1
|
Salemi S, Prange JA, Baumgartner V, Mohr-Haralampieva D, Eberli D. Adult stem cell sources for skeletal and smooth muscle tissue engineering. Stem Cell Res Ther 2022; 13:156. [PMID: 35410452 PMCID: PMC8996587 DOI: 10.1186/s13287-022-02835-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/24/2022] [Indexed: 12/13/2023] Open
Abstract
INTRODUCTION Tissue engineering is an innovative field with enormous developments in recent years. These advances are not only in the understanding of how stem cells can be isolated, cultured and manipulated but also in their potential for clinical applications. Thus, tissue engineering when applied to skeletal and smooth muscle cells is an area that bears high benefit for patients with muscular diseases or damage. Most of the recent research has been focused on use of adult stem cells. These cells have the ability to rejuvenate and repair damaged tissues and can be derived from different organs and tissue sources. Recently there are several different types of adult stem cells, which have the potential to function as a cell source for tissue engineering of skeletal and smooth muscles. However, to build neo-tissues there are several challenges which have to be addressed, such as the selection of the most suitable stem cell type, isolation techniques, gaining control over its differentiation and proliferation process. CONCLUSION The usage of adult stem cells for muscle engineering applications is promising. Here, we summarize the status of research on the use of adult stem cells for cell transplantation in experimental animals and humans. In particular, the application of skeletal and smooth muscle engineering in pre-clinical and clinical trials will be discussed.
Collapse
Affiliation(s)
- Souzan Salemi
- grid.412004.30000 0004 0478 9977Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, Wagistrasse 21, 4.OG, 8952 Schlieren, Switzerland
| | - Jenny A. Prange
- grid.412004.30000 0004 0478 9977Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, Wagistrasse 21, 4.OG, 8952 Schlieren, Switzerland
| | - Valentin Baumgartner
- grid.412004.30000 0004 0478 9977Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, Wagistrasse 21, 4.OG, 8952 Schlieren, Switzerland
| | - Deana Mohr-Haralampieva
- grid.412004.30000 0004 0478 9977Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, Wagistrasse 21, 4.OG, 8952 Schlieren, Switzerland
| | - Daniel Eberli
- grid.412004.30000 0004 0478 9977Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, Wagistrasse 21, 4.OG, 8952 Schlieren, Switzerland
| |
Collapse
|
2
|
Marcinczyk M, Dunn A, Haas G, Madsen J, Scheidt R, Patel K, Talovic M, Garg K. The Effect of Laminin-111 Hydrogels on Muscle Regeneration in a Murine Model of Injury. Tissue Eng Part A 2019; 25:1001-1012. [PMID: 30426851 PMCID: PMC9839345 DOI: 10.1089/ten.tea.2018.0200] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
IMPACT STATEMENT Extremity injuries make up the most common survivable injuries in vehicular accidents and modern military conflicts. A majority of these injuries involve volumetric muscle loss (VML). The potential for donor site morbidity may limit the clinical use of autologous muscle grafts for VML injuries. Treatments that can improve the regeneration of functional muscle tissue are critically needed to improve limb salvage and reduce the rate of delayed amputations. The development of a laminin-111-enriched fibrin hydrogel will offer a potentially transformative and "off-the-shelf" clinically relevant therapy for functional skeletal muscle regeneration.
Collapse
Affiliation(s)
- Madison Marcinczyk
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri
| | - Andrew Dunn
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri
| | - Gabriel Haas
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri
| | - Josh Madsen
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri
| | - Robert Scheidt
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri
| | - Krishna Patel
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri
| | - Muhamed Talovic
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri
| | - Koyal Garg
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, St. Louis, Missouri.,Address correspondence to: Koyal Garg, PhD, Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, 3507 Lindell Boulevard, St. Louis, MO 63103
| |
Collapse
|
3
|
Dunn A, Talovic M, Patel K, Patel A, Marcinczyk M, Garg K. Biomaterial and stem cell-based strategies for skeletal muscle regeneration. J Orthop Res 2019; 37:1246-1262. [PMID: 30604468 DOI: 10.1002/jor.24212] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 12/13/2018] [Indexed: 02/04/2023]
Abstract
Adult skeletal muscle can regenerate effectively after mild physical or chemical insult. Muscle trauma or disease can overwhelm this innate capacity for regeneration and result in heightened inflammation and fibrotic tissue deposition resulting in loss of structure and function. Recent studies have focused on biomaterial and stem cell-based therapies to promote skeletal muscle regeneration following injury and disease. Many stem cell populations besides satellite cells are implicated in muscle regeneration. These stem cells include but are not limited to mesenchymal stem cells, adipose-derived stem cells, hematopoietic stem cells, pericytes, fibroadipogenic progenitors, side population cells, and CD133+ stem cells. However, several challenges associated with their isolation, availability, delivery, survival, engraftment, and differentiation have been reported in recent studies. While acellular scaffolds offer a relatively safe and potentially off-the-shelf solution to cell-based therapies, they are often unable to stimulate host cell migration and activity to a level that would result in clinically meaningful regeneration of traumatized muscle. Combining stem cells and biomaterials may offer a viable therapeutic strategy that may overcome the limitations associated with these therapies when they are used in isolation. In this article, we review the stem cell populations that can stimulate muscle regeneration in vitro and in vivo. We also discuss the regenerative potential of combination therapies that utilize both stem cell and biomaterials for the treatment of skeletal muscle injury and disease. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1246-1262, 2019.
Collapse
Affiliation(s)
- Andrew Dunn
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Muhamed Talovic
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Krishna Patel
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Anjali Patel
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Madison Marcinczyk
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Koyal Garg
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| |
Collapse
|
4
|
Tierney MT, Stec MJ, Rulands S, Simons BD, Sacco A. Muscle Stem Cells Exhibit Distinct Clonal Dynamics in Response to Tissue Repair and Homeostatic Aging. Cell Stem Cell 2018; 22:119-127.e3. [PMID: 29249462 PMCID: PMC5945549 DOI: 10.1016/j.stem.2017.11.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 09/15/2017] [Accepted: 11/07/2017] [Indexed: 10/18/2022]
Abstract
The clonal complexity of adult stem cell pools is progressively lost during homeostatic turnover in several tissues, suggesting a decrease in the number of stem cells with distinct clonal origins. The functional impact of reduced complexity on stem cell pools, and how different tissue microenvironments may contribute to such a reduction, are poorly understood. Here, we performed clonal multicolor lineage tracing of skeletal muscle stem cells (MuSCs) to address these questions. We found that MuSC clonal complexity is maintained during aging despite heterogenous reductions in proliferative capacity, allowing aged muscle to mount a clonally diverse, albeit diminished, response to injury. In contrast, repeated bouts of tissue repair cause a progressive reduction in MuSC clonal complexity indicative of neutral drift. Consistently, biostatistical modeling suggests that MuSCs undergo symmetric expansions with stochastic fate acquisition during tissue repair. These findings establish distinct principles that underlie stem cell dynamics during homeostatic aging and muscle regeneration.
Collapse
Affiliation(s)
- Matthew T Tierney
- Graduate School of Biomedical Sciences, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA; Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Michael J Stec
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Steffen Rulands
- Cavendish Laboratory, Department of Physics, J. J. Thomson Avenue, Cambridge CB3 0HE, UK; The Wellcome Trust/Cancer Research UK Gurdon Institute and Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Benjamin D Simons
- Cavendish Laboratory, Department of Physics, J. J. Thomson Avenue, Cambridge CB3 0HE, UK; The Wellcome Trust/Cancer Research UK Gurdon Institute and Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Alessandra Sacco
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
5
|
Passipieri JA, Baker HB, Siriwardane M, Ellenburg MD, Vadhavkar M, Saul JM, Tomblyn S, Burnett L, Christ GJ. Keratin Hydrogel Enhances In Vivo Skeletal Muscle Function in a Rat Model of Volumetric Muscle Loss. Tissue Eng Part A 2017; 23:556-571. [PMID: 28169594 DOI: 10.1089/ten.tea.2016.0458] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Volumetric muscle loss (VML) injuries exceed the considerable intrinsic regenerative capacity of skeletal muscle, resulting in permanent functional and cosmetic deficits. VML and VML-like injuries occur in military and civilian populations, due to trauma and surgery as well as due to a host of congenital and acquired diseases/syndromes. Current therapeutic options are limited, and new approaches are needed for a more complete functional regeneration of muscle. A potential solution is human hair-derived keratin (KN) biomaterials that may have significant potential for regenerative therapy. The goal of these studies was to evaluate the utility of keratin hydrogel formulations as a cell and/or growth factor delivery vehicle for functional muscle regeneration in a surgically created VML injury in the rat tibialis anterior (TA) muscle. VML injuries were treated with KN hydrogels in the absence and presence of skeletal muscle progenitor cells (MPCs), and/or insulin-like growth factor 1 (IGF-1), and/or basic fibroblast growth factor (bFGF). Controls included VML injuries with no repair (NR), and implantation of bladder acellular matrix (BAM, without cells). Initial studies conducted 8 weeks post-VML injury indicated that application of keratin hydrogels with growth factors (KN, KN+IGF-1, KN+bFGF, and KN+IGF-1+bFGF, n = 8 each) enabled a significantly greater functional recovery than NR (n = 7), BAM (n = 8), or the addition of MPCs to the keratin hydrogel (KN+MPC, KN+MPC+IGF-1, KN+MPC+bFGF, and KN+MPC+IGF-1+bFGF, n = 8 each) (p < 0.05). A second series of studies examined functional recovery for as many as 12 weeks post-VML injury after application of keratin hydrogels in the absence of cells. A significant time-dependent increase in functional recovery of the KN, KN+bFGF, and KN+IGF+bFGF groups was observed, relative to NR and BAM implantation, achieving as much as 90% of the maximum possible functional recovery. Histological findings from harvested tissue at 12 weeks post-VML injury documented significant increases in neo-muscle tissue formation in all keratin treatment groups as well as diminished fibrosis, in comparison to both BAM and NR. In conclusion, keratin hydrogel implantation promoted statistically significant and physiologically relevant improvements in functional outcomes post-VML injury to the rodent TA muscle.
Collapse
Affiliation(s)
- J A Passipieri
- 1 Biomedical Engineering Department, University of Virginia , Charlottesville, Virginia.,2 Wake Forest Institute for Regenerative Medicine, Wake Forest University , Winston-Salem, North Carolina
| | - H B Baker
- 2 Wake Forest Institute for Regenerative Medicine, Wake Forest University , Winston-Salem, North Carolina.,3 Fischell Department of Bioengineering, University of Maryland , College Park, Maryland
| | - Mevan Siriwardane
- 2 Wake Forest Institute for Regenerative Medicine, Wake Forest University , Winston-Salem, North Carolina
| | | | - Manasi Vadhavkar
- 2 Wake Forest Institute for Regenerative Medicine, Wake Forest University , Winston-Salem, North Carolina
| | - Justin M Saul
- 5 Department of Chemical, Paper and Biomedical Engineering, Miami University , Oxford, Ohio
| | - Seth Tomblyn
- 4 KeraNetics, LLC , Winston-Salem, North Carolina
| | - Luke Burnett
- 4 KeraNetics, LLC , Winston-Salem, North Carolina
| | - George J Christ
- 1 Biomedical Engineering Department, University of Virginia , Charlottesville, Virginia.,2 Wake Forest Institute for Regenerative Medicine, Wake Forest University , Winston-Salem, North Carolina.,6 Orthopaedics Department, University of Virginia , Charlottesville, Virginia
| |
Collapse
|
6
|
Tierney MT, Gromova A, Sesillo FB, Sala D, Spenlé C, Orend G, Sacco A. Autonomous Extracellular Matrix Remodeling Controls a Progressive Adaptation in Muscle Stem Cell Regenerative Capacity during Development. Cell Rep 2016; 14:1940-52. [PMID: 26904948 PMCID: PMC4778082 DOI: 10.1016/j.celrep.2016.01.072] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 09/22/2015] [Accepted: 01/24/2016] [Indexed: 12/20/2022] Open
Abstract
Muscle stem cells (MuSCs) exhibit distinct behavior during successive phases of developmental myogenesis. However, how their transition to adulthood is regulated is poorly understood. Here, we show that fetal MuSCs resist progenitor specification and exhibit altered division dynamics, intrinsic features that are progressively lost postnatally. After transplantation, fetal MuSCs expand more efficiently and contribute to muscle repair. Conversely, niche colonization efficiency increases in adulthood, indicating a balance between muscle growth and stem cell pool repopulation. Gene expression profiling identified several extracellular matrix (ECM) molecules preferentially expressed in fetal MuSCs, including tenascin-C, fibronectin, and collagen VI. Loss-of-function experiments confirmed their essential and stagespecific role in regulating MuSC function. Finally, fetal-derived paracrine factors were able to enhance adult MuSC regenerative potential. Together, these findings demonstrate that MuSCs change the way in which they remodel their microenvironment to direct stem cell behavior and support the unique demands of muscle development or repair.
Collapse
Affiliation(s)
- Matthew Timothy Tierney
- Graduate School of Biomedical Sciences, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA; Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Anastasia Gromova
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA; Biomedical Sciences Graduate Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0685, USA
| | - Francesca Boscolo Sesillo
- Graduate School of Biomedical Sciences, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA; Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - David Sala
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Caroline Spenlé
- Inserm U1109, MN3T Team, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, 3 Avenue Molière, 67200 Strasbourg, France; Université de Strasbourg, 67000 Strasbourg, France; LabEx Medalis, Université de Strasbourg, 67000 Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France
| | - Gertraud Orend
- Inserm U1109, MN3T Team, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, 3 Avenue Molière, 67200 Strasbourg, France; Université de Strasbourg, 67000 Strasbourg, France; LabEx Medalis, Université de Strasbourg, 67000 Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France
| | - Alessandra Sacco
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
7
|
Combinations of Kinase Inhibitors Protecting Myoblasts against Hypoxia. PLoS One 2015; 10:e0126718. [PMID: 26042811 PMCID: PMC4456388 DOI: 10.1371/journal.pone.0126718] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 04/07/2015] [Indexed: 01/13/2023] Open
Abstract
Cell-based therapies to treat skeletal muscle disease are limited by the poor survival of donor myoblasts, due in part to acute hypoxic stress. After confirming that the microenvironment of transplanted myoblasts is hypoxic, we screened a kinase inhibitor library in vitro and identified five kinase inhibitors that protected myoblasts from cell death or growth arrest in hypoxic conditions. A systematic, combinatorial study of these compounds further improved myoblast viability, showing both synergistic and additive effects. Pathway and target analysis revealed CDK5, CDK2, CDC2, WEE1, and GSK3β as the main target kinases. In particular, CDK5 was the center of the target kinase network. Using our recently developed statistical method based on elastic net regression we computationally validated the key role of CDK5 in cell protection against hypoxia. This method provided a list of potential kinase targets with a quantitative measure of their optimal amount of relative inhibition. A modified version of the method was also able to predict the effect of combinations using single-drug response data. This work is the first step towards a broadly applicable system-level strategy for the pharmacology of hypoxic damage.
Collapse
|
8
|
Stilhano RS, Martins L, Ingham SJM, Pesquero JB, Huard J. Gene and cell therapy for muscle regeneration. Curr Rev Musculoskelet Med 2015; 8:182-187. [PMID: 25899573 DOI: 10.1007/s12178-015-9268-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Skeletal muscle injury and healing are multifactorial processes, involving three steps of healing: (1) degeneration and inflammation, (2) regeneration, and (3) fibrosis. Fibrous tissue hinders the muscle's complete recovery and current therapies fail in achieving total muscle recovery. Gene and cell therapy (or both) are potential future treatments for severe muscular injuries. Stem cells' properties associated with growth factors or/and cytokines can improve muscle healing and permit long-term recovery.
Collapse
Affiliation(s)
- Roberta Sessa Stilhano
- Biophysics Department, Federal University of São Paulo - UNIFESP, Rua Mirassol, 207 - Vila Clementino, 04044-010 São Paulo, Brazil
| | - Leonardo Martins
- Biophysics Department, Federal University of São Paulo - UNIFESP, Rua Mirassol, 207 - Vila Clementino, 04044-010 São Paulo, Brazil
| | | | - João Bosco Pesquero
- Biophysics Department, Federal University of São Paulo - UNIFESP, Rua Mirassol, 207 - Vila Clementino, 04044-010 São Paulo, Brazil
| | - Johnny Huard
- Stem Cell Research Center, Department of Orthopaedic Surgery, and Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA USA
| |
Collapse
|
9
|
Ward CL, Ji L, Corona BT. An Autologous Muscle Tissue Expansion Approach for the Treatment of Volumetric Muscle Loss. Biores Open Access 2015; 4:198-208. [PMID: 26309796 PMCID: PMC4497650 DOI: 10.1089/biores.2015.0009] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Volumetric muscle loss (VML) is a hallmark of orthopedic trauma with no current standard of care. As a potential therapy for some VML indications, autologous minced muscle grafts (1 mm(3) pieces of muscle) are effective in promoting remarkable de novo fiber regeneration. But they require ample donor muscle tissue and therefore may be limited in their application for large clinical VML. Here, we tested the hypothesis that autologous minced grafts may be volume expanded in a collagen hydrogel, allowing for the use of lesser autologous muscle while maintaining regenerative and functional efficacy. The results of the study indicate that 50% (but not 75%) less minced graft tissue suspended in a collagen hydrogel promoted a functional improvement similar to that of a 100% minced graft repair. However, approximately half of the number of fibers regenerated de novo with 50% graft repair. Moreover, the fibers that regenerated had a smaller cross-sectional area. These findings support the concept of using autologous minced grafts for the regeneration of muscle tissue after VML, but indicate the need to identify optimal carrier materials for expansion.
Collapse
Affiliation(s)
- Catherine L. Ward
- US Army Institute of Surgical Research, Extremity Trauma and Regenerative Medicine, Fort Sam Houston, Texas
| | - Lisa Ji
- US Army Institute of Surgical Research, Extremity Trauma and Regenerative Medicine, Fort Sam Houston, Texas
| | - Benjamin T. Corona
- US Army Institute of Surgical Research, Extremity Trauma and Regenerative Medicine, Fort Sam Houston, Texas
| |
Collapse
|
10
|
Bone marrow-derived mesenchymal cell differentiation toward myogenic lineages: facts and perspectives. BIOMED RESEARCH INTERNATIONAL 2014; 2014:762695. [PMID: 25054145 PMCID: PMC4099119 DOI: 10.1155/2014/762695] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 06/04/2014] [Indexed: 12/11/2022]
Abstract
Bone marrow-derived mesenchymal stem cells (BM-MSCs) are valuable platforms for new therapies based on regenerative medicine. BM-MSCs era is coming of age since the potential of these cells is increasingly demonstrated. In fact, these cells give origin to osteoblasts, chondroblasts, and adipocyte precursors in vitro, and they can also differentiate versus other mesodermal cell types like skeletal muscle precursors and cardiomyocytes. In our short review, we focus on the more recent manipulations of BM-MSCs toward skeletal and heart muscle differentiation, a growing field of obvious relevance considering the toll of muscle disease (i.e., muscular dystrophies), the heavier toll of heart disease in developed countries, and the still not completely understood mechanisms of muscle differentiation and repair.
Collapse
|
11
|
Systemic delivery of human mesenchymal stromal cells combined with IGF-1 enhances muscle functional recovery in LAMA2 dy/2j dystrophic mice. Stem Cell Rev Rep 2013; 9:93-109. [PMID: 22664740 DOI: 10.1007/s12015-012-9380-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The combination of cell therapy with growth factors could be a useful approach to treat progressive muscular dystrophies. Here, we demonstrate, for the first time, that IGF-1 considerably enhances the myogenesis of human umbilical cord (UC) mesenchymal stromal cells (MSCs) in vitro and that IGF-1 enhances interaction and restoration of dystrophin expression in co-cultures of MSCs and muscle cells from Duchenne patients. In vivo studies showed that human MSCs were able to reach the skeletal muscle of LAMA2(dy/2j) dystrophic mice, through systemic delivery, without immunosuppression. Moreover, we showed, for the first time, that IGF-1 injected systemically together with MSCs markedly reduced muscle inflammation and fibrosis, and significantly improved muscle strength in dystrophic mice. Our results suggest that a combined treatment with IGF-1 and MSCs enhances efficiency of muscle repair and, therefore, should be further considered as a potential therapeutic approach in muscular dystrophies.
Collapse
|
12
|
Majka SM, Miller HL, Sullivan T, Erickson PF, Kong R, Weiser-Evans M, Nemenoff R, Moldovan R, Morandi SA, Davis JA, Klemm DJ. Adipose lineage specification of bone marrow-derived myeloid cells. Adipocyte 2012; 1:215-229. [PMID: 23700536 PMCID: PMC3609111 DOI: 10.4161/adip.21496] [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] [Indexed: 12/18/2022] Open
Abstract
We have reported the production of white adipocytes in adipose tissue from hematopoietic progenitors arising from bone marrow. However, technical challenges have hindered detection of this adipocyte population by certain other laboratories. These disparate results highlight the need for sensitive and definitive techniques to identify bone marrow progenitor (BMP)-derived adipocytes. In these studies we exploited new models and methods to enhance detection of this adipocyte population. Here we showed that confocal microscopy with spectrum acquisition could effectively identify green fluorescent protein (GFP) positive BMP-derived adipocytes by matching their fluorescence spectrum to that of native GFP. Likewise, imaging flow cytometry made it possible to visualize intact unilocular and multilocular GFP-positive BMP-derived adipocytes and distinguished them from non-fluorescent adipocytes and cell debris in the cytometer flow stream. We also devised a strategy to detect marker genes in flow-enriched adipocytes from which stromal cells were excluded. This technique also proved to be an efficient means for detecting genetically labeled adipocytes and should be applicable to models in which marker gene expression is low or absent. Finally, in vivo imaging of mice transplanted with BM from adipocyte-targeted luciferase donors showed a time-dependent increase in luciferase activity, with the bulk of luciferase activity confined to adipocytes rather than stromal cells. These results confirmed and extended our previous reports and provided proof-of-principle for sensitive techniques and models for detection and study of these unique cells.
Collapse
|
13
|
Steyn FJ, Ngo ST, Lee JD, Leong JW, Buckley AJ, Veldhuis JD, McCombe PA, Chen C, Bellingham MC. Impairments to the GH-IGF-I axis in hSOD1G93A mice give insight into possible mechanisms of GH dysregulation in patients with amyotrophic lateral sclerosis. Endocrinology 2012; 153:3735-46. [PMID: 22621959 DOI: 10.1210/en.2011-2171] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
GH deficiency has been found in subjects with amyotrophic lateral sclerosis (ALS). Disrupted endocrine function could contribute to the progressive muscle loss and hypermetabolism seen in ALS. It is not possible to study all the elements of the GH-IGF-I axis in ALS patients. Consequently, it remains unclear whether dysfunctional GH secretion contributes to disease pathogenesis and why GH and IGF-I directed treatment strategies are ineffective in human ALS. The hSOD1(G93A) transgenic mouse model is useful for the detailed investigation of the pathogenesis of ALS. We report that symptomatic male hSOD1(G93A) transgenic mice exhibit a deficiency in GH secretion similar to that seen in human ALS. Further characterization of the GH-IGF-I axis in hSOD1(G93A) mice reveals central and peripheral abnormalities that are not found in wild-type age-matched controls. Specifically, we observe aberrant endogenous pulsatile GH secretion, reduced pituitary GH content, and decreased circulating levels of IGF-I, indicating global GH deficiency in hSOD1(G93A) mice. Furthermore, a reduction in the expression of the IGF-I receptor α-subunit in skeletal muscle and lumbar spinal cords of hSOD1(G93A) mice suggests impaired IGF-I signaling within these tissues. This is the first account of disrupted GH secretion in a transgenic mouse model of ALS. These observations are essential for the development of effective GH and IGF-I targeted therapies in ALS.
Collapse
Affiliation(s)
- F J Steyn
- School of Biomedical Sciences, University of Queensland, St. Lucia 4072, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Patil AS, Sable RB, Kothari RM. Role of insulin-like growth factors (IGFs), their receptors and genetic regulation in the chondrogenesis and growth of the mandibular condylar cartilage. J Cell Physiol 2012; 227:1796-804. [PMID: 21732349 DOI: 10.1002/jcp.22905] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Growth of the mandibular condylar cartilage (MCC) is reviewed as a function of genetic and epigenetic factors. The growth centers around the differential spatial concentration of the chondrocytes, influence of growth factors like TGF-β and heterogeneity in the number of IGF receptors, control the action of IGF. Besides these factors, growth of the mandibular condyle is influenced by differential response of chondrocytes as a function of their source/ageing, which in turn is regulated by TGF-β, BMPs and IGFs. While IGF-1 promotes proteoglycan synthesis and survival of the chondrocytes to maintain cartilage homeostasis, TGF-β synergistically catalysed the effect of IGF-1, while BMPs catalysed proteolysis as and when physiologically needed. To understand these processes, role of IGF-1 and its six receptors is at the center to a number of physiological processes being regulated by its mode of application for the growth and differentiation. Probing deeper, biological functions of IGFs seemed to depend on their level of free status rather than bound status to respective IGF-binding proteins (IGF-BPs), considered prerequisite to modulate their biological functions. Genetic regulation of their secretion has thrown light on their insulin-like structural homology, level and response in osteo-arthritis (OA), rheumatic arthritis (RA) and diabetes type-II. Biochemistry and spatial distribution of IGF receptors in different domains exerts control on IGF-1 activities. In ultimate analysis, IGF-axis conserved during the evolution to regulate cell growth and proliferation affect nearly every organ in the body as judged from the techniques determining skeletal maturity and decision making dependent on it for orthodontic, orthognathic/orthopedic and dental implant applications.
Collapse
Affiliation(s)
- Amol S Patil
- Department of Orthodontics and Dentofacial Orthopedics, Bharati Vidyapeeth Dental College and Hospital, Bharati Vidyapeeth Deemed University, Pune, Maharashtra, India.
| | | | | |
Collapse
|
15
|
Ma X, Zhang S, Zhou J, Chen B, Shang Y, Gao T, Wang X, Xie H, Chen F. Clone-derived human AF-amniotic fluid stem cells are capable of skeletal myogenic differentiation in vitro and in vivo. J Tissue Eng Regen Med 2012; 6:598-613. [PMID: 22396316 DOI: 10.1002/term.462] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Revised: 05/02/2011] [Accepted: 07/01/2011] [Indexed: 12/14/2022]
Affiliation(s)
| | | | | | | | | | | | - Xue Wang
- Department of Urology; Shanghai Children's Hospital; Shanghai Jiao Tong University School of Medicine; Shanghai; People's Republic of China
| | - Hua Xie
- Department of Urology; Shanghai Children's Hospital; Shanghai Jiao Tong University School of Medicine; Shanghai; People's Republic of China
| | | |
Collapse
|
16
|
Asakura A. Skeletal Muscle-derived Hematopoietic Stem Cells: Muscular Dystrophy Therapy by Bone Marrow Transplantation. ACTA ACUST UNITED AC 2012; Suppl 11. [PMID: 24524008 PMCID: PMC3918728 DOI: 10.4172/2157-7633.s11-005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
For postnatal growth and regeneration of skeletal muscle, satellite cells, a self-renewing pool of muscle stem cells, give rise to daughter myogenic precursor cells that contribute to the formation of new muscle fibers. In addition to this key myogenic cell class, adult skeletal muscle also contains hematopoietic stem cell and progenitor cell populations which can be purified as a side population (SP) fraction or as a hematopoietic marker CD45-positive cell population. These muscle-derived hematopoietic stem/progenitor cell populations are surprisingly capable of differentiation into hematopoietic cells both after transplantation into irradiated mice and during in vitro colony formation assay. Therefore, these muscle-derived hematopoietic stem/progenitor cells appear to have characteristics similar to classical hematopoietic stem/progenitor cells found in bone marrow. This review outlines recent findings regarding hematopoietic stem/progenitor cell populations residing in adult skeletal muscle and discusses their myogenic potential along with their role in the stem cell niche and related cell therapies for approaching treatment of Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Atsushi Asakura
- Stem Cell Institute, Paul and Sheila Wellstone Muscular Dystrophy Center, Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
17
|
Ieronimakis N, Hays A, Reyes M. Bone marrow-derived cells do not engraft into skeletal muscle microvasculature but promote angiogenesis after acute injury. Exp Hematol 2011; 40:238-249.e3. [PMID: 22155292 DOI: 10.1016/j.exphem.2011.12.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Revised: 11/15/2011] [Accepted: 12/03/2011] [Indexed: 12/28/2022]
Abstract
The skeletal muscle is supported by a vast network of microvessels with the capacity to regenerate in response to injury. However, the dynamics of microvascular repair and the origin of reconstituted endothelial cells in the skeletal muscle are poorly understood. A growing body of literature exists to indicate bone marrow (BM)-derived cells engraft into regenerating vascular endothelium and muscle macrovasculature. Therefore, we investigated the extent of BM contribution to skeletal muscle microvasculature after acute injury. Because reporters and markers commonly used to trace donor BM cells are not endothelial specific but are also expressed by leukocytes, we generated novel BM chimeras utilizing Tie2-green fluorescent protein BM cells transplanted into CD31 and Caveolin-1 knockout recipients. In turn, we surveyed BM vascular contribution, not just by the presence of green fluorescent protein, but also CD31 and Caveolin-1, respectively. After stable BM reconstitution, chimera limb muscles were cardiotoxin (CTX) injured and examined 21 days post-injury for the presence of green fluorescent protein, CD31, and Caveolin-1. Acute muscle injury by CTX is characterized by initial microvasculature death followed by rapid endothelial regeneration within 14 days post-damage. Histological analysis of injured and uninjured contralateral limb muscles revealed a complete absence of BM engraftment in the muscle vasculature of wild-type and CD31/Caveolin-1 knockout chimeras. In contrast, F4/80(+) cells isolated from CTX-injured muscle, expressed endothelial-related markers and promoted angiogenesis in vitro. Therefore, despite the absence of BM engraftment to regenerated skeletal muscle microvasculature, macrophages recruited after injury promote angiogenesis and, in turn, vascular regeneration.
Collapse
Affiliation(s)
- Nicholas Ieronimakis
- Departments of Pathology and Lab Medicine, University of Washington School of Medicine, Seattle, Wash., USA
| | | | | |
Collapse
|
18
|
Janebodin K, Horst OV, Ieronimakis N, Balasundaram G, Reesukumal K, Pratumvinit B, Reyes M. Isolation and characterization of neural crest-derived stem cells from dental pulp of neonatal mice. PLoS One 2011; 6:e27526. [PMID: 22087335 PMCID: PMC3210810 DOI: 10.1371/journal.pone.0027526] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Accepted: 10/19/2011] [Indexed: 01/09/2023] Open
Abstract
Dental pulp stem cells (DPSCs) are shown to reside within the tooth and play an important role in dentin regeneration. DPSCs were first isolated and characterized from human teeth and most studies have focused on using this adult stem cell for clinical applications. However, mouse DPSCs have not been well characterized and their origin(s) have not yet been elucidated. Herein we examined if murine DPSCs are neural crest derived and determined their in vitro and in vivo capacity. DPSCs from neonatal murine tooth pulp expressed embryonic stem cell and neural crest related genes, but lacked expression of mesodermal genes. Cells isolated from the Wnt1-Cre/R26R-LacZ model, a reporter of neural crest-derived tissues, indicated that DPSCs were Wnt1-marked and therefore of neural crest origin. Clonal DPSCs showed multi-differentiation in neural crest lineage for odontoblasts, chondrocytes, adipocytes, neurons, and smooth muscles. Following in vivo subcutaneous transplantation with hydroxyapatite/tricalcium phosphate, based on tissue/cell morphology and specific antibody staining, the clones differentiated into odontoblast-like cells and produced dentin-like structure. Conversely, bone marrow stromal cells (BMSCs) gave rise to osteoblast-like cells and generated bone-like structure. Interestingly, the capillary distribution in the DPSC transplants showed close proximity to odontoblasts whereas in the BMSC transplants bone condensations were distant to capillaries resembling dentinogenesis in the former vs. osteogenesis in the latter. Thus we demonstrate the existence of neural crest-derived DPSCs with differentiation capacity into cranial mesenchymal tissues and other neural crest-derived tissues. In turn, DPSCs hold promise as a source for regenerating cranial mesenchyme and other neural crest derived tissues.
Collapse
Affiliation(s)
- Kajohnkiart Janebodin
- Department of Oral Health Sciences, School of Dentistry, University of Washington, Seattle, Washington, United States of America
- Department of Anatomy, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Orapin V. Horst
- Departments of Dental Public Health Sciences and Endodontics, School of Dentistry, University of Washington, Seattle, Washington, United States of America
| | - Nicholas Ieronimakis
- Department of Pathology, Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Gayathri Balasundaram
- Department of Pathology, Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Kanit Reesukumal
- Department of Clinical Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Busadee Pratumvinit
- Department of Clinical Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Morayma Reyes
- Department of Oral Health Sciences, School of Dentistry, University of Washington, Seattle, Washington, United States of America
- Department of Pathology, Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
19
|
Lu H, Huang D, Ransohoff RM, Zhou L. Acute skeletal muscle injury: CCL2 expression by both monocytes and injured muscle is required for repair. FASEB J 2011; 25:3344-55. [PMID: 21697550 DOI: 10.1096/fj.10-178939] [Citation(s) in RCA: 156] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
CC chemokine ligand 2 (CCL2), a ligand of CC chemokine receptor 2 (CCR2), is essential to mount an adequate inflammatory response to repair acute skeletal muscle injury. We studied the mechanisms by which CCL2 regulates muscle inflammation and regeneration. Mobilization of monocytes/macrophages (MOs/MPs) but not lymphocytes or neutrophils was impaired from bone marrow to blood and from blood to injured muscles in Ccl2(-/-) mice. This was accompanied by poor phagocytosis, reduced up-regulation of insulin-like growth factor-1 (IGF-1), and impaired muscle regeneration. Bone marrow transfer from wild-type mice to irradiated Ccr2(-/-) but not Ccl2(-/-) mice restored muscle inflammation. Intravenously injected CCL2-deficient bone marrow monocytes could not enter wild-type injured muscles as well as wild-type bone marrow monocytes. Intravenously injected wild-type bone marrow monocytes could not enter CCL2-deficient injured muscles as well as wild-type injured muscles. CCL2 stimulated IGF-1 expression by wild-type but not CCR2-deficient intramuscular macrophages. A single intramuscular injection of IGF-1, but not PBS, markedly improved muscle regeneration in Ccl2(-/-) mice. We conclude that CCL2 is a major ligand of CCR2 to recruit MOs/MPs into injured muscles to conduct phagocytosis and produce IGF-1 for injury repair. CCL2 needs to be expressed by bone marrow cells, circulating monocytes, and injured muscle tissue cells to recruit MOs/MPs into injured muscles. CCL2/CCR2 signaling also up-regulates IGF-1 expression by intramuscular macrophages to promote acute skeletal muscle injury repair.
Collapse
Affiliation(s)
- Haiyan Lu
- Neuroinflammation Research Center, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave/S90, Cleveland, OH 44195, USA
| | | | | | | |
Collapse
|
20
|
Gentile A, Toietta G, Pazzano V, Tsiopoulos VD, Giglio AF, Crea F, Pompilio G, Capogrossi MC, Di Rocco G. Human epicardium-derived cells fuse with high efficiency with skeletal myotubes and differentiate toward the skeletal muscle phenotype: a comparison study with stromal and endothelial cells. Mol Biol Cell 2011; 22:581-92. [PMID: 21209317 PMCID: PMC3046056 DOI: 10.1091/mbc.e10-06-0537] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
EPDCs fuse with skeletal myotubes with higher efficiency when compared to MSCs and endothelial cells. Independently from the cell origin, all nuclei recruited inside myotubes express muscle-specific genes. VCAM1 expression in nonmuscle cells is induced by soluble factors secreted by myotubes, and its function is required for fusion to occur. Recent studies have underscored a role for the epicardium as a source of multipotent cells. Here, we investigate the myogenic potential of adult human epicardium-derived cells (EPDCs) and analyze their ability to undergo skeletal myogenesis when cultured with differentiating primary myoblasts. Results are compared to those obtained with mesenchymal stromal cells (MSCs) and with endothelial cells, another mesodermal derivative. We demonstrate that EPDCs spontaneously fuse with pre-existing myotubes with an efficiency that is significantly higher than that of other cells. Although at a low frequency, endothelial cells may also contribute to myotube formation. In all cases analyzed, after entering the myotube, nonmuscle nuclei are reprogrammed to express muscle-specific genes. The fusion competence of nonmyogenic cells in vitro parallels their ability to reconstitute dystrophin expression in mdx mice. We additionally show that vascular cell adhesion molecule 1 (VCAM1) expression levels of nonmuscle cells are modulated by soluble factors secreted by skeletal myoblasts and that VCAM1 function is required for fusion to occur. Finally, treatment with interleukin (IL)-4 or IL-13, two cytokines released by differentiating myotubes, increases VCAM1 expression and enhances the rate of fusion of EPDCs and MSCs, but not that of endothelial cells.
Collapse
Affiliation(s)
- Antonietta Gentile
- Laboratorio di Patologia Vascolare, Istituto Dermopatico dell'Immacolata-IRCCS, 00167 Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Cell Fusion and Tissue Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 713:161-75. [DOI: 10.1007/978-94-007-0763-4_10] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
22
|
Xynos A, Corbella P, Belmonte N, Zini R, Manfredini R, Ferrari G. Bone marrow-derived hematopoietic cells undergo myogenic differentiation following a Pax-7 independent pathway. Stem Cells 2010; 28:965-73. [PMID: 20333749 DOI: 10.1002/stem.418] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Several reports showed that hematopoietic stem cells (HSCs) participate in muscle regeneration, raising hope for their therapeutic potential for degenerative muscle diseases. However, proof that HSCs are able to reprogram their fate and enter a myogenic pathway, remains elusive. We demonstrate that murine bone marrow (BM)-derived hematopoietic cells, carrying reporter genes controlled by muscle-specific regulatory elements from the Myf5, myosin light chain (MLC3F), or MCK genes, are induced by myoblasts to activate muscle-specific genes. This potential resides in the more undifferentiated progenitors, expressing surface markers typical of HSCs. Comparative gene expression profiling of CD45(+)/Sca1(+) cells isolated from muscle or BM shows that hematopoietic cells participate to muscle regeneration, by undergoing a profound although incomplete myogenic reprogramming on interaction with the muscle microenviroment. These cells undergo specification and differentiation independently from Pax7 and MyoD, and lack Pax7-associated properties, such as self-renewal and proliferation, distinguishing from satellite cells. Our findings indicate that hematopoietic cells, on seeding in the muscle, become a distinct cell population endowed with myogenic potential.
Collapse
Affiliation(s)
- Alexandros Xynos
- San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), San Raffaele Scientific Institute, Milan, Italy
| | | | | | | | | | | |
Collapse
|
23
|
Hammers DW, Matheny RW, Sell C, Adamo ML, Walters TJ, Estep JS, Farrar RP. Impairment of IGF-I expression and anabolic signaling following ischemia/reperfusion in skeletal muscle of old mice. Exp Gerontol 2010; 46:265-72. [PMID: 21094246 DOI: 10.1016/j.exger.2010.11.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Revised: 09/17/2010] [Accepted: 11/02/2010] [Indexed: 12/26/2022]
Abstract
With the advancement of age, skeletal muscle undergoes a progressive decline in mass, function, and regenerative capacity. Previously, our laboratory has reported an age-reduction in recovery and local induction of IGF-I gene expression with age following tourniquet (TK)-induced skeletal muscle ischemia/reperfusion (I/R). In this study, young (6 mo) and old (24-28 mo) mice were subjected to 2h of TK-induced ischemia of the hindlimb followed by 1, 3, 5, or 7 days of reperfusion. Real time-PCR analysis revealed clear age-related reductions and temporal alterations in the expression of IGF-I and individual IGF-I Ea and Eb splice variants. ELISA verified a reduction of IGF-I peptide with age following 7 day recovery from TK. Western blotting showed that the phosphorylation of Akt, mTOR, and FoxO3, all indicators of anabolic activity, were reduced in the muscles of old mice. These data indicate that an age-related impairment of IGF-I expression and intracellular signaling does exist following injury, and potentially has a role in the impaired recovery of skeletal muscle with age.
Collapse
Affiliation(s)
- David W Hammers
- Department of Kinesiology, The University of Texas, 1 University Station D3700, Austin, TX 78712, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Otto A, Patel K. Signalling and the control of skeletal muscle size. Exp Cell Res 2010; 316:3059-66. [DOI: 10.1016/j.yexcr.2010.04.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Accepted: 04/12/2010] [Indexed: 11/26/2022]
|
25
|
Vinciguerra M, Hede M, Rosenthal N. Comments on Point:Counterpoint: IGF is/is not the major physiological regulator of muscle mass. IGF-1 is a major regulator of muscle mass during growth but not for adult myofiber hypertrophy. J Appl Physiol (1985) 2010; 108:1831. [PMID: 20527703 DOI: 10.1152/japplphysiol.00312.2010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
26
|
Lu H, Huang D, Saederup N, Charo IF, Ransohoff RM, Zhou L. Macrophages recruited via CCR2 produce insulin-like growth factor-1 to repair acute skeletal muscle injury. FASEB J 2010; 25:358-69. [PMID: 20889618 DOI: 10.1096/fj.10-171579] [Citation(s) in RCA: 197] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
CC chemokine receptor 2 (CCR2) is essential to acute skeletal muscle injury repair. We studied the subpopulation of inflammatory cells recruited via CCR2 signaling and their cellular functions with respect to muscle regeneration. Mobilization of monocytes/macrophages (MOs/MPs), but not lymphocytes or neutrophils, was impaired from bone marrow to blood and from blood to injured muscle in Ccr2(-/-) mice. While the Ly-6C(+) but not the Ly-6C(-) subset of MOs/MPs was significantly reduced in blood, both subsets were drastically reduced in injured muscle of Ccr2(-/-) mice. Expression of insulin-like growth factor-1 (IGF-I) was markedly up-regulated in injured muscle of wild-type but not Ccr2(-/-) mice. IGF-I was strongly expressed by macrophages within injured muscle, more prominently by the Ly-6C(-) subset. A single injection of IGF-I, but not PBS, into injured muscle to replace IGF-I remarkably improved muscle regeneration in Ccr2(-/-) mice. CCR2 was not detected in myogenic cells or capillary endothelial cells in injured muscle to suggest its direct involvement in muscle regeneration or angiogenesis. We conclude that CCR2 is essential to acute skeletal muscle injury repair primarily by recruiting Ly-6C(+) MOs/MPs. Within injured muscle, these cells conduct phagocytosis, contribute to accumulation of intramuscular Ly-6C(-) macrophages, and produce a high level of IGF-I to promote muscle regeneration.
Collapse
Affiliation(s)
- Haiyan Lu
- Neuroinflammation Research Center, Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave./S90, Cleveland, OH 44195, USA
| | | | | | | | | | | |
Collapse
|
27
|
de la Garza-Rodea AS, van der Velde I, Boersma H, Gonçalves MAFV, van Bekkum DW, de Vries AAF, Knaän-Shanzer S. Long-term contribution of human bone marrow mesenchymal stromal cells to skeletal muscle regeneration in mice. Cell Transplant 2010; 20:217-31. [PMID: 20719081 DOI: 10.3727/096368910x522117] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are attractive for cellular therapy of muscular dystrophies as they are easy to procure, can be greatly expanded ex vivo, and contribute to skeletal muscle repair in vivo. However, detailed information about the contribution of bone marrow (BM)-derived human MSCs (BM-hMSCs) to skeletal muscle regeneration in vivo is very limited. Here, we present the results of a comprehensive study of the fate of LacZ-tagged BM-hMSCs following implantation in cardiotoxin (CTX)-injured tibialis anterior muscles (TAMs) of immunodeficient mice. β-Galactosidase-positive (β-gal(+)) human-mouse hybrid myofibers (HMs) were counted in serial cross sections over the full length of the treated TAMs of groups of mice at monthly intervals. The number of human cells was estimated using chemiluminescence assays. While the number of human cells declined gradually to about 10% of the injected cells at 60 days after transplantation, the number of HMs increased from day 10 onwards, reaching 104 ± 39.1 per TAM at 4 months postinjection. β-gal(+) cells and HMs were distributed over the entire muscle, indicating migration of the former from the central injection site to the ends of the TAMs. The identification of HMs that stained positive for human spectrin suggests myogenic reprogramming of hMSC nuclei. In summary, our findings reveal that BM-hMSCs continue to participate in the regeneration/remodeling of CTX-injured TAMs, resulting in ±5% HMs at 4 months after damage induction. Moreover, donor-derived cells were shown to express genetic information, both endogenous and transgenic, in recipient myofibers.
Collapse
Affiliation(s)
- Anabel S de la Garza-Rodea
- Virus and Stem Cell Biology Laboratory, Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
28
|
Misteli H, Wolff T, Füglistaler P, Gianni-Barrera R, Gürke L, Heberer M, Banfi A. High-throughput flow cytometry purification of transduced progenitors expressing defined levels of vascular endothelial growth factor induces controlled angiogenesis in vivo. Stem Cells 2010; 28:611-9. [PMID: 20039367 DOI: 10.1002/stem.291] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Delivery of therapeutic genes by genetically modified progenitors is a powerful tool for regenerative medicine. However, many proteins remain localized within or around the expressing cell, and heterogeneous expression levels can lead to reduced efficacy or increased toxicity. For example, the matrix-binding vascular endothelial growth factor (VEGF) can induce normal, stable, and functional angiogenesis or aberrant angioma growth depending on its level of expression in the microenvironment around each producing cell, and not on its total dose. To overcome this limitation, we developed a flow cytometry-based method to rapidly purify transduced cells expressing desired levels of a therapeutic transgene. Primary mouse myoblasts were transduced with a bicistronic retrovirus expressing VEGF linked to a nonfunctional, truncated form of the syngenic molecule CD8a. By using a clonal population uniformly expressing a known VEGF level as a reference, cells producing similar VEGF amounts were rapidly sorted from the primary population on the basis of their CD8a fluorescence intensity. A single round of sorting with a suitably designed gate yielded a purified population that induced robust, normal, and stable angiogenesis, and completely avoided angioma growth, which was instead always caused by the heterogeneous parent population. This clinically applicable high-throughput technique allowed the delivery of highly controlled VEGF levels in vivo, leading to significantly improved safety without compromising efficacy. Furthermore, when applied to other suitable progenitor populations, this technique could help overcome a significant obstacle in the development of safe and efficacious vascularization strategies in the fields of regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Heidi Misteli
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | | | | | | | | | | | | |
Collapse
|
29
|
Stewart CE, Pell JM. POINT: IGF IS THE MAJOR PHYSIOLOGICAL REGULATOR OF MUSCLE MASS. J Appl Physiol (1985) 2010; 108:1820-1; discussion 1823-4; author reply 1832. [DOI: 10.1152/japplphysiol.01246.2009] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- C. E. Stewart
- Institute for Biomedical Research into Human Movement and Health Manchester, UK
| | - J. M. Pell
- The Babraham Institute Babraham Research Campus Babraham, Cambridge, UK
| |
Collapse
|
30
|
Cosgrove BD, Sacco A, Gilbert PM, Blau HM. A home away from home: challenges and opportunities in engineering in vitro muscle satellite cell niches. Differentiation 2009; 78:185-94. [PMID: 19751902 PMCID: PMC2801624 DOI: 10.1016/j.diff.2009.08.004] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Accepted: 08/11/2009] [Indexed: 12/24/2022]
Abstract
Satellite cells are skeletal muscle stem cells with a principal role in postnatal skeletal muscle regeneration. Satellite cells, like many tissue-specific adult stem cells, reside in a quiescent state in an instructive, anatomically defined niche. The satellite cell niche constitutes a distinct membrane-enclosed compartment within the muscle fiber, containing a diversity of biochemical and biophysical signals that influence satellite cell function. A major limitation to the study and clinical utility of satellite cells is that upon removal from the muscle fiber and plating in traditional plastic tissue culture platforms, their muscle stem cell properties are rapidly lost. Clearly, the maintenance of stem cell function is critically dependent on in vivo niche signals, highlighting the need to create novel in vitro microenvironments that allow for the maintenance and propagation of satellite cells while retaining their potential to function as muscle stem cells. Here, we discuss how emerging biomaterials technologies offer great promise for engineering in vitro microenvironments to meet these challenges. In engineered biomaterials, signaling molecules can be presented in a manner that more closely mimics cell-cell and cell-matrix interactions, and matrices can be fabricated with diverse rigidities that approximate in vivo tissues. The development of in vitro microenvironments in which niche features can be systematically modulated will be instrumental not only to future insights into muscle stem cell biology and therapeutic approaches to muscle diseases and muscle wasting with aging, but also will provide a paradigm for the analysis of numerous adult tissue-specific stem cells.
Collapse
Affiliation(s)
- Benjamin D. Cosgrove
- Baxter Laboratory in Genetic Pharmacology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, USA
- Stem Cell Biology and Regenerative Medicine Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Alessandra Sacco
- Baxter Laboratory in Genetic Pharmacology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Stem Cell Biology and Regenerative Medicine Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Penney M. Gilbert
- Baxter Laboratory in Genetic Pharmacology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Stem Cell Biology and Regenerative Medicine Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Helen M. Blau
- Baxter Laboratory in Genetic Pharmacology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, USA
- Stem Cell Biology and Regenerative Medicine Institute, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
31
|
Otto A, Collins-Hooper H, Patel K. The origin, molecular regulation and therapeutic potential of myogenic stem cell populations. J Anat 2009; 215:477-97. [PMID: 19702867 DOI: 10.1111/j.1469-7580.2009.01138.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Satellite cells, originating in the embryonic dermamyotome, reside beneath the myofibre of mature adult skeletal muscle and constitute the tissue-specific stem cell population. Recent advances following the identification of markers for these cells (including Pax7, Myf5, c-Met and CD34) (CD, cluster of differentiation; c-Met, mesenchymal epithelial transition factor) have led to a greater understanding of the role played by satellite cells in the regeneration of new skeletal muscle during growth and following injury. In response to muscle damage, satellite cells harbour the ability both to form myogenic precursors and to self-renew to repopulate the stem cell niche following myofibre damage. More recently, other stem cell populations including bone marrow stem cells, skeletal muscle side population cells and mesoangioblasts have also been shown to have myogenic potential in culture, and to be able to form skeletal muscle myofibres in vivo and engraft into the satellite cell niche. These cell types, along with satellite cells, have shown potential when used as a therapy for skeletal muscle wasting disorders where the intrinsic stem cell population is genetically unable to repair non-functioning muscle tissue. Accurate understanding of the mechanisms controlling satellite cell lineage progression and self-renewal as well as the recruitment of other stem cell types towards the myogenic lineage is crucial if we are to exploit the power of these cells in combating myopathic conditions. Here we highlight the origin, molecular regulation and therapeutic potential of all the major cell types capable of undergoing myogenic differentiation and discuss their potential therapeutic application.
Collapse
Affiliation(s)
- A Otto
- School of Biological Sciences, Hopkins Building, University of Reading, Whiteknights Campus, Reading, Berkshire, UK
| | | | | |
Collapse
|
32
|
The distribution and characterization of skeletal muscle lesions in dysferlin-deficient SJL and A/J mice. ACTA ACUST UNITED AC 2009; 62:509-17. [PMID: 19615872 DOI: 10.1016/j.etp.2009.06.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Accepted: 06/27/2009] [Indexed: 11/23/2022]
Abstract
The pathogenesis of limb-girdle muscular dystrophy type 2B (LGMD2B) dysferlinopathy remains to be investigated. The distribution and characterization of skeletal muscle lesions were examined in two different LGMD2B mouse models, SJL and A/J mice (at 10 and 35 weeks old), in association with the endoplasmic reticulum (ER) stress. SJL mice showed an earlier age of onset and a faster progression of skeletal muscle lesions as compared with those of A/J mice; the sensitivity difference to muscular dystrophic lesions between SJL and A/J mice was observed in the lumbar muscles (particularly, lumbar longissimus and sublumbar muscles); the lesions seen mainly in SJL mice at 35 weeks old consisted of degeneration, necrosis, fatty infiltration, variation in muscle fiber size and atrophy in muscle fibers. Enzyme-histochemically, the fast-twitch muscle fiber was predominant for the degenerative changes seen in the rectus femoris and lateral longissimus muscles of SJL mice. Immunohistochemically, the main reactive cell type observed in and around degenerative and/or necrotic muscle fibers was macrophages, demonstrable with an anti-F4/80 antibody. Because the analyses of spliced XBP1 mRNA, a marker of ER stress, did not show the increased expression, it was considered that ER stress did not affect the progression of skeletal muscle lesions in SJL mice with the advanced stage of dysferlinopathy.
Collapse
|
33
|
Abstract
Both skeletal muscle and bone marrow tissue contain myogenic stem cells. The population residing in muscles is heterogenic. Predominant in number are "typical" satellite cells - muscle progenitors migrating from somites during embryonic life. Another population is group of multipotent muscle stem cells which, at least in part, are derived from bone marrow. These cells are tracked by gradient of growth factors releasing from muscle during injury or exercise. Recruited bone marrow-derived cells gradually change their phenotype becoming muscle stem cells and eventually can attain satellite cell position and express Pax7 protein. Mesenchymal stem cells (MSC) isolated directly from bone marrow also display myogenic potential, although methods of induction of myogenic differentiation in vitro have not been optimized yet. Concerning efforts of exploiting myogenic stem cells in cell-mediated therapies it is important to understand the cause of impaired regenerative potential of aged muscle. Up to now, most of research data suggest that majority of age related changes in skeletal muscles are reversible, thus depending on extrinsic factors. However, irreversible intrinsic features of muscle stem cells are also taken into consideration.
Collapse
|
34
|
Sacco A, Doyonnas R, Kraft P, Vitorovic S, Blau HM. Self-renewal and expansion of single transplanted muscle stem cells. Nature 2008; 456:502-6. [PMID: 18806774 DOI: 10.1038/nature07384] [Citation(s) in RCA: 635] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2008] [Accepted: 09/01/2008] [Indexed: 01/25/2023]
Abstract
Adult muscle satellite cells have a principal role in postnatal skeletal muscle growth and regeneration. Satellite cells reside as quiescent cells underneath the basal lamina that surrounds muscle fibres and respond to damage by giving rise to transient amplifying cells (progenitors) and myoblasts that fuse with myofibres. Recent experiments showed that, in contrast to cultured myoblasts, satellite cells freshly isolated or satellite cells derived from the transplantation of one intact myofibre contribute robustly to muscle repair. However, because satellite cells are known to be heterogeneous, clonal analysis is required to demonstrate stem cell function. Here we show that when a single luciferase-expressing muscle stem cell is transplanted into the muscle of mice it is capable of extensive proliferation, contributes to muscle fibres, and Pax7(+)luciferase(+) mononucleated cells can be readily re-isolated, providing evidence of muscle stem cell self-renewal. In addition, we show using in vivo bioluminescence imaging that the dynamics of muscle stem cell behaviour during muscle repair can be followed in a manner not possible using traditional retrospective histological analyses. By imaging luciferase activity, real-time quantitative and kinetic analyses show that donor-derived muscle stem cells proliferate and engraft rapidly after injection until homeostasis is reached. On injury, donor-derived mononucleated cells generate massive waves of cell proliferation. Together, these results show that the progeny of a single luciferase-expressing muscle stem cell can both self-renew and differentiate after transplantation in mice, providing new evidence at the clonal level that self-renewal is an autonomous property of a single adult muscle stem cell.
Collapse
Affiliation(s)
- Alessandra Sacco
- Baxter Laboratory in Genetic Pharmacology, Department of Microbiology and Immunology, Stem Cell Institute, Stanford University School of Medicine, Stanford, California 94305-5175, USA
| | | | | | | | | |
Collapse
|
35
|
Schwartz M, Ziv Y. Immunity to self and self-maintenance: a unified theory of brain pathologies. Trends Immunol 2008; 29:211-9. [DOI: 10.1016/j.it.2008.01.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2007] [Revised: 01/05/2008] [Accepted: 01/09/2008] [Indexed: 10/22/2022]
|
36
|
Johansson CB, Youssef S, Koleckar K, Holbrook C, Doyonnas R, Corbel SY, Steinman L, Rossi FMV, Blau HM. Extensive fusion of haematopoietic cells with Purkinje neurons in response to chronic inflammation. Nat Cell Biol 2008; 10:575-83. [PMID: 18425116 DOI: 10.1038/ncb1720] [Citation(s) in RCA: 186] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2008] [Accepted: 04/09/2008] [Indexed: 12/13/2022]
Abstract
Transplanted bone marrow-derived cells (BMDCs) have been reported to fuse with cells of diverse tissues, but the extremely low frequency of fusion has led to the view that such events are biologically insignificant. Nonetheless, in mice with a lethal recessive liver disease (tyrosinaemia), transplantation of wild-type BMDCs restored liver function by cell fusion and prevented death, indicating that cell fusion can have beneficial effects. Here we report that chronic inflammation resulting from severe dermatitis or autoimmune encephalitis leads to robust fusion of BMDCs with Purkinje neurons and formation of hundreds of binucleate heterokaryons per cerebellum, a 10-100-fold higher frequency than previously reported. Single haematopoietic stem-cell transplants showed that the fusogenic cell is from the haematopoietic lineage and parabiosis experiments revealed that fusion can occur without irradiation. Transplantation of rat bone marrow into mice led to activation of dormant rat Purkinje neuron-specific genes in BMDC nuclei after fusion with mouse Purkinje neurons, consistent with nuclear reprogramming. The precise neurological role of these heterokaryons awaits elucidation, but their frequency in brain after inflammation is clearly much higher than previously appreciated.
Collapse
Affiliation(s)
- Clas B Johansson
- Baxter Laboratory in Genetic Pharmacology, Stanford University School of Medicine, Stanford, CA 94305-5175, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Stolzing A, Hescheler J, Sethe S. Fusion and Regenerative Therapies: Is Immortality Really Recessive? Rejuvenation Res 2007; 10:571-86. [DOI: 10.1089/rej.2007.0570] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
|
38
|
Dimmeler S, Burchfield J, Zeiher AM. Cell-based therapy of myocardial infarction. Arterioscler Thromb Vasc Biol 2007; 28:208-16. [PMID: 17951319 DOI: 10.1161/atvbaha.107.155317] [Citation(s) in RCA: 253] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cell-based therapy is a promising option for treatment of ischemic diseases. Several cell types have experimentally been shown to increase the functional recovery of the heart after ischemia by physically forming new blood vessels, differentiating to cardiac myocytes and--additionally or alternatively--by providing proangiogenic and antiapoptotic factors promoting tissue repair in a paracrine manner. Clinical studies preferentially used adult bone marrow-derived cells for the treatment of patients with acute myocardial infarction. Most of the studies suggested that cell therapy reduced the infarct size and improved cardiac contractile function. However, cell therapy is in its early stages, and various questions remain. For example, the identification of those patients who benefit most from cell therapy, the optimal cell type and number for patient with acute and chronic diseases, the best time and way of cell delivery, and the mechanisms of action by which cells exhibit beneficial effects, need to be further evaluated. Although no major safety concerns were raised during the initial clinical trials, several potential side effects need to be carefully monitored. The present review article summarizes the results of the clinical studies and discusses the open issues.
Collapse
Affiliation(s)
- Stefanie Dimmeler
- Molecular Cardiology, Department of Internal Medicine III, University of Frankfurt, Theodor Stern-Kai 7, 60590 Frankfurt, Germany.
| | | | | |
Collapse
|
39
|
Mills P, Dominique JC, Lafrenière JF, Bouchentouf M, Tremblay JP. A synthetic mechano growth factor E Peptide enhances myogenic precursor cell transplantation success. Am J Transplant 2007; 7:2247-59. [PMID: 17845560 DOI: 10.1111/j.1600-6143.2007.01927.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Myogenic precursor cell (MPC) transplantation is a good strategy to introduce dystrophin expression in muscles of Duchenne muscular dystrophy (DMD) patients. Insulin-like growth factor (IGF-1) promotes MPC activities, such as survival, proliferation, migration and differentiation, which could enhance the success of their transplantation. Alternative splicing of the IGF-1 mRNA produces different muscle isoforms. The mechano growth factor (MGF) is an isoform, especially expressed after a mechanical stress. A 24 amino acids peptide corresponding to the C-terminal part of the MGF E domain (MGF-Ct24E peptide) was synthesized. This peptide had been shown to enhance the proliferation and delay the terminal differentiation of C(2)C(12) myoblasts. The present study showed that the MGF-Ct24E peptide improved human MPC transplantation by modulating their proliferation and differentiation. Indeed, intramuscular or systemic delivery of this synthetic peptide significantly promoted engraftment of human MPCs in mice. In vitro experiments demonstrated that the MGF-Ct24E peptide enhanced MPC proliferation by a different mechanism than the binding to the IGF-1 receptor. Moreover, MGF-Ct24E peptide delayed human MPC differentiation while having no outcome on survival. Those combined effects are probably responsible for the enhanced transplantation success. Thus, the MGF-Ct24E peptide is an interesting agent to increase MPC transplantation success in DMD patients.
Collapse
Affiliation(s)
- P Mills
- Unité de recherche en Génétique humaine, Centre Hospitalier de l'Université Laval, Ste-Foy, Québec, Canada
| | | | | | | | | |
Collapse
|
40
|
Castaldi L, Serra C, Moretti F, Messina G, Paoletti R, Sampaolesi M, Torgovnick A, Baiocchi M, Padula F, Pisaniello A, Molinaro M, Cossu G, Levrero M, Bouché M. Bisperoxovanadium, a phospho-tyrosine phosphatase inhibitor, reprograms myogenic cells to acquire a pluripotent, circulating phenotype. FASEB J 2007; 21:3573-83. [PMID: 17601985 DOI: 10.1096/fj.06-7454com] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Satellite cells are the main source of myogenic progenitors in postnatal skeletal muscle, but their use in cell therapy for muscle disorders is limited because these cells cannot be delivered through circulation and they are rapidly exhausted in severe myopathies. The search for alternative donor cells is ongoing, but none of the candidates so far show all the features required for successful colonization and repair of diseased muscle. In this study, we show that bisperoxovanadium, a phospho-tyrosine phosphatase inhibitor, induces myogenic cells to acquire a gene expression profile and a differentiation potential consistent with the phenotype of a circulating precursors, while maintaining their myogenic potential. These effects are mediated, at least in part, by NF-kappaB activation through the Tyr42-IkappaB-alpha phosphorylation, as shown by the expression of the dominant negative mutant form of the p50 NF-kappaB subunit. Moreover, when bisperoxovanadium-treated cells are injected into the femoral artery of alpha-sarcoglican null dystrophic mice, they are able to circulate and to reach muscle tissue; importantly, they contribute to muscle regeneration, as shown by the expression of alpha-sarcoglican in some fibers. Our observations indicate that bisperoxovanadium, or similar compounds, may prove very valuable to obtain and to expand, from committed cells, multipotent cell populations suitable for gene-cell therapy applications and may help to understand the molecular basis of genome reprogramming and "stem-ness."
Collapse
Affiliation(s)
- L Castaldi
- Department of Histology and Med. Embr., Univ. of Rome La Sapienza, Via A. Scarpa 14, 00161 Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Yoshida K, Obata S, Ono M, Esaki M, Maejima T, Sawada H. TPA-induced multinucleation of a mesenchymal stem cell-like clone is mediated primarily by karyokinesis without cytokinesis, although cell-cell fusion also occurs. Eur J Cell Biol 2007; 86:461-71. [PMID: 17599648 DOI: 10.1016/j.ejcb.2007.04.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2006] [Revised: 04/03/2007] [Accepted: 04/13/2007] [Indexed: 10/23/2022] Open
Abstract
The 5F9A cell, which is a mesenchymal stem cell-like clone established from rat bone marrow substrate adherent cells, can differentiate into adipocytes and osteoblasts in vitro under the appropriate conditions. Multinucleated cells could be also induced by 12-O-tetradecanoylphorbol 13-acetate (TPA) in 5F9A cells. This effect was mediated by protein kinase C. Possible mechanisms of multinucleation by TPA were hypothesized to be either karyokinesis without cytokinesis or cell-cell fusion. By observation using time-lapse phase-contrast microscopy, we determined that the multinucleated cells were generated mainly by karyokinesis without cytokinesis. Cell fusion was studied using time-lapse photography, and confocal laser scanning microscopy using two differentially labeled cells. These techniques demonstrated that multinucleated 5F9A cells could be produced by cell fusion, albeit at a low frequency. We conclude that multinucleated 5F9A cells are formed primarily by karyokinesis without cytokinesis, although some cells are also formed by cell-cell fusion.
Collapse
Affiliation(s)
- Keiichiro Yoshida
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Fukuura 3-9, Kanazawa-ku, Yokohama, Kanagawa-ken 236-0004, Japan.
| | | | | | | | | | | |
Collapse
|
42
|
Herzog EL, Van Arnam J, Hu B, Zhang J, Chen Q, Haberman AM, Krause DS. Lung‐specific nuclear reprogramming is accompanied by heterokaryon formation and Y chromosome loss following bone marrow transplantation and secondary inflammation. FASEB J 2007; 21:2592-601. [PMID: 17449722 DOI: 10.1096/fj.06-7861com] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cell fusion is one mechanism by which bone marrow-derived cells (BMDCs) take on the gene expression pattern of nonhematopoietic cells. This process occurs in a number of organs with postengraftment injury but has never been found in the lung. We performed bone marrow (BM) transplant in a murine model of lung inflammation to test whether transplanted BMDCs develop lung-specific gene expression by fusing with diseased pneumocytes. Mice lacking the lung-specific protein surfactant protein C (Sp-C) were lethally irradiated, transplanted with sex mismatched wild-type marrow, and sacrificed 6 months later. Nineteen/38 recipients exhibited Sp-C mRNA (RT-PCR) and/or protein (mean 0.95+/-1.18 Sp-C+ cells per 1000 type II pneumocytes by confocal microscopy). In male recipients of female BM, 65% of Sp-C + cells contained the Y chromosome, indicating their origin from fusion. Only 28% of Sp-C+ cells in female recipients of male BMDCs contained the Y chromosome, suggesting that 72% of Sp-C-expressing cells lost the Y chromosome. In the setting of post-transplant inflammation, pneumocyte-specific reprogramming of transplanted BMDCs predominantly derives from heterokaryon formation. This process does not reverse inflammation caused by Sp-C deficiency; nevertheless, further investigation may identify phenotypes benefiting from such an approach.
Collapse
Affiliation(s)
- Erica L Herzog
- Yale University School of Medicine, Internal Medicine-Pulmonary and Critical Care Division, 333 Cedar St., TAC 441-S, New Haven CT 06511, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Moreno RJ, Messi ML, Zheng Z, Wang ZM, Ye P, D'Ercole JA, Delbono O. Role of sustained overexpression of central nervous system IGF-I in the age-dependent decline of mouse excitation-contraction coupling. J Membr Biol 2007; 212:147-61. [PMID: 17334835 DOI: 10.1007/s00232-006-0044-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2006] [Revised: 09/18/2006] [Indexed: 12/20/2022]
Abstract
We investigated the effects of exclusive and sustained transgenic overexpression of insulin-like growth factor (IGF)-I in the central nervous system (CNS) on the age-dependent decline in muscle strength, excitation-contraction coupling, muscle innervation and neuromuscular junction postterminal architecture. We found that (1) transgenic IGF-I overexpression in the CNS does not modify the decline in extensor digitorum longus (EDL) and soleus muscle weight with aging and (2) strength significantly decreases in transgenic (Tg) compared to wild-type mice. The latter finding is consistent with (3) the decreased absolute and specific force measured in the EDL muscle in vitro and (4) the decreased charge movement and peak intracellular Ca(2+) mobilization in individual muscle fibers from old IGF-I Tg mice compared to young wild-type mice, which also is associated with (5) decreased dihydropyridine receptor alpha(1)-subunit expression in old compared to young IGF-I Tg mice. (6) Tg IGF-I prevents a change in muscle fiber type that is associated with (7) improved muscle innervation and postterminal neuromuscular structure. (8) IGF-I is expressed extensively across the spinal cord gray matter and the lateral motor column. Our results raise questions about the timing and cell location of CNS IGF-I overexpression necessary to prevent or to ameliorate age-dependent alterations in the structure and function of skeletal muscle.
Collapse
Affiliation(s)
- Ramón Jiménez Moreno
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Dedja A, Zaglia T, Dall'Olmo L, Chioato T, Thiene G, Fabris L, Ancona E, Schiaffino S, Ausoni S, Cozzi E. Hybrid cardiomyocytes derived by cell fusion in heterotopic cardiac xenografts. FASEB J 2006; 20:2534-6. [PMID: 17077278 DOI: 10.1096/fj.06-6586fje] [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: 01/06/2023]
Abstract
Cardiomyocytes expressing host markers, such as the Y chromosome in sex-mismatched transplants, have been described in human allografts, suggesting that circulating cells can contribute to cardiac regeneration. It has not been established, however, whether host-derived cardiomyocytes result from transdifferentiation of stem cells or cell fusion. To address this issue, we used heterotopic heart xenografts and looked for markers of donor and recipient cells. Golden Syrian hamsters or transgenic mice expressing nuclear beta-galactosidase under the control of the cardiac troponin I promoter served as organ donors, while GFP+ transgenic rats were used as recipients. GFP+ cells, including abundant CD-45+ inflammatory cells and rare undifferentiated cells expressing early cardiac markers (GATA-4 or MEF2C), were found in xenografts harvested two weeks after surgery. In addition, rare GFP+ mature cardiomyocytes were found in 7 of 8 hamster xenografts and 6 of 6 mouse xenografts. The proportion of these cells was very low (0.0001% to 0.0344% in hamster xenografts) but similar to the one observed in control rat heart allografts. Without exception, all GFP+ cardiomyocytes also expressed donor markers, i.e., hamster membrane antigens or lacZ, so they must derive from cell fusion, not transdifferentiation.
Collapse
Affiliation(s)
- Arben Dedja
- Department of Medical and Surgical Sciences, University of Padova, Padova, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Foster K, Foster H, Dickson JG. Gene therapy progress and prospects: Duchenne muscular dystrophy. Gene Ther 2006; 13:1677-85. [PMID: 17066097 DOI: 10.1038/sj.gt.3302877] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a severe muscle wasting disorder affecting 1/3500 male births. There is currently no effective treatment, but gene therapy approaches are offering viable avenues for treatment development. The last 10 years have seen the development of a number of strategies and tools for muscle gene therapy. However, the major hurdle has been the inability to deliver vectors at high enough efficiency via a systemic route. The last 2-3 years (reviewed here) have seen unrivalled progress in efficient systemic delivery of viral and non-viral gene transfer agents and antisense oligonucleotides. This progress, coupled with the successful completion of the first gene therapy clinical trial for DMD, has led to three more clinical trials planned for the immediate future.
Collapse
Affiliation(s)
- K Foster
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, Surrey, UK
| | | | | |
Collapse
|
46
|
Wang J, Wang N, Xie J, Walton SC, McKown RL, Raab RW, Ma P, Beck SL, Coffman GL, Hussaini IM, Laurie GW. Restricted epithelial proliferation by lacritin via PKCalpha-dependent NFAT and mTOR pathways. ACTA ACUST UNITED AC 2006; 174:689-700. [PMID: 16923831 PMCID: PMC1761701 DOI: 10.1083/jcb.200605140] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Renewal of nongermative epithelia is poorly understood. The novel mitogen "lacritin" is apically secreted by several nongermative epithelia. We tested 17 different cell types and discovered that lacritin is preferentially mitogenic or prosecretory for those types that normally contact lacritin during its glandular outward flow. Mitogenesis is dependent on lacritin's C-terminal domain, which can form an alpha-helix with a hydrophobic face, as per VEGF's and PTHLP's respective dimerization or receptor-binding domain. Lacritin targets downstream NFATC1 and mTOR. The use of inhibitors or siRNA suggests that lacritin mitogenic signaling involves Galpha(i) or Galpha(o)-PKCalpha-PLC-Ca2+-calcineurin-NFATC1 and Galpha(i) or Galpha(o)-PKCalpha-PLC-phospholipase D (PLD)-mTOR in a bell-shaped, dose-dependent manner requiring the Ca2+ sensor STIM1, but not TRPC1. This pathway suggests the placement of transiently dephosphorylated and perinuclear Golgi-translocated PKCalpha upstream of both Ca2+ mobilization and PLD activation in a complex with PLCgamma2. Outward flow of lacritin from secretory cells through ducts may generate a proliferative/secretory field as a different unit of cellular renewal in nongermative epithelia where luminal structures predominate.
Collapse
Affiliation(s)
- Jiahu Wang
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22904, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Carrington JL, Bellino FL. Developing a research agenda in biogerontology: physiological systems. SCIENCE OF AGING KNOWLEDGE ENVIRONMENT : SAGE KE 2006; 2006:pe17. [PMID: 16807481 DOI: 10.1126/sageke.2006.10.pe17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The Biology of Aging Program (BAP) at the National Institute on Aging supports research in many areas, including processes of cell senescence and apoptosis, genetic influences on aging, and how aging leads to tissue dysfunction. Several approaches to research on aging physiological systems are described, along with BAP programmatic efforts to enhance and support that research. Understanding the relation between aging and tissue dysfunction has led to new insights into how health can be improved for aged individuals.
Collapse
Affiliation(s)
- Jill L Carrington
- Biology of Aging Program, National Institute on Aging, Bethesda, MD 20892, USA.
| | | |
Collapse
|