor cells ameliorates knee osteoarthritis in streptozotocin-induced diabetic mice. J Orthop Res 2017; 35:1886-1893. [PMID: 27935109 DOI: 10.1002/jor.23503] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 12/05/2016] [Indexed: 02/04/2023]
Abstract
The epidemiological and experimental evidence suggests that diabetes can be an independent risk factor for osteoarthritis. The osteoarthritis-like cartilage damage has been shown in streptozotocin-induced diabetic mice. The therapeutic effects of human skeletal muscle-derived progenitor cells (HSMPCs) on diabetic osteoarthritis still remain unclear. Here, we investigated the therapeutic potential of HSMPCs on diabetic knee osteoarthritis. The in vitro chondrogenic ability of HSMPCs was determined by pellet culture assay. Male mice were used to develop the model of streptozotocin-induced type 1 diabetes and its related osteoarthritis. HSMPCs were injected intra-articularly to rescue osteoarthritis. Protein expressions of advanced glycation end-products, cyclooxygenase-2, and type-2 collagen in tissues were determined by immunohistochemistry. The pellet culture assay showed that HSMPCs cultured in differentiation medium for chondrogenesis significantly produced larger pellets with an overproduction of extracellular matrix than in growth medium. In in vivo experiments, intra-articular injection of HSMPCs for 4 weeks significantly prevented the progression of degenerative changes in the cartilage of streptozotocin-induced diabetic mice, including an obvious increase of total articular cartilage thickness and a decrease of fibrous cartilage thickness. HSMPCs transplantation also exerted the decline in advanced glycation end-products and cyclooxygenase-2 protein expression, but increased the type-2 collagen protein expression in streptozotocin-induced osteoarthritic cartilages. Moreover, HSMPCs transplantation also inhibited the increased serum interleukin-6 and matrix metalloproteinase-3 levels in diabetic mice. These results demonstrated for the first time that HSMPCs transplantation ameliorates cartilage degeneration in diabetes-related osteoarthritis mice. These findings suggest that HSMPCs transplantation may apply as a potential therapeutic use of diabetes-related osteoarthritis. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:1886-1893, 2017.
Collapse
Affiliation(s)
- Ding-Cheng Chan
- Department of Geriatrics and Gerontology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chen-Yuan Chiu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Cell and Tissue Engineering, Changhua Christian Hospital, Changhua, Taiwan
| | - Kuo-Cheng Lan
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Te-I Weng
- Department of Forensic Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Rong-Sen Yang
- Departments of Orthopaedics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
4
|
Adaptive Immune Response Impairs the Efficacy of Autologous Transplantation of Engineered Stem Cells in Dystrophic Dogs. Mol Ther 2016; 24:1949-1964. [PMID: 27506452 DOI: 10.1038/mt.2016.163] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 08/03/2016] [Indexed: 12/27/2022] Open
Abstract
Duchenne muscular dystrophy is the most common genetic muscular dystrophy. It is caused by mutations in the dystrophin gene, leading to absence of muscular dystrophin and to progressive degeneration of skeletal muscle. We have demonstrated that the exon skipping method safely and efficiently brings to the expression of a functional dystrophin in dystrophic CD133+ cells injected scid/mdx mice. Golden Retriever muscular dystrophic (GRMD) dogs represent the best preclinical model of Duchenne muscular dystrophy, mimicking the human pathology in genotypic and phenotypic aspects. Here, we assess the capacity of intra-arterial delivered autologous engineered canine CD133+ cells of restoring dystrophin expression in Golden Retriever muscular dystrophy. This is the first demonstration of five-year follow up study, showing initial clinical amelioration followed by stabilization in mild and severe affected Golden Retriever muscular dystrophy dogs. The occurrence of T-cell response in three Golden Retriever muscular dystrophy dogs, consistent with a memory response boosted by the exon skipped-dystrophin protein, suggests an adaptive immune response against dystrophin.
Collapse
|
5
|
Growth of bone marrow and skeletal muscle side population stem cells in suspension culture. Methods Mol Biol 2015; 1210:51-61. [PMID: 25173160 DOI: 10.1007/978-1-4939-1435-7_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
The ability to efficiently isolate and expand various stem cell populations in vitro is crucial for successful translation of cell-based therapies to the clinical setting. One such heterogeneous population that possesses a remarkable potential for the development of cell-based treatments for a variety of degenerative diseases and disorders is called the Side Population (SP). For many years, investigators have isolated these primitive cells based upon their ability to efflux the fluorophore Hoechst 33342. This attribute enabled separation of SP cells derived from multiple tissue sources from other endogenous cell populations using fluorescence-activated cell sorting (FACS). While all tissue-specific SP fractions appear to contain cells with multi-potent stem cell activity, the therapeutic utility of these cells has yet to be fully realized because of the scarcity of this fraction in vivo. In view of that, we developed a method to expand adult murine bone marrow and skeletal muscle-derived SP cells in vitro. Here, we describe a spinner-flask culture system that supports the growth of SP cells in suspension when they are combined with feeder cells cultured on spherical microcarriers. In this way, their distinguishing biological characteristics can be maintained, attachment-stimulated differentiation is avoided, and therapeutically relevant quantities of SP cells are generated. Modification of the described procedure may permit expansion of the SP from other relevant tissue sources and our method is amenable to establishing compliance with current good manufacturing practices.
Collapse
|
6
|
Sirabella D, De Angelis L, Berghella L. Sources for skeletal muscle repair: from satellite cells to reprogramming. J Cachexia Sarcopenia Muscle 2013; 4:125-36. [PMID: 23314905 PMCID: PMC3684700 DOI: 10.1007/s13539-012-0098-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 11/28/2012] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscle regeneration is the process that ensures tissue repair after damage by injury or in degenerative diseases such as muscular dystrophy. Satellite cells, the adult skeletal muscle progenitor cells, are commonly considered to be the main cell type involved in skeletal muscle regeneration. Their mechanism of action in this process is extensively characterized. However, evidence accumulated in the last decade suggests that other cell types may participate in skeletal muscle regeneration. Although their actual contribution to muscle formation and regeneration is still not clear; if properly manipulated, these cells may become new suitable and powerful sources for cell therapy of skeletal muscle degenerative diseases. Mesoangioblasts, vessel associated stem/progenitor cells with high proliferative, migratory and myogenic potential, are very good candidates for clinical applications and are already in clinical experimentation. In addition, pluripotent stem cells are very promising sources for regeneration of most tissues, including skeletal muscle. Conditions such as muscle cachexia or aging that severely alter homeostasis may be counteracted by transplantation of donor and/or recruitment and activation of resident muscle stem/progenitor cells. Advantages and limitations of different cell therapy approaches will be discussed.
Collapse
Affiliation(s)
- Dario Sirabella
- />Department of Biomedical Engineering, Columbia University, 2920 Broadway, New York, NY 10027-7164 USA
| | - Luciana De Angelis
- />DAHFMO, Unit of Histology and Medical Embryology, University of Roma “La Sapienza”, Via Scarpa, 14, 00161 Rome, Italy
| | - Libera Berghella
- />IRCCS Fondazione S. Lucia, Via del Fosso di Fiorano, 64, 00143 Rome, Italy
- />HudsonAlpha Institute for Biotechnology, 601 Genome Way, Huntsville, AL 35806 USA
| |
Collapse
|
7
|
Fuoco C, Salvatori ML, Biondo A, Shapira-Schweitzer K, Santoleri S, Antonini S, Bernardini S, Tedesco FS, Cannata S, Seliktar D, Cossu G, Gargioli C. Injectable polyethylene glycol-fibrinogen hydrogel adjuvant improves survival and differentiation of transplanted mesoangioblasts in acute and chronic skeletal-muscle degeneration. Skelet Muscle 2012; 2:24. [PMID: 23181356 PMCID: PMC3579757 DOI: 10.1186/2044-5040-2-24] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Accepted: 10/25/2012] [Indexed: 01/03/2023] Open
Abstract
UNLABELLED BACKGROUND Cell-transplantation therapies have attracted attention as treatments for skeletal-muscle disorders; however, such research has been severely limited by poor cell survival. Tissue engineering offers a potential solution to this problem by providing biomaterial adjuvants that improve survival and engraftment of donor cells. METHODS In this study, we investigated the use of intra-muscular transplantation of mesoangioblasts (vessel-associated progenitor cells), delivered with an injectable hydrogel biomaterial directly into the tibialis anterior (TA) muscle of acutely injured or dystrophic mice. The hydrogel cell carrier, made from a polyethylene glycol-fibrinogen (PF) matrix, is polymerized in situ together with mesoangioblasts to form a resorbable cellularized implant. RESULTS Mice treated with PF and mesoangioblasts showed enhanced cell engraftment as a result of increased survival and differentiation compared with the same cell population injected in aqueous saline solution. CONCLUSION Both PF and mesoangioblasts are currently undergoing separate clinical trials: their combined use may increase chances of efficacy for localized disorders of skeletal muscle.
Collapse
Affiliation(s)
- Claudia Fuoco
- Department of Biology, Tor Vergata Rome University, Rome, Italy
| | | | - Antonella Biondo
- Division of Regenerative Medicine, San Raffaele Scientific Institute, Milan, Italy
| | | | - Sabrina Santoleri
- Division of Regenerative Medicine, San Raffaele Scientific Institute, Milan, Italy
| | | | | | - Francesco Saverio Tedesco
- Division of Regenerative Medicine, San Raffaele Scientific Institute, Milan, Italy
- Department of Cell and Developmental Biology, UCL, London, UK
| | - Stefano Cannata
- Department of Biology, Tor Vergata Rome University, Rome, Italy
| | - Dror Seliktar
- Faculty of Biomedical Engineering, Technion – Israel Institute of Technology, Haifa, Israel
| | - Giulio Cossu
- Division of Regenerative Medicine, San Raffaele Scientific Institute, Milan, Italy
- Department of Cell and Developmental Biology, UCL, London, UK
| | - Cesare Gargioli
- Department of Biology, Tor Vergata Rome University, Rome, Italy
- IRCCS MultiMedica, Milan, Italy
| |
Collapse
|
8
|
Trophic actions of bone marrow-derived mesenchymal stromal cells for muscle repair/regeneration. Cells 2012; 1:832-50. [PMID: 24710532 PMCID: PMC3901134 DOI: 10.3390/cells1040832] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 09/28/2012] [Accepted: 10/09/2012] [Indexed: 12/30/2022] Open
Abstract
Bone marrow-derived mesenchymal stromal cells (BM-MSCs) represent the leading candidate cell in tissue engineering and regenerative medicine. These cells can be easily isolated, expanded in vitro and are capable of providing significant functional benefits after implantation in the damaged muscle tissues. Despite their plasticity, the participation of BM-MSCs to new muscle fiber formation is controversial; in fact, emerging evidence indicates that their therapeutic effects occur without signs of long-term tissue engraftment and involve the paracrine secretion of cytokines and growth factors with multiple effects on the injured tissue, including modulation of inflammation and immune reaction, positive extracellular matrix (ECM) remodeling, angiogenesis and protection from apoptosis. Recently, a new role for BM-MSCs in the stimulation of muscle progenitor cells proliferation has been demonstrated, suggesting the potential ability of these cells to influence the fate of local stem cells and augment the endogenous mechanisms of repair/regeneration in the damaged tissues.
Collapse
|
9
|
Sassoli C, Pini A, Chellini F, Mazzanti B, Nistri S, Nosi D, Saccardi R, Quercioli F, Zecchi-Orlandini S, Formigli L. Bone marrow mesenchymal stromal cells stimulate skeletal myoblast proliferation through the paracrine release of VEGF. PLoS One 2012; 7:e37512. [PMID: 22815682 PMCID: PMC3398011 DOI: 10.1371/journal.pone.0037512] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 04/24/2012] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are the leading cell candidates in the field of regenerative medicine. These cells have also been successfully used to improve skeletal muscle repair/regeneration; however, the mechanisms responsible for their beneficial effects remain to be clarified. On this basis, in the present study, we evaluated in a co-culture system, the ability of bone-marrow MSCs to influence C2C12 myoblast behavior and analyzed the cross-talk between the two cell types at the cellular and molecular level. We found that myoblast proliferation was greatly enhanced in the co-culture as judged by time lapse videomicroscopy, cyclin A expression and EdU incorporation. Moreover, myoblasts immunomagnetically separated from MSCs after co-culture expressed higher mRNA and protein levels of Notch-1, a key determinant of myoblast activation and proliferation, as compared with the single culture. Notch-1 intracellular domain and nuclear localization of Hes-1, a Notch-1 target gene, were also increased in the co-culture. Interestingly, the myoblastic response was mainly dependent on the paracrine release of vascular endothelial growth factor (VEGF) by MSCs. Indeed, the addition of MSC-derived conditioned medium (CM) to C2C12 cells yielded similar results as those observed in the co-culture and increased the phosphorylation and expression levels of VEGFR. The treatment with the selective pharmacological VEGFR inhibitor, KRN633, resulted in a marked attenuation of the receptor activation and concomitantly inhibited the effects of MSC-CM on C2C12 cell growth and Notch-1 signaling. In conclusion, this study provides novel evidence for a role of MSCs in stimulating myoblast cell proliferation and suggests that the functional interaction between the two cell types may be exploited for the development of new and more efficient cell-based skeletal muscle repair strategies.
Collapse
Affiliation(s)
- Chiara Sassoli
- Department of Human Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy
| | - Alessandro Pini
- Department of Human Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy
| | - Flaminia Chellini
- Department of Human Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy
| | - Benedetta Mazzanti
- Department of Hematology, Cord Blood Bank, Careggi Hospital, University of Florence, Florence, Italy
| | - Silvia Nistri
- Department of Human Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy
| | - Daniele Nosi
- Department of Human Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy
| | - Riccardo Saccardi
- Department of Hematology, Cord Blood Bank, Careggi Hospital, University of Florence, Florence, Italy
| | - Franco Quercioli
- National Institute of Optics (INO), National Research Council (CNR), Sesto Fiorentino, Florence, Italy
| | - Sandra Zecchi-Orlandini
- Department of Human Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy
| | - Lucia Formigli
- Department of Human Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy
- * E-mail:
| |
Collapse
|
10
|
Cassano M, Berardi E, Crippa S, Toelen J, Barthelemy I, Micheletti R, Chuah M, Vandendriessche T, Debyser Z, Blot S, Sampaolesi M. Alteration of cardiac progenitor cell potency in GRMD dogs. Cell Transplant 2012; 21:1945-67. [PMID: 22513051 DOI: 10.3727/096368912x638919] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Among the animal models of Duchenne muscular dystrophy (DMD), the Golden Retriever muscular dystrophy (GRMD) dog is considered the best model in terms of size and pathological onset of the disease. As in human patients presenting with DMD or Becker muscular dystrophies (BMD), the GRMD is related to a spontaneous X-linked mutation of dystrophin and is characterized by myocardial lesions. In this respect, GRMD is a useful model to explore cardiac pathogenesis and for the development of therapeutic protocols. To investigate whether cardiac progenitor cells (CPCs) isolated from healthy and GRMD dogs may differentiate into myocardial cell types and to test the feasibility of cell therapy for cardiomyopathies in a preclinical model of DMD, CPCs were isolated from cardiac biopsies of healthy and GRMD dogs. Gene profile analysis revealed an active cardiac transcription network in both healthy and GRMD CPCs. However, GRMD CPCs showed impaired self-renewal and cardiac differentiation. Population doubling and telomerase analyses highlighted earlier senescence and proliferation impairment in progenitors isolated from GRMD cardiac biopsies. Immunofluorescence analysis revealed that only wt CPCs showed efficient although not terminal cardiac differentiation, consistent with the upregulation of cardiac-specific proteins and microRNAs. Thus, the pathological condition adversely influences the cardiomyogenic differentiation potential of cardiac progenitors. Using PiggyBac transposon technology we marked CPCs for nuclear dsRed expression, providing a stable nonviral gene marking method for in vivo tracing of CPCs. Xenotransplantation experiments in neonatal immunodeficient mice revealed a valuable contribution of CPCs to cardiomyogenesis with homing differences between wt and dystrophic progenitors. These results suggest that cardiac degeneration in dystrophinopathies may account for the progressive exhaustion of local cardiac progenitors and shed light on cardiac stemness in physiological and pathological conditions. Furthermore, we provide essential information that canine CPCs may be used to alleviate cardiac involvement in a large preclinical model of DMD.
Collapse
Affiliation(s)
- M Cassano
- Laboratory of Translational Cardiomyology, Stem Cell Institute, Department of Development and Regeneration, University of Leuven (KU Leuven), Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Wright KT, El Masri W, Osman A, Chowdhury J, Johnson WEB, Franchina M, Lanzoni G, Cantoni S, Cavallini C, Bianchi F, Tazzari PL, Pasquinelli G, Foroni L, Ventura C, Grossi A, Bagnara GP. Concise review: Bone marrow for the treatment of spinal cord injury: mechanisms and clinical applications. Stem Cells 2011; 29:169-78. [PMID: 21732476 PMCID: PMC3083520 DOI: 10.1002/stem.570] [Citation(s) in RCA: 164] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Transplantation of bone marrow stem cells into spinal cord lesions enhances axonal regeneration and promotes functional recovery in animal studies. There are two types of adult bone marrow stem cell; hematopoietic stem cells (HSCs), and mesenchymal stem cells (MSCs). The mechanisms by which HSCs and MSCs might promote spinal cord repair following transplantation have been extensively investigated. The objective of this review is to discuss these mechanisms; we briefly consider the controversial topic of HSC and MSC transdifferentiation into central nervous system cells but focus on the neurotrophic, tissue sparing, and reparative action of MSC grafts in the context of the spinal cord injury (SCI) milieu. We then discuss some of the specific issues related to the translation of HSC and MSC therapies for patients with SCI and present a comprehensive critique of the current bone marrow cell clinical trials for the treatment of SCI to date.
Collapse
Affiliation(s)
- Karina T Wright
- Spinal Studies and Midlands Centre for Spinal Injuries, RJAH Orthopaedic Hospital, Oswestry, Shropshire, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
MacLean S, Khan WS, Malik AA, Anand S, Snow M. The potential of stem cells in the treatment of skeletal muscle injury and disease. Stem Cells Int 2011; 2012:282348. [PMID: 22220178 PMCID: PMC3246792 DOI: 10.1155/2012/282348] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Accepted: 09/18/2011] [Indexed: 01/05/2023] Open
Abstract
Tissue engineering is a pioneering field with huge advances in recent times. These advances are not only in the understanding of how cells can be manipulated but also in potential clinical applications. Thus, tissue engineering, when applied to skeletal muscle cells, is an area of huge prospective benefit to patients with muscle disease/damage. This could include damage to muscle from trauma and include genetic abnormalities, for example, muscular dystrophies. Much of this research thus far has been focused on satellite cells, however, mesenchymal stem cells have more recently come to the fore. In particular, results of trials and further research into their use in heart failure, stress incontinence, and muscular dystrophies are eagerly awaited. Although no doubt, stem cells will have much to offer in the future, the results of further research still limit their use.
Collapse
Affiliation(s)
- S. MacLean
- University of Manchester, Manchester M13 9PL, UK
| | - W. S. Khan
- Institute of Orthpaedics and Musculoskeletal Science, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, UK
| | - A. A. Malik
- Spinal Deformity Unit, Royal National Orthopaedic Hospital, Brockley Hill, Stanmore, Middlesex HA7 4LP, UK
| | - S. Anand
- Stepping Hill Hospital, Stockport SK2 7JE, UK
| | - M. Snow
- Department of Sports Inury, Royal National Orthopaedic Hospital, Birimingham B31 2AP, UK
| |
Collapse
|
13
|
Myoblast-mediated gene therapy via encephalomyosynangiosis – A novel strategy for local delivery of gene products to the brain surface. J Neurosci Methods 2011; 201:61-6. [DOI: 10.1016/j.jneumeth.2011.07.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2011] [Revised: 07/11/2011] [Accepted: 07/12/2011] [Indexed: 11/18/2022]
|
14
|
Nagy G, Gyurcsik B, Hoffmann EA, Körtvélyesi T. Theoretical design of a specific DNA-Zinc-finger protein interaction with semi-empirical quantum chemical methods. J Mol Graph Model 2011; 29:928-34. [PMID: 21470886 DOI: 10.1016/j.jmgm.2011.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 02/24/2011] [Accepted: 03/03/2011] [Indexed: 10/18/2022]
Abstract
The interactions of a zinc-finger (ZF) protein with DNA containing the specific recognition site of the ZF and with a non-specific DNA were studied with the semi-empirical quantum chemical method of PM6/Mozyme. The ZF protein (1MEY)-DNA complex structures were generated by docking calculations. The complex structures were reoptimized with the PM6/Mozyme method with implicit solvation in water. The structures were also calculated in the gas phase. The interaction enthalpies between the protein and DNA within the complexes obtained in the PM6/Mozyme with solvation optimized structures were calculated with the single-point PM6-DH2/Mozyme method (PM6 with dispersion, H-bond correction and Mozyme) with solvation. The results supported the specific and non-specific interactions in the complexes obtained from the docking experiments. The binding enthalpies of the specific and non-specific DNA binding to the protein differed significantly. The interactions between the nucleic acid strands in duplexes were also evaluated; these interactions between the base pairs were different because of the different "G…C:A…T" ratios in the DNA molecules studied. The stacking interactions between the nucleic bases were also characterized in the DNA duplexes.
Collapse
Affiliation(s)
- Gábor Nagy
- Department of Physical Chemistry and Material Sciences, University of Szeged, Aradi Vértanuk tere 1, H-6720 Szeged, Hungary
| | | | | | | |
Collapse
|
15
|
Meng J, Muntoni F, Morgan JE. Stem cells to treat muscular dystrophies – Where are we? Neuromuscul Disord 2011; 21:4-12. [DOI: 10.1016/j.nmd.2010.10.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 09/13/2010] [Accepted: 10/08/2010] [Indexed: 12/18/2022]
|
16
|
Sciorati C, Buono R, Azzoni E, Casati S, Ciuffreda P, D'Angelo G, Cattaneo D, Brunelli S, Clementi E. Co-administration of ibuprofen and nitric oxide is an effective experimental therapy for muscular dystrophy, with immediate applicability to humans. Br J Pharmacol 2010; 160:1550-60. [PMID: 20590643 PMCID: PMC2938824 DOI: 10.1111/j.1476-5381.2010.00809.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 03/08/2010] [Accepted: 03/11/2010] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND AND PURPOSE Current therapies for muscular dystrophy are based on corticosteroids. Significant side effects associated with these therapies have prompted several studies aimed at identifying possible alternative strategies. As inflammation and defects of nitric oxide (NO) generation are key pathogenic events in muscular dystrophies, we have studied the effects of combining the NO donor isosorbide dinitrate (ISDN) and the non-steroidal anti-inflammatory drug ibuprofen. EXPERIMENTAL APPROACH alpha-Sarcoglycan-null mice were treated for up to 8 months with ISDN (30 mg.kg(-1)) plus ibuprofen (50 mg.kg(-1)) administered daily in the diet. Effects of ISDN and ibuprofen alone were assessed in parallel. Drug effects on animal motility and muscle function, muscle damage, inflammatory infiltrates and cytokine levels, as well as muscle regeneration including assessment of endogenous stem cell pool, were measured at selected time points. KEY RESULTS Combination of ibuprofen and ISDN stimulated regeneration capacity, of myogenic precursor cells, reduced muscle necrotic damage and inflammation. Muscle function in terms of free voluntary movement and resistance to exercise was maintained throughout the time window analysed. The effects of ISDN and ibuprofen administered separately were transient and significantly lower than those induced by their combination. CONCLUSIONS AND IMPLICATIONS Co-administration of NO and ibuprofen provided synergistic beneficial effects in a mouse model of muscular dystrophy, leading to an effective therapy. Our results open the possibility of immediate clinical testing of a combination of ISDN and ibuprofen in dystrophic patients, as both components are approved for use in humans, with a good safety profile.
Collapse
Affiliation(s)
- Clara Sciorati
- San Raffaele Scientific Institute, Stem Cell Research InstituteMilan, Italy
| | - Roberta Buono
- Unit of Clinical Pharmacology, Department of Preclinical Sciences, University Hospital ‘Luigi Sacco’, Università di MilanoMilan, Italy,
| | - Emanuele Azzoni
- San Raffaele Scientific Institute, Stem Cell Research InstituteMilan, Italy
- Department of Experimental Medicine, University of Milano-BicoccaMonza, Italy
| | - Silvana Casati
- Unit of Clinical Pharmacology, Department of Preclinical Sciences, University Hospital ‘Luigi Sacco’, Università di MilanoMilan, Italy,
| | - Pierangela Ciuffreda
- Unit of Clinical Pharmacology, Department of Preclinical Sciences, University Hospital ‘Luigi Sacco’, Università di MilanoMilan, Italy,
| | | | - Dario Cattaneo
- Unit of Clinical Pharmacology, Department of Preclinical Sciences, University Hospital ‘Luigi Sacco’, Università di MilanoMilan, Italy,
| | - Silvia Brunelli
- San Raffaele Scientific Institute, Stem Cell Research InstituteMilan, Italy
- Department of Experimental Medicine, University of Milano-BicoccaMonza, Italy
| | - Emilio Clementi
- Unit of Clinical Pharmacology, Department of Preclinical Sciences, University Hospital ‘Luigi Sacco’, Università di MilanoMilan, Italy,
- E. Medea Scientific InstituteBosisio Parini, Italy
| |
Collapse
|
17
|
Martelly I, Singabraya D, Vandebrouck A, Papy-Garcia D, Cognard C, Raymond G, Guillet-Deniau I, Courty J, Constantin B. Glycosaminoglycan mimetics trigger IP3-dependent intracellular calcium release in myoblasts. Matrix Biol 2010; 29:317-29. [PMID: 20193761 DOI: 10.1016/j.matbio.2010.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Revised: 02/05/2010] [Accepted: 02/05/2010] [Indexed: 11/18/2022]
Abstract
Glycosaminoglycans (GAG) are sulfated polysaccharides that play an important role in regulating cell functions. GAG mimetics called RGTAs (for ReGeneraTing Agents) have been shown to stimulate tissue repair. In particular they accelerate myogenesis, in part via their heparin-mimetic property towards growth factors. RGTAs also increase activity of calcium-dependent intracellular protease suggesting an effect on calcium cellular homeostasis. This effect was presently investigated on myoblasts in vitro using one member of the RGTA family molecule named OTR4120. We have shown that OTR4120 or heparin induced transient increases of intracellular calcium concentration ([Ca(2+)]i) in pre-fusing myoblasts from both mouse SolD7 cell line and rat skeletal muscle satellite cells grown in primary culture by mobilising sarcoplasmic reticulum store. This [Ca(2+)]i was not mediated by ryanodine receptors but instead resulted from stimulation of the Inositol-3 phosphate-phospholipase C activation pathway. OTR4120-induced calcium transient was not mediated through an ATP, nor a tyrosine kinase, nor an acetylcholine receptor but principally through serotonin 5-HT2A receptor. This original finding shows that the GAG mimetic can induce calcium signal through serotonin receptors and the IP3 pathway may be relevant to its ability to favour myoblast differentiation. It supports a novel and unexpected function of GAGs in the regulation of calcium homeostasis.
Collapse
Affiliation(s)
- Isabelle Martelly
- Laboratoire de Recherche sur la Croissance Cellulaire, la Réparation et la Régénération Tissulaires (CRRET), UMR 7149-CNRS, Université Paris-Est Créteil, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Fiaschi T, Tedesco FS, Giannoni E, Diaz-Manera J, Parri M, Cossu G, Chiarugi P. Globular adiponectin as a complete mesoangioblast regulator: role in proliferation, survival, motility, and skeletal muscle differentiation. Mol Biol Cell 2010; 21:848-59. [PMID: 20089845 PMCID: PMC2836966 DOI: 10.1091/mbc.e09-04-0310] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
This article shows that globular adiponectin regulates vital cues of mesoangioblast, such as proliferation, survival, and migration toward myotubes and the myogenic properties. In vivo experiments confirm that globular adiponectin increases the survival, engraftment, and localization to muscle of mesoangioblasts in α-sarcoglycan-null mice. Mesoangioblasts are progenitor endowed with multipotent mesoderm differentiation ability. Despite the promising results obtained with mesoangioblast transplantation in muscle dystrophy, an improvement of their efficient engrafting and survival within damaged muscles, as well as their ex vivo activation/expansion and commitment toward myogenic lineage, is highly needed and should greatly increase their therapeutic potential. We show that globular adiponectin, an adipokine endowed with metabolic and differentiating functions for muscles, regulates vital cues of mesoangioblast cell biology. The adipokine drives mesoangioblasts to entry cell cycle and strongly counteracts the apoptotic process triggered by growth factor withdrawal, thereby serving as an activating and prosurvival stem cell factor. In addition, adiponectin provides a specific protection against anoikis, the apoptotic death due to lack of anchorage to extracellular matrix, suggesting a key protective role for these nonresident stem cells after systemic injection. Finally, adiponectin behaves as a chemoattractive factor toward mature myotubes and stimulates their differentiation toward the skeletal muscle lineage, serving as a positive regulator in mesoangioblast homing to injured or diseased muscles. We conclude that adiponectin exerts several advantageous effects on mesoangioblasts, potentially valuable to improve their efficacy in cell based therapies of diseased muscles.
Collapse
Affiliation(s)
- Tania Fiaschi
- Department of Biochemical Science, University of Florence, 50134 Florence, Italy
| | | | | | | | | | | | | |
Collapse
|
19
|
Tedesco FS, Dellavalle A, Diaz-Manera J, Messina G, Cossu G. Repairing skeletal muscle: regenerative potential of skeletal muscle stem cells. J Clin Invest 2010; 120:11-9. [PMID: 20051632 DOI: 10.1172/jci40373] [Citation(s) in RCA: 458] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Skeletal muscle damaged by injury or by degenerative diseases such as muscular dystrophy is able to regenerate new muscle fibers. Regeneration mainly depends upon satellite cells, myogenic progenitors localized between the basal lamina and the muscle fiber membrane. However, other cell types outside the basal lamina, such as pericytes, also have myogenic potency. Here, we discuss the main properties of satellite cells and other myogenic progenitors as well as recent efforts to obtain myogenic cells from pluripotent stem cells for patient-tailored cell therapy. Clinical trials utilizing these cells to treat muscular dystrophies, heart failure, and stress urinary incontinence are also briefly outlined.
Collapse
Affiliation(s)
- Francesco Saverio Tedesco
- Division of Regenerative Medicine, San Raffaele Scientific Institute, 58 via Olgettina, Milan, Italy
| | | | | | | | | |
Collapse
|
20
|
Quattrocelli M, Cassano M, Crippa S, Perini I, Sampaolesi M. Cell therapy strategies and improvements for muscular dystrophy. Cell Death Differ 2009; 17:1222-9. [DOI: 10.1038/cdd.2009.160] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
21
|
Abstract
Tissue and organ regeneration proceed in a coordinated manner to restore proper function after trauma. Vertebrate skeletal muscle has a remarkable ability to regenerate after repeated and complete destruction of the tissue, yet limited information is available on how muscle stem and progenitor cells, and other nonmuscle cells, reestablish homeostasis after the regenerative process. The genetic pathways that regulate the establishment of skeletal muscle in the embryo have been studied extensively, and many of the genes that govern muscle stem cell maintenance and commitment are redeployed during adult homeostasis and regeneration. Therefore, correlates can be made between embryonic muscle development and postnatal regeneration. However, there are some important distinctions between prenatal development and regeneration - in the context of the cells, niche, anatomy and the regulatory genes employed. The similarities and distinctions between these two scenarios are the focus of this review.
Collapse
Affiliation(s)
- S Tajbakhsh
- Stem Cells & Development, Department of Developmental Biology, Pasteur Institute, CNRS URA, Paris, France.
| |
Collapse
|
22
|
Gayraud-Morel B, Chrétien F, Tajbakhsh S. Skeletal muscle as a paradigm for regenerative biology and medicine. Regen Med 2009; 4:293-319. [PMID: 19317647 DOI: 10.2217/17460751.4.2.293] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Tissue development and regeneration share common features, since modules of regulatory pathways and transcription factors that are crucial for prenatal development are redeployed for tissue reconstruction after trauma. Regenerative medicine has therefore gained important insights through the study of developmental and regenerative biology. Moreover, diverse experimental models have been used to investigate the regeneration process in different tissues and organs. Paradoxically, little is known regarding the relative contribution of stem cells with respect to the supporting tissue during tissue regeneration. Particular attention will be given to mouse models using distinct injury paradigms to investigate the regenerative biology of skeletal muscle. An understanding of the response of stem and parenchymal cells is crucial for the development of clinical strategies to combat the normal decline in tissue performance during aging or its reconstitution after trauma and during disease. This review addresses these issues, focusing on muscle regeneration and how different factors, including genes, cells and the environment, impinge on this process.
Collapse
Affiliation(s)
- Barbara Gayraud-Morel
- Stem Cells & Development, Department of Developmental Biology, Pasteur Institute, CNRS URA 2578, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France
| | | | | |
Collapse
|
23
|
Jazedje T, Secco M, Vieira NM, Zucconi E, Gollop TR, Vainzof M, Zatz M. Stem cells from umbilical cord blood do have myogenic potential, with and without differentiation induction in vitro. J Transl Med 2009; 7:6. [PMID: 19144182 PMCID: PMC2633316 DOI: 10.1186/1479-5876-7-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Accepted: 01/14/2009] [Indexed: 01/09/2023] Open
Abstract
The dystrophin gene, located at Xp21, codifies dystrophin, which is part of a protein complex responsible for the membrane stability of muscle cells. Its absence on muscle causes Duchenne Muscular Dystrophy (DMD), a severe disorder, while a defect of muscle dystrophin causes Becker Muscular Dystrophy (DMB), a milder disease. The replacement of the defective muscle through stem cells transplantation is a possible future treatment for these patients. Our objective was to analyze the potential of CD34+ stem cells from umbilical cord blood to differentiate in muscle cells and express dystrophin, in vitro. Protein expression was analyzed by Immunofluorescence, Western Blotting (WB) and Reverse Transcriptase – Polymerase Chain Reaction (RT-PCR). CD34+ stem cells and myoblasts from a DMD affected patient started to fuse with muscle cells immediately after co-cultures establishment. Differentiation in mature myotubes was observed after 15 days and dystrophin-positive regions were detected through Immunofluorescence analysis. However, WB or RT-PCR analysis did not detect the presence of normal dystrophin in co-cultures of CD34+ and DMD or DMB affected patients' muscle cells. In contrast, some CD34+ stem cells differentiated in dystrophin producers' muscle cells, what was observed by WB, reinforcing that this progenitor cell has the potential to originate muscle dystrophin in vitro, and not just in vivo like reported before.
Collapse
Affiliation(s)
- Tatiana Jazedje
- Department of Biology, Human Genome Research Center, São Paulo, Brazil.
| | | | | | | | | | | | | |
Collapse
|
24
|
Cassano M, Biressi S, Finan A, Benedetti L, Omes C, Boratto R, Martin F, Allegretti M, Broccoli V, Cusella De Angelis G, Comoglio PM, Basilico C, Torrente Y, Michieli P, Cossu G, Sampaolesi M. Magic-factor 1, a partial agonist of Met, induces muscle hypertrophy by protecting myogenic progenitors from apoptosis. PLoS One 2008; 3:e3223. [PMID: 18795097 PMCID: PMC2528937 DOI: 10.1371/journal.pone.0003223] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2008] [Accepted: 08/20/2008] [Indexed: 01/18/2023] Open
Abstract
Background Hepatocyte Growth Factor (HGF) is a pleiotropic cytokine of mesenchymal origin that mediates a characteristic array of biological activities including cell proliferation, survival, motility and morphogenesis. Its high affinity receptor, the tyrosine kinase Met, is expressed by a wide range of tissues and can be activated by either paracrine or autocrine stimulation. Adult myogenic precursor cells, the so called satellite cells, express both HGF and Met. Following muscle injury, autocrine HGF-Met stimulation plays a key role in promoting activation and early division of satellite cells, but is shut off in a second phase to allow myogenic differentiation. In culture, HGF stimulation promotes proliferation of muscle precursors thereby inhibiting their differentiation. Methodology/Principal Findings Magic-Factor 1 (Met-Activating Genetically Improved Chimeric Factor-1 or Magic-F1) is an HGF-derived, engineered protein that contains two Met-binding domains repeated in tandem. It has a reduced affinity for Met and, in contrast to HGF it elicits activation of the AKT but not the ERK signaling pathway. As a result, Magic-F1 is not mitogenic but conserves the ability to promote cell survival. Here we show that Magic-F1 protects myogenic precursors against apoptosis, thus increasing their fusion ability and enhancing muscular differentiation. Electrotransfer of Magic-F1 gene into adult mice promoted muscular hypertrophy and decreased myocyte apoptosis. Magic-F1 transgenic mice displayed constitutive muscular hypertrophy, improved running performance and accelerated muscle regeneration following injury. Crossing of Magic-F1 transgenic mice with α-sarcoglycan knock-out mice –a mouse model of muscular dystrophy– or adenovirus-mediated Magic-F1 gene delivery resulted in amelioration of the dystrophic phenotype as measured by both anatomical/histological analysis and functional tests. Conclusions/Significance Because of these features Magic-F1 represents a novel molecular tool to counteract muscle wasting in major muscular diseases such as cachexia or muscular dystrophy.
Collapse
Affiliation(s)
- Marco Cassano
- Translational Cardiomyology, Stem Cell Institute Leuven (SCIL), KULeuven, Leuven, Belgium
- Department of Histology and Medical Embryology, University of Rome Sapienza, Rome, Italy
| | | | - Amanda Finan
- Stem Cell Research Institute, H. S. Raffaele, Milan, Italy
| | | | - Claudia Omes
- Human Anatomy, University of Pavia, Pavia, Italy
| | | | | | | | - Vania Broccoli
- Stem Cell Research Institute, H. S. Raffaele, Milan, Italy
| | | | - Paolo M. Comoglio
- Division of Molecular Oncology, Institute for Cancer Research and Treatment (IRCC), Candiolo (Torino), Italy
| | - Cristina Basilico
- Division of Molecular Oncology, Institute for Cancer Research and Treatment (IRCC), Candiolo (Torino), Italy
| | - Yvan Torrente
- Department of Neurological Science, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Paolo Michieli
- Division of Molecular Oncology, Institute for Cancer Research and Treatment (IRCC), Candiolo (Torino), Italy
| | - Giulio Cossu
- Stem Cell Research Institute, H. S. Raffaele, Milan, Italy
| | - Maurilio Sampaolesi
- Translational Cardiomyology, Stem Cell Institute Leuven (SCIL), KULeuven, Leuven, Belgium
- Human Anatomy, University of Pavia, Pavia, Italy
- * E-mail:
| |
Collapse
|
25
|
Bellinger AM, Mongillo M, Marks AR. Stressed out: the skeletal muscle ryanodine receptor as a target of stress. J Clin Invest 2008; 118:445-53. [PMID: 18246195 DOI: 10.1172/jci34006] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Over the past century, understanding the mechanisms underlying muscle fatigue and weakness has been the focus of much investigation. However, the dominant theory in the field, that lactic acidosis causes muscle fatigue, is unlikely to tell the whole story. Recently, dysregulation of sarcoplasmic reticulum (SR) Ca(2+) release has been associated with impaired muscle function induced by a wide range of stressors, from dystrophy to heart failure to muscle fatigue. Here, we address current understandings of the altered regulation of SR Ca(2+) release during chronic stress, focusing on the role of the SR Ca(2+) release channel known as the type 1 ryanodine receptor.
Collapse
Affiliation(s)
- Andrew M Bellinger
- Clyde and Helen Wu Center for Molecular Cardiology, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA
| | | | | |
Collapse
|
26
|
Kazuki Y, Hoshiya H, Kai Y, Abe S, Takiguchi M, Osaki M, Kawazoe S, Katoh M, Kanatsu-Shinohara M, Inoue K, Kajitani N, Yoshino T, Shirayoshi Y, Ogura A, Shinohara T, Barrett JC, Oshimura M. Correction of a genetic defect in multipotent germline stem cells using a human artificial chromosome. Gene Ther 2008; 15:617-24. [PMID: 18305578 DOI: 10.1038/sj.gt.3303091] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Human artificial chromosomes (HACs) have several advantages as gene therapy vectors, including stable episomal maintenance that avoids insertional mutations and the ability to carry large gene inserts including regulatory elements. Multipotent germline stem (mGS) cells have a great potential for gene therapy because they can be generated from an individual's testes, and when reintroduced can contribute to the specialized function of any tissue. As a proof of concept, we herein report the functional restoration of a genetic deficiency in mouse p53-/- mGS cells, using a HAC with a genomic human p53 gene introduced via microcell-mediated chromosome transfer. The p53 phenotypes of gene regulation and radiation sensitivity were complemented by introducing the p53-HAC and the cells differentiated into several different tissue types in vivo and in vitro. Therefore, the combination of using mGS cells with HACs provides a new tool for gene and cell therapies. The next step is to demonstrate functional restoration using animal models for future gene therapy.
Collapse
Affiliation(s)
- Y Kazuki
- Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science, Tottori University, Yonago, Tottori, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Cossu G, Sampaolesi M. New therapies for Duchenne muscular dystrophy: challenges, prospects and clinical trials. Trends Mol Med 2007; 13:520-6. [PMID: 17983835 DOI: 10.1016/j.molmed.2007.10.003] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2007] [Revised: 10/10/2007] [Accepted: 10/10/2007] [Indexed: 02/05/2023]
Abstract
Muscular dystrophies primarily affect skeletal muscle. Mutations in a large number of genes, mainly encoding cytoskeletal proteins, cause different forms of dystrophy that compromise patient mobility and quality of life, and in the most severe cases lead to complete paralysis and premature death. Although muscular dystrophies still lack an effective therapy, several novel strategies are entering or are ready to enter clinical trials. Here we review the main experimental strategies, namely drug, gene and cell therapies, outlining their goals and limitations. We also provide an update of ongoing or planned clinical trials based on these strategies.
Collapse
Affiliation(s)
- Giulio Cossu
- Stem Cell Research Institute, Dibit, H. San Raffaele, 58 Via Olgettina, 20132 Milan, Italy.
| | | |
Collapse
|
28
|
Onori A, Desantis A, Buontempo S, Di Certo MG, Fanciulli M, Salvatori L, Passananti C, Corbi N. The artificial 4-zinc-finger protein Bagly binds human utrophin promoter A at the endogenous chromosomal site and activates transcription. Biochem Cell Biol 2007; 85:358-65. [PMID: 17612630 DOI: 10.1139/o07-015] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Our aim is to upregulate the expression of the dystrophin-related gene utrophin in Duchenne muscular dystrophy, in this way complementing the lack of dystrophin function. To achieve utrophin upregulation, we designed and engineered synthetic zinc-finger based transcription factors. We have previously shown that the artificial 3-zinc-finger protein Jazz, fused with the appropriate effector domain, is able to drive the transcription of a test gene from utrophin promoter A. Here we report a novel artificial 4-zinc-finger protein, Bagly, which binds with optimized affinity-specificity to a 12 bp DNA target sequence that is internal to human utrophin promoter A. Bagly was generated adding to Jazz protein an extra-fourth zinc finger, derived from transcription factor YY1. Importantly, the Bagly DNA target sequence is statistically present in the human genome only 210 times, about 60 fewer times than the 9 bp Jazz DNA target sequence. Thanks to its additional zinc-finger domain, Bagly protein shows enhanced transcriptional activity. Moreover, we demonstrated Bagly's effective access and binding to active chromatin in the chromosomal context and its ability to upregulate endogenous utrophin.
Collapse
Affiliation(s)
- Annalisa Onori
- Istituto di Biologia e Patologia Molecolari, CNR, c/o Regina Elena Cancer Institute, Via delle Messi d'Oro 156, 00158 Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Mattei E, Corbi N, Di Certo MG, Strimpakos G, Severini C, Onori A, Desantis A, Libri V, Buontempo S, Floridi A, Fanciulli M, Baban D, Davies KE, Passananti C. Utrophin up-regulation by an artificial transcription factor in transgenic mice. PLoS One 2007; 2:e774. [PMID: 17712422 PMCID: PMC1942121 DOI: 10.1371/journal.pone.0000774] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2007] [Accepted: 07/23/2007] [Indexed: 12/27/2022] Open
Abstract
Duchenne Muscular Dystrophy (DMD) is a severe muscle degenerative disease, due to absence of dystrophin. There is currently no effective treatment for DMD. Our aim is to up-regulate the expression level of the dystrophin related gene utrophin in DMD, complementing in this way the lack of dystrophin functions. To this end we designed and engineered several synthetic zinc finger based transcription factors. In particular, we have previously shown that the artificial three zinc finger protein named Jazz, fused with the appropriate effector domain, is able to drive the transcription of a test gene from the utrophin promoter “A”. Here we report on the characterization of Vp16-Jazz-transgenic mice that specifically over-express the utrophin gene at the muscular level. A Chromatin Immunoprecipitation assay (ChIP) demonstrated the effective access/binding of the Jazz protein to active chromatin in mouse muscle and Vp16-Jazz was shown to be able to up-regulate endogenous utrophin gene expression by immunohistochemistry, western blot analyses and real-time PCR. To our knowledge, this is the first example of a transgenic mouse expressing an artificial gene coding for a zinc finger based transcription factor. The achievement of Vp16-Jazz transgenic mice validates the strategy of transcriptional targeting of endogenous genes and could represent an exclusive animal model for use in drug discovery and therapeutics.
Collapse
Affiliation(s)
- Elisabetta Mattei
- Istituto di Neurobiologia e Medicina Molecolare, Consiglio Nazionale delle Ricerche, European Brain Research Institute, Rome, Italy
- Italian Association for Cancer Research, Roman Oncogenomic Center, Rome, Italy
| | - Nicoletta Corbi
- Istituto di Biologia e Patologia Molecolari, Consiglio Nazionale delle Ricerche, Regina Elena Cancer Institute, Rome, Italy
| | - Maria Grazia Di Certo
- Istituto di Neurobiologia e Medicina Molecolare, Consiglio Nazionale delle Ricerche, European Brain Research Institute, Rome, Italy
- Department of Experimental Medicine, University of L'Aquila, L'Aquila, Italy
| | - Georgios Strimpakos
- Istituto di Neurobiologia e Medicina Molecolare, Consiglio Nazionale delle Ricerche, European Brain Research Institute, Rome, Italy
| | - Cinzia Severini
- Istituto di Neurobiologia e Medicina Molecolare, Consiglio Nazionale delle Ricerche, European Brain Research Institute, Rome, Italy
| | - Annalisa Onori
- Istituto di Biologia e Patologia Molecolari, Consiglio Nazionale delle Ricerche, Regina Elena Cancer Institute, Rome, Italy
| | - Agata Desantis
- Istituto di Biologia e Patologia Molecolari, Consiglio Nazionale delle Ricerche, Regina Elena Cancer Institute, Rome, Italy
- Department of Experimental Medicine, University of L'Aquila, L'Aquila, Italy
| | - Valentina Libri
- Istituto di Biologia e Patologia Molecolari, Consiglio Nazionale delle Ricerche, Regina Elena Cancer Institute, Rome, Italy
| | - Serena Buontempo
- Istituto di Biologia e Patologia Molecolari, Consiglio Nazionale delle Ricerche, Regina Elena Cancer Institute, Rome, Italy
| | - Aristide Floridi
- Department of Experimental Medicine, University of L'Aquila, L'Aquila, Italy
- Laboratory B, Regina Elena Cancer Institute, Rome, Italy
| | - Maurizio Fanciulli
- Italian Association for Cancer Research, Roman Oncogenomic Center, Rome, Italy
- Laboratory B, Regina Elena Cancer Institute, Rome, Italy
| | - Dilair Baban
- Department of Physiology, Anatomy and Genetics, Medical Research Council Functional Genetics Unit, University of Oxford, Oxford, United Kingdom
| | - Kay E. Davies
- Department of Physiology, Anatomy and Genetics, Medical Research Council Functional Genetics Unit, University of Oxford, Oxford, United Kingdom
| | - Claudio Passananti
- Italian Association for Cancer Research, Roman Oncogenomic Center, Rome, Italy
- Istituto di Biologia e Patologia Molecolari, Consiglio Nazionale delle Ricerche, Regina Elena Cancer Institute, Rome, Italy
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
30
|
Hinkle RT, Lefever FR, Dolan ET, Reichart DL, Dietrich JA, Gropp KE, Thacker RI, Demuth JP, Stevens PJ, Qu XA, Varbanov AR, Wang F, Isfort RJ. Corticortophin releasing factor 2 receptor agonist treatment significantly slows disease progression in mdx mice. BMC Med 2007; 5:18. [PMID: 17626629 PMCID: PMC1936998 DOI: 10.1186/1741-7015-5-18] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2006] [Accepted: 07/12/2007] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Duchenne muscular dystrophy results from mutation of the dystrophin gene, causing skeletal and cardiac muscle loss of function. The mdx mouse model of Duchenne muscular dystrophy is widely utilized to evaluate the potential of therapeutic regimens to modulate the loss of skeletal muscle function associated with dystrophin mutation. Importantly, progressive loss of diaphragm function is the most consistent striated muscle effect observed in the mdx mouse model, which is the same as in patients suffering from Duchenne muscular dystrophy. METHODS Using the mdx mouse model, we have evaluated the effect that corticotrophin releasing factor 2 receptor (CRF2R) agonist treatment has on diaphragm function, morphology and gene expression. RESULTS We have observed that treatment with the potent CRF2R-selective agonist PG-873637 prevents the progressive loss of diaphragm specific force observed during aging of mdx mice. In addition, the combination of PG-873637 with glucocorticoids not only prevents the loss of diaphragm specific force over time, but also results in recovery of specific force. Pathological analysis of CRF2R agonist-treated diaphragm muscle demonstrates that treatment reduces fibrosis, immune cell infiltration, and muscle architectural disruption. Gene expression analysis of CRF2R-treated diaphragm muscle showed multiple gene expression changes including globally decreased immune cell-related gene expression, decreased extracellular matrix gene expression, increased metabolism-related gene expression, and, surprisingly, modulation of circadian rhythm gene expression. CONCLUSION Together, these data demonstrate that CRF2R activation can prevent the progressive degeneration of diaphragm muscle associated with dystrophin gene mutation.
Collapse
Affiliation(s)
- Richard T Hinkle
- Research Division, Procter & Gamble Pharmaceuticals, Mason, OH, USA
| | - Frank R Lefever
- Research Division, Procter & Gamble Pharmaceuticals, Mason, OH, USA
| | | | | | | | - Kathryn E Gropp
- Research Division, Procter & Gamble Pharmaceuticals, Mason, OH, USA
| | - Robert I Thacker
- Department of Pathobiology and Molecular Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Jeffrey P Demuth
- Research Division, Procter & Gamble Pharmaceuticals, Mason, OH, USA
| | - Paula J Stevens
- Research Division, Procter & Gamble Pharmaceuticals, Mason, OH, USA
| | - Xiaoyan A Qu
- Research Division, Procter & Gamble Pharmaceuticals, Mason, OH, USA
| | - Alex R Varbanov
- Research Division, Procter & Gamble Pharmaceuticals, Mason, OH, USA
| | - Feng Wang
- Research Division, Procter & Gamble Pharmaceuticals, Mason, OH, USA
| | - Robert J Isfort
- Research Division, Procter & Gamble Pharmaceuticals, Mason, OH, USA
| |
Collapse
|
31
|
Donati C, Cencetti F, Nincheri P, Bernacchioni C, Brunelli S, Clementi E, Cossu G, Bruni P. Sphingosine 1-Phosphate Mediates Proliferation and Survival of Mesoangioblasts. Stem Cells 2007; 25:1713-9. [PMID: 17464089 DOI: 10.1634/stemcells.2006-0725] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mesoangioblasts are stem cells capable of differentiating in various mesodermal tissues and are presently regarded as suitable candidates for cell therapy of muscle degenerative diseases, as well as myocardial infarction. The enhancement of their proliferation and survival after injection in vivo could greatly improve their ability to repopulate damaged tissues. In this study, we show that the bioactive sphingolipid sphingosine 1-phosphate (S1P) regulates critical functions of mesoangioblast cell biology. S1P evoked a full mitogenic response in mesoangioblasts, measured by labeled thymidine incorporation and cell counting. Moreover, S1P strongly counteracted the apoptotic process triggered by stimuli as diverse as serum deprivation, C2-ceramide treatment, or staurosporine treatment, as assessed by cell counting, as well as histone-associated fragments and caspase-3 activity determinations. S1P acts both as an intracellular messenger and through specific membrane receptors. Real-time polymerase chain reaction analysis revealed that mesoangioblasts express the S1P-specific receptor S1P3 and, to a minor extent, S1P1 and S1P2. By using S1P receptor subtype-specific agonists and antagonists, we found that the proliferative response to S1P was mediated mainly by S1P2. By contrast, the antiapoptotic effect did not implicate S1P receptors. These findings demonstrate an important role of S1P in mesoangioblast proliferation and survival and indicate that targeting modulation of S1P-dependent signaling pathways may be used to improve the efficiency of muscle repair by these cells. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Chiara Donati
- Dipartimento di Scienze Biochimiche and Instituto Interuniversitario di Miotogia, Università di Firenze, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Nunes VA, Cavaçana N, Canovas M, Strauss BE, Zatz M. Stem cells from umbilical cord blood differentiate into myotubes and express dystrophin in vitro only after exposure to in vivo muscle environment. Biol Cell 2007; 99:185-96. [PMID: 17166095 DOI: 10.1042/bc20060075] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND INFORMATION Duchenne muscular dystrophy is a disease characterized by progressive and irreversible muscle degeneration for which there is no therapy. HUCB (human umbilical cord blood) has been considered as an important source of haematopoietic and mesenchymal stem cells, each having been shown to differentiate into distinct cell types. However, it remains unclear if these cells are able to differentiate into muscle cells. RESULTS We have showed that stem cells from HUCB did not differentiate into myotubes or express dystrophin when cultured in muscle-conditioned medium or with human muscle cells. However, delivery of GFP (green fluorescent protein)-transduced mononucleated cells from HUCB, which comprises both haematopoietic and mesenchymal populations, into quadriceps muscle of mdx (mouse dystrophy X-chromosome linked) mice resulted in the expression of human myogenic markers. After recovery of these cells from mdx muscle and in vitro cultivation, they were able to fuse and form GFP-positive myotubes that expressed dystrophin. CONCLUSIONS These results indicate that chemical factors and cell-to-cell contact provided by in vitro conditions were not enough to trigger the differentiation of stem cells into muscle cells. Nevertheless, we showed that the HUCB-derived stem cells were capable of acquiring a muscle phenotype after exposure to an in vivo muscle environment, which was required to activate the differentiation programme.
Collapse
Affiliation(s)
- Viviane A Nunes
- Human Genome Research Center, Department of Genetics and Evolutionary Biology, University of Sao Paulo, Sao Paulo, SP, Brazil.
| | | | | | | | | |
Collapse
|
33
|
Sciorati C, Galvez BG, Brunelli S, Tagliafico E, Ferrari S, Cossu G, Clementi E. Ex vivo treatment with nitric oxide increases mesoangioblast therapeutic efficacy in muscular dystrophy. J Cell Sci 2007; 119:5114-23. [PMID: 17158915 DOI: 10.1242/jcs.03300] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Muscular dystrophies are characterized by primary wasting of skeletal muscle for which no satisfactory therapy is available. Studies in animal models have shown that stem cell-based therapies may improve the outcome of the disease, and that mesoangioblasts are promising stem cells in this respect. The efficacy of mesoangioblasts in yielding extensive muscle repair is, however, still limited. We found that mesoangioblasts treated with nitric oxide (NO) donors and injected intra-arterially in alpha-sarcoglycan-null dystrophic mice have a significantly enhanced ability to migrate to dystrophic muscles, to resist their apoptogenic environment and engraft into them, yielding a significant recovery of alpha-sarcolgycan expression. In vitro NO-treated mesoangioblasts displayed an enhanced chemotactic response to myotubes, cytokines and growth factors generated by the dystrophic muscle. In addition, they displayed an increased ability to fuse with myotubes and differentiating myoblasts and to survive when exposed to cytotoxic stimuli similar to those present in the dystrophic muscle. All the effects of NO were cyclic GMP-dependent since they were mimicked by treatment with the membrane permeant cyclic-GMP analogue 8-bromo-cGMP and prevented by inhibiting guanylate cyclase. We conclude that NO donors exert multiple beneficial effects on mesoangioblasts that may be used to increase their efficacy in cell therapy of muscular dystrophies.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Cell Differentiation/drug effects
- Cell Movement/drug effects
- Cells, Cultured
- Cyclic GMP/analogs & derivatives
- Cyclic GMP/pharmacology
- Gene Expression Profiling
- Immunohistochemistry
- Mesoderm/cytology
- Mesoderm/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Muscular Dystrophy, Animal/genetics
- Muscular Dystrophy, Animal/therapy
- Nitric Oxide Donors/pharmacology
- Sarcoglycans/deficiency
- Sarcoglycans/genetics
- Stem Cell Transplantation
- Stem Cells/cytology
- Stem Cells/drug effects
- Stem Cells/metabolism
Collapse
Affiliation(s)
- Clara Sciorati
- Stem Cell Research Institute, H San Raffaele Scientific Institute, 20132, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
34
|
Term Amniotic membrane is a high throughput source for multipotent Mesenchymal Stem Cells with the ability to differentiate into endothelial cells in vitro. BMC DEVELOPMENTAL BIOLOGY 2007; 7:11. [PMID: 17313666 PMCID: PMC1810523 DOI: 10.1186/1471-213x-7-11] [Citation(s) in RCA: 290] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2006] [Accepted: 02/21/2007] [Indexed: 11/10/2022]
Abstract
BACKGROUND Term Amniotic membrane (AM) is a very attractive source of Mesenchymal Stem Cells (MSCs) due to the fact that this fetal tissue is usually discarded without ethical conflicts, leading to high efficiency in MSC recovery with no intrusive procedures. Here we confirmed that term AM, as previously reported in the literature, is an abundant source of hMSCs; in particular we further investigated the AM differentiation potential by assessing whether these cells may also be committed to the angiogenic fate. In agreement with the recommendation of the International Society for Cellular Therapy, the mesenchymal cells herein investigated were named Amniotic Membrane-human Mesenchymal Stromal Cells (AM-hMSC). RESULTS The recovery of hMSCs and their in vitro expansion potential were greater in amniotic membrane than in bone marrow stroma. At flow cytometry analysis AM-hMSCs showed an immunophenotypical profile, i.e., positive for CD105, CD73, CD29, CD44, CD166 and negative for CD14, CD34, CD45, consistent with that reported for bone marrow-derived MSCs. In addition, amniotic membrane-isolated cells underwent in vitro osteogenic (von Kossa stain), adipogenic (Oil Red-O stain), chondrogenic (collagen type II immunohistochemichal detection) and myogenic (RT-PCR MyoD and Myogenin expression as well as desmin immunohistochemical detection) differentiation. In angiogenic experiments, a spontaneous differentiation into endothelial cells was detected by in vitro matrigel assay and this behaviour has been enhanced through Vascular Endothelial Growth Factor (VEGF) induction. According to these findings, VEGF receptor 1 and 2 (FLT-1 and KDR) were basally expressed in AM-hMSCs and the expression of endothelial-specific markers like FLT-1 KDR, ICAM-1 increased after exposure to VEGF together with the occurrence of CD34 and von Willebrand Factor positive cells. CONCLUSION The current study suggests that AM-hMSCs may emerge as a remarkable tool for the cell therapy of multiple diseased tissues. AM-hMSCs may potentially assist both bone and cartilage repair, nevertheless, due to their angiogenic potential, they may also pave the way for novel approaches in the development of tissue-engineered vascular grafts which are useful when vascularization of ischemic tissues is required.
Collapse
|
35
|
Dellavalle A, Sampaolesi M, Tonlorenzi R, Tagliafico E, Sacchetti B, Perani L, Innocenzi A, Galvez BG, Messina G, Morosetti R, Li S, Belicchi M, Peretti G, Chamberlain JS, Wright WE, Torrente Y, Ferrari S, Bianco P, Cossu G. Pericytes of human skeletal muscle are myogenic precursors distinct from satellite cells. Nat Cell Biol 2007; 9:255-67. [PMID: 17293855 DOI: 10.1038/ncb1542] [Citation(s) in RCA: 700] [Impact Index Per Article: 38.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2006] [Accepted: 01/30/2007] [Indexed: 02/08/2023]
Abstract
Cells derived from blood vessels of human skeletal muscle can regenerate skeletal muscle, similarly to embryonic mesoangioblasts. However, adult cells do not express endothelial markers, but instead express markers of pericytes, such as NG2 proteoglycan and alkaline phosphatase (ALP), and can be prospectively isolated from freshly dissociated ALP(+) cells. Unlike canonical myogenic precursors (satellite cells), pericyte-derived cells express myogenic markers only in differentiated myotubes, which they form spontaneously with high efficiency. When transplanted into severe combined immune deficient-X-linked, mouse muscular dystrophy (scid-mdx) mice, pericyte-derived cells colonize host muscle and generate numerous fibres expressing human dystrophin. Similar cells isolated from Duchenne patients, and engineered to express human mini-dystrophin, also give rise to many dystrophin-positive fibres in vivo. These data show that myogenic precursors, distinct from satellite cells, are associated with microvascular walls in the human skeletal muscle, may represent a correlate of embryonic 'mesoangioblasts' present after birth and may be a promising candidate for future cell-therapy protocols in patients.
Collapse
MESH Headings
- Adolescent
- Adult
- Adult Stem Cells/cytology
- Adult Stem Cells/metabolism
- Adult Stem Cells/transplantation
- Aged
- Animals
- Antigens, CD/analysis
- Cell Culture Techniques/methods
- Child
- Child, Preschool
- Female
- Humans
- Male
- Mice
- Mice, Inbred mdx
- Mice, Nude
- Mice, SCID
- Middle Aged
- Muscle Proteins/analysis
- Muscle Proteins/genetics
- Muscle, Skeletal/chemistry
- Muscle, Skeletal/cytology
- Muscle, Skeletal/physiology
- Muscular Dystrophy, Duchenne/physiopathology
- Muscular Dystrophy, Duchenne/surgery
- Pericytes/chemistry
- Pericytes/cytology
- Pericytes/transplantation
- Regeneration/physiology
- Satellite Cells, Skeletal Muscle/cytology
- Satellite Cells, Skeletal Muscle/metabolism
- Satellite Cells, Skeletal Muscle/transplantation
- Stem Cell Transplantation/methods
- Treatment Outcome
Collapse
Affiliation(s)
- Arianna Dellavalle
- Stem Cell Research Institute, San Raffaele Scientific Institute, 58 Via Olgettina, 20132 Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Brunelli S, Sciorati C, D'Antona G, Innocenzi A, Covarello D, Galvez BG, Perrotta C, Monopoli A, Sanvito F, Bottinelli R, Ongini E, Cossu G, Clementi E. Nitric oxide release combined with nonsteroidal antiinflammatory activity prevents muscular dystrophy pathology and enhances stem cell therapy. Proc Natl Acad Sci U S A 2006; 104:264-9. [PMID: 17182743 PMCID: PMC1765447 DOI: 10.1073/pnas.0608277104] [Citation(s) in RCA: 138] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Duchenne muscular dystrophy is a relatively common disease that affects skeletal muscle, leading to progressive paralysis and death. There is currently no resolutive therapy. We have developed a treatment in which we combined the effects of nitric oxide with nonsteroidal antiinflammatory activity by using HCT 1026, a nitric oxide-releasing derivative of flurbiprofen. Here, we report the results of long-term (1-year) oral treatment with HCT 1026 of two murine models for limb girdle and Duchenne muscular dystrophies (alpha-sarcoglycan-null and mdx mice). In both models, HCT 1026 significantly ameliorated the morphological, biochemical, and functional phenotype in the absence of secondary effects, efficiently slowing down disease progression. HCT 1026 acted by reducing inflammation, preventing muscle damage, and preserving the number and function of satellite cells. HCT 1026 was significantly more effective than the corticosteroid prednisolone, which was analyzed in parallel. As an additional beneficial effect, HCT 1026 enhanced the therapeutic efficacy of arterially delivered donor stem cells, by increasing 4-fold their ability to migrate and reconstitute muscle fibers. The therapeutic strategy we propose is not selective for a subset of mutations; it provides ground for immediate clinical experimentation with HCT 1026 alone, which is approved for use in humans; and it sets the stage for combined therapies with donor or autologous, genetically corrected stem cells.
Collapse
Affiliation(s)
- Silvia Brunelli
- *Department of Experimental Medicine, University of Milano–Bicocca, 20052 Monza, Italy
- San Raffaele Scientific Institute, Stem Cell Research Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Clara Sciorati
- San Raffaele Scientific Institute, Stem Cell Research Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Giuseppe D'Antona
- Department of Experimental Medicine, University of Pavia, 27100 Pavia, Italy
| | - Anna Innocenzi
- San Raffaele Scientific Institute, Stem Cell Research Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Diego Covarello
- San Raffaele Scientific Institute, Stem Cell Research Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Beatriz G. Galvez
- San Raffaele Scientific Institute, Stem Cell Research Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Cristiana Perrotta
- San Raffaele Scientific Institute, Stem Cell Research Institute, Via Olgettina 58, 20132 Milan, Italy
- Department of Preclinical Sciences, University of Milano, 20157 Milan, Italy
| | - Angela Monopoli
- Nicox Research Institute, Via Ariosto 21, 20091 Bresso, Italy
| | - Francesca Sanvito
- San Raffaele Scientific Institute, Stem Cell Research Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Roberto Bottinelli
- Department of Experimental Medicine, University of Pavia, 27100 Pavia, Italy
| | - Ennio Ongini
- Nicox Research Institute, Via Ariosto 21, 20091 Bresso, Italy
| | - Giulio Cossu
- San Raffaele Scientific Institute, Stem Cell Research Institute, Via Olgettina 58, 20132 Milan, Italy
- Department of Biology, University of Milano, 20130 Milan, Italy
- **To whom correspondence may be addressed at:
Stem Cell Research Institute, H. San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy. E-mail:
or
| | - Emilio Clementi
- San Raffaele Scientific Institute, Stem Cell Research Institute, Via Olgettina 58, 20132 Milan, Italy
- E. Medea Scientific Institute, 23842 Bosisio Parini, Italy; and
- Department of Preclinical Sciences, University of Milano, 20157 Milan, Italy
- **To whom correspondence may be addressed at:
Stem Cell Research Institute, H. San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy. E-mail:
or
| |
Collapse
|
37
|
Brunelli S, Rovere-Querini P, Sciorati C, Manfredi AA, Clementi E. Nitric oxide: emerging concepts about its use in cell-based therapies. Expert Opin Investig Drugs 2006; 16:33-43. [PMID: 17155852 DOI: 10.1517/13543784.16.1.33] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Regenerative medicine is an emerging clinical discipline in which cell-based therapies are used to restore the functions of damaged or defective tissues and organs. Along with the well-established use of cells derived from bone marrow or pancreatic islets, novel approaches of cell therapy have recently emerged that appear particularly promising; that is, those using cell-based vaccines and stem cells. This review focuses on the recent developments of these experimental therapeutic approaches and their drawbacks, with specific focus on dendritic cell vaccines in tumours and mesoangioblasts in muscular dystrophies. The authors discuss how the unique properties of a gaseous messenger, NO, may be exploited to overcome some of the drawbacks of these cell-based approaches in combined therapies based on NO-releasing drugs and cell delivery.
Collapse
Affiliation(s)
- Silvia Brunelli
- University of Milano-Bicocca, Department of Experimental, Environmental Medicine and Medical Biotechnology, 20052 Monza, Italy
| | | | | | | | | |
Collapse
|
38
|
Sampaolesi M, Blot S, D'Antona G, Granger N, Tonlorenzi R, Innocenzi A, Mognol P, Thibaud JL, Galvez BG, Barthélémy I, Perani L, Mantero S, Guttinger M, Pansarasa O, Rinaldi C, Cusella De Angelis MG, Torrente Y, Bordignon C, Bottinelli R, Cossu G. Mesoangioblast stem cells ameliorate muscle function in dystrophic dogs. Nature 2006; 444:574-9. [PMID: 17108972 DOI: 10.1038/nature05282] [Citation(s) in RCA: 528] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2006] [Accepted: 09/26/2006] [Indexed: 12/18/2022]
Abstract
Duchenne muscular dystrophy remains an untreatable genetic disease that severely limits motility and life expectancy in affected children. The only animal model specifically reproducing the alterations in the dystrophin gene and the full spectrum of human pathology is the golden retriever dog model. Affected animals present a single mutation in intron 6, resulting in complete absence of the dystrophin protein, and early and severe muscle degeneration with nearly complete loss of motility and walking ability. Death usually occurs at about 1 year of age as a result of failure of respiratory muscles. Here we report that intra-arterial delivery of wild-type canine mesoangioblasts (vessel-associated stem cells) results in an extensive recovery of dystrophin expression, normal muscle morphology and function (confirmed by measurement of contraction force on single fibres). The outcome is a remarkable clinical amelioration and preservation of active motility. These data qualify mesoangioblasts as candidates for future stem cell therapy for Duchenne patients.
Collapse
Affiliation(s)
- Maurilio Sampaolesi
- San Raffaele Scientific Institute, Università Vita e Salute, Stem Cell Research Institute, Via Olgettina 58, 20132 Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Guttinger M, Tafi E, Battaglia M, Coletta M, Cossu G. Allogeneic mesoangioblasts give rise to alpha-sarcoglycan expressing fibers when transplanted into dystrophic mice. Exp Cell Res 2006; 312:3872-9. [DOI: 10.1016/j.yexcr.2006.08.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2006] [Revised: 08/09/2006] [Accepted: 08/16/2006] [Indexed: 10/24/2022]
|
40
|
Di Rocco G, Iachininoto MG, Tritarelli A, Straino S, Zacheo A, Germani A, Crea F, Capogrossi MC. Myogenic potential of adipose-tissue-derived cells. J Cell Sci 2006; 119:2945-52. [PMID: 16825428 DOI: 10.1242/jcs.03029] [Citation(s) in RCA: 169] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Adipose-tissue-derived mesenchymal stem cells can be directed towards a myogenic phenotype in vitro by the addition of specific inductive media. However, the ability of these or other adipose-tissue-associated cells to respond to ;natural' myogenic cues such as a myogenic environment has never been investigated in detail. Here, we provide evidence that a restricted subpopulation of freshly harvested adipose-tissue-derived cells possesses an intrinsic myogenic potential and can spontaneously differentiate into skeletal muscle. Conversion of adipose-tissue-derived cells to a myogenic phenotype is enhanced by co-culture with primary myoblasts in the absence of cell contact and is maximal when the two cell types are co-cultured in the same plate. Conversely, in vitro expanded adipose-tissue-derived mesenchymal stem cells require direct contact with muscle cells to generate skeletal myotubes. Finally, we show that uncultured adipose-tissue-associated cells have a high regenerative capacity in vivo since they can be incorporated into muscle fibers following ischemia and can restore significantly dystrophin expression in mdx mice.
Collapse
Affiliation(s)
- Giuliana Di Rocco
- Laboratorio di Biologia Vascolare e Terapia Genica, Centro Cardiologico Fondazione Monzino, Via Parea 4, 20138 Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Gross DA, Chappert P, Leboeuf M, Monteilhet V, Van Wittenberghe L, Danos O, Davoust J. Simple conditioning with monospecific CD4+CD25+ regulatory T cells for bone marrow engraftment and tolerance to multiple gene products. Blood 2006; 108:1841-8. [PMID: 16741251 DOI: 10.1182/blood-2006-02-011981] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
A major impediment to gene replacement therapy is immune elimination of genetically modified cells. In principle, this can be dealt with by inducing a strong, specific, and enduring tolerance through engraftment of transgene-modified autologous bone marrow (BM). Because usual myeloablation and/or immunosuppression are risk factors in most pathologies, we assessed the potential of monospecific CD4(+)CD25(+) regulatory T cells (Tregs) to engraft minor-mismatched BM without preconditioning. We found that as few as 5 x 10(4) Tregs directed to the male DBY protein promote the engraftment of foreign male BM into sex-mismatched female hosts, establishing sustained chimerism in all hematopoeitic compartments. We achieved concomitantly strong tolerance to all foreign antigens expressed in the BM, likely occurring through induction of anergy and/or deletion of antidonor T cells. Chimerism was obtained in thymectomized mice too, underlining the major role of peripheral tolerance mechanisms in our system. This allowed us to engraft gene-modified tissues while preserving full immunocompetence to third-party antigens. Our results demonstrate that very few donor-specific Tregs are effective as the sole conditioning to induce mixed molecular chimerism and long-term tolerance to multiple foreign antigens.
Collapse
Affiliation(s)
- David-Alexandre Gross
- Genethon, CNRS UMR 8115, 1bis, rue de l'Internationale, BP 60, 91002 Evry Cedex, France.
| | | | | | | | | | | | | |
Collapse
|
42
|
Young LS, Searle PF, Onion D, Mautner V. Viral gene therapy strategies: from basic science to clinical application. J Pathol 2006; 208:299-318. [PMID: 16362990 DOI: 10.1002/path.1896] [Citation(s) in RCA: 196] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A major impediment to the successful application of gene therapy for the treatment of a range of diseases is not a paucity of therapeutic genes, but the lack of an efficient non-toxic gene delivery system. Having evolved to deliver their genes to target cells, viruses are currently the most effective means of gene delivery and can be manipulated to express therapeutic genes or to replicate specifically in certain cells. Gene therapy is being developed for a range of diseases including inherited monogenic disorders and cardiovascular disease, but it is in the treatment of cancer that this approach has been most evident, resulting in the recent licensing of a gene therapy for the routine treatment of head and neck cancer in China. A variety of virus vectors have been employed to deliver genes to cells to provide either transient (eg adenovirus, vaccinia virus) or permanent (eg retrovirus, adeno-associated virus) transgene expression and each approach has its own advantages and disadvantages. Paramount is the safety of these virus vectors and a greater understanding of the virus-host interaction is key to optimizing the use of these vectors for routine clinical use. Recent developments in the modification of the virus coat allow more targeted approaches and herald the advent of systemic delivery of therapeutic viruses. In the context of cancer, the ability of attenuated viruses to replicate specifically in tumour cells has already yielded some impressive results in clinical trials and bodes well for the future of this approach, particularly when combined with more traditional anti-cancer therapies.
Collapse
Affiliation(s)
- Lawrence S Young
- Cancer Research UK Institute for Cancer Studies, University of Birmingham Medical School, UK.
| | | | | | | |
Collapse
|
43
|
Abstract
Skeletal muscle is the largest organ in the human body, and plays an important role in body movement and metabolism. Skeletal muscle mass is lost in genetic disorders such as muscular dystrophy, muscle wasting and ageing. Chemicals and proteins that restore muscle mass and function are potential drugs that can improve human health and could be used in the clinic. Myostatin is a muscle-specific member of the transforming growth factor (TGF)-beta superfamily that plays an essential role in the negative regulation of muscle growth. Inhibition of myostatin activity is a promising therapeutic method for restoring muscle mass and strength. Potential inhibitors of myostatin include follistatin domain-containing proteins, myostatin propeptide, myostatin antibodies and chemical compounds. These inhibitors could be beneficial for the development of clinical drugs for the treatment of muscular disorders. Bone morphogenetic protein (BMP) plays a significant role in the development of neuromuscular architecture and its proper functions. Modulation of BMP activity could be beneficial for muscle function in muscular disorders. This review will describe the current progress in therapy for muscular disorders, emphasising the importance of myostatin as a drug target.
Collapse
Affiliation(s)
- Kunihiro Tsuchida
- Institute for Comprehensive Medical Science, Division for Therapies against Intractable Diseases, Fujita Health University, Toyoake, Aichi 470-1192, Japan.
| |
Collapse
|
44
|
Gonçalves MAFV, Holkers M, Cudré-Mauroux C, van Nierop GP, Knaän-Shanzer S, van der Velde I, Valerio D, de Vries AAF. Transduction of myogenic cells by retargeted dual high-capacity hybrid viral vectors: robust dystrophin synthesis in duchenne muscular dystrophy muscle cells. Mol Ther 2006; 13:976-86. [PMID: 16443396 DOI: 10.1016/j.ymthe.2005.11.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2005] [Revised: 10/25/2005] [Accepted: 11/13/2005] [Indexed: 11/24/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by mutations in the dystrophin gene (DMD), making it amenable to gene- or cell-based therapies. Another possible treatment entails the combination of both principles by transplantation of autologous myogenic cells after their genetic complementation. This approach requires efficient and stable transduction of these cells with recombinant DMD. Recently, we generated a dual high-capacity (hc) adenovirus (Ad)-adeno-associated virus (AAV) hybrid vector (HV) that can deliver two full-length dystrophin-encoding modules into target cells. We showed that HV transduction of human cells containing AAV Rep proteins leads to the insertion of foreign DNA into the AAVS1 locus. Here, we improved HV entry into muscle cells from DMD patients. After having verified that these cells barely express the coxsackie B virus and Ad receptor (CAR), which constitutes the attachment molecule for Ad serotype 5 (Ad5) fibers, we equipped dual hcAd/AAV HV particles with Ad serotype 50 fiber domains to achieve CAR-independent uptake. These retargeted vectors complemented much more efficiently the genetic defect of dystrophin-defective myoblasts and myotubes than their isogenic counterparts with conventional Ad5 fibers. Importantly, the accumulation of beta-dystroglycan along the membranes of vector-treated DMD myotubes indicated proper assembly of dystrophin-associated glycoprotein complexes.
Collapse
Affiliation(s)
- Manuel A F V Gonçalves
- Gene Therapy Section, Department of Molecular Cell Biology, Leiden University Medical Center, Wassenaarseweg 72, 2333 AL Leiden, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Gonçalves MAFV, de Vries AAF, Holkers M, van de Watering MJM, van der Velde I, van Nierop GP, Valerio D, Knaän-Shanzer S. Human mesenchymal stem cells ectopically expressing full-length dystrophin can complement Duchenne muscular dystrophy myotubes by cell fusion. Hum Mol Genet 2005; 15:213-21. [PMID: 16321987 DOI: 10.1093/hmg/ddi438] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most prevalent inheritable muscle disease. It is caused by mutations in the approximately 2.5-megabase dystrophin (Dys) encoding gene. Therapeutic attempts at DMD have relied on injection of allogeneic Dys-positive myoblasts. The immune rejection of these cells and their limited availability have prompted the search for alternative therapies and sources of myogenic cells. Stem cell-based gene therapy aims to restore tissue function by the transplantation of gene-corrected autologous cells. It depends on (i) the capacity of stem cells to participate in tissue regeneration and (ii) the efficient genetic correction of defective autologous stem cells. We explored the potential of bone marrow-derived human mesenchymal stem cells (hMSCs) genetically modified with the full-length Dys-coding sequence to engage in myogenesis. By tagging hMSCs with enhanced green fluorescent protein (EGFP) or the membrane dye PKH26, we demonstrated that they could participate in myotube formation when cultured together with differentiating human myoblasts. Experiments performed with EGFP-marked hMSCs and DsRed-labeled DMD myoblasts revealed that the EGFP-positive DMD myotubes were also DsRed-positive indicating that hMSCs participate in human myogenesis through cellular fusion. Finally, we showed that hMSCs transduced with a tropism-modified high-capacity hybrid viral vector encoding full-length Dys could complement the genetic defect of DMD myotubes.
Collapse
Affiliation(s)
- Manuel A F V Gonçalves
- Department of Molecular Cell Biology, Leiden University Medical Center, Wassenaarseweg 72, 2333 AL Leiden, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Cossu G, Biressi S. Satellite cells, myoblasts and other occasional myogenic progenitors: Possible origin, phenotypic features and role in muscle regeneration. Semin Cell Dev Biol 2005; 16:623-31. [PMID: 16118057 DOI: 10.1016/j.semcdb.2005.07.003] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In the vertebrate embryo, skeletal muscle originates from somites and is formed in discrete steps by different classes of progenitor cells. After myotome formation, embryonic myoblasts give rise to primary fibers in the embryo, while fetal myoblasts give rise to secondary fibers, initially smaller and surrounding primary fibers. Satellite cells appear underneath the newly formed basal lamina that develops around each fiber, and contribute to post-natal growth and regeneration of muscle fibers. Recently, different types of non somitic stem-progenitor cells have been shown to contribute to muscle regeneration. The origin of these different cell types and their possible lineage relationships with other myogenic cells as well as their possible role in muscle regeneration will be discussed. Finally, possible use of different myogenic cells in experimental protocols of cell therapy will be briefly outlined.
Collapse
Affiliation(s)
- Giulio Cossu
- Stem Cell Research Institute, Dibit, H. San Raffaele, via Olgettina 58, 20132 Milan, Italy.
| | | |
Collapse
|
47
|
Harcourt LJ, Holmes AG, Gregorevic P, Schertzer JD, Stupka N, Plant DR, Lynch GS. Interleukin-15 administration improves diaphragm muscle pathology and function in dystrophic mdx mice. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 166:1131-41. [PMID: 15793293 PMCID: PMC1602390 DOI: 10.1016/s0002-9440(10)62333-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/07/2004] [Indexed: 12/13/2022]
Abstract
Interleukin (IL)-15, a cytokine expressed in skeletal muscle, has been shown to have muscle anabolic effects in vitro and to slow muscle wasting in rats with cancer cachexia. Whether IL-15 has therapeutic potential for diseases such as Duchenne muscular dystrophy (DMD) is unknown. We examined whether IL-15 administration could ameliorate the dystrophic pathology in the diaphragm muscle of the mdx mouse, an animal model for DMD. Four weeks of IL-15 treatment improved diaphragm strength, a highly significant finding because respiratory function is a mortality predictor in DMD. Enhanced diaphragm function was associated with increased muscle fiber cross-sectional area and decreased collagen infiltration. IL-15 administration was not associated with changes in T-cell populations or alterations in specific components of the ubiquitin proteasome pathway. To determine the effects of IL-15 on myofiber regeneration, muscles of IL-15-treated and untreated wild-type mice were injured myotoxically, and their functional recovery was assessed. IL-15 had a mild anabolic effect, increasing fiber cross-sectional area after 2 and 6 days but not after 10 days. Our findings demonstrate that IL-15 administration improves the pathophysiology of dystrophic muscle and highlight a possible therapeutic role for IL-15 in the treatment of neuromuscular disorders especially in which muscle wasting is indicated.
Collapse
Affiliation(s)
- Leah J Harcourt
- Department of Physiology, University of Melbourne, Victoria, Australia, 3010
| | | | | | | | | | | | | |
Collapse
|
48
|
Cossu G. Fusion of bone marrow-derived stem cells with striated muscle may not be sufficient to activate muscle genes. J Clin Invest 2005; 114:1540-3. [PMID: 15578085 PMCID: PMC529291 DOI: 10.1172/jci23733] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Several studies have demonstrated the existence of pluripotent bone marrow-derived stem cells capable of homing to injured cardiac and skeletal muscle; however, there has been little evidence demonstrating the induction of tissue-specific endogenous genes in donor stem cells following engraftment. A new study in this issue reports an intriguing finding that raises additional concerns relating to stem cell plasticity and stem cell therapy in an already heated and controversial field. The study demonstrates that wild-type bone marrow-derived side population stem cells are indeed readily incorporated into both skeletal and cardiac muscle when transplanted into mice that lack delta-sarcoglycan -- a model of cardiomyopathy and muscular dystrophy. However, these cells fail to express sarcoglycan and thus to repair the tissue, which suggests that this stem cell population has limited potential for cardiac and skeletal muscle regeneration.
Collapse
Affiliation(s)
- Giulio Cossu
- Stem Cell Research Institute, DIBIT, Milan, Italy.
| |
Collapse
|