1
|
Redden JT, Cohen DJ, Olson LC, Bendale G, Isaacs JE, Schwartz Z, McClure MJ. Neurotization of decellularized muscle graft increases de novo type I slow muscle fiber formation and large fiber size frequency. Acta Biomater 2025; 191:244-259. [PMID: 39551332 DOI: 10.1016/j.actbio.2024.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/22/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
Volumetric muscle loss (VML) injuries are the result of extreme trauma from battlefield injuries, tumor ablations, and other physical traumas such as car crash injuries. The abrupt loss of muscle restricts the tissue's remaining regenerative capacity, leading to loss of satellite cells, peripheral nerve connections, and aberrant fibrosis. Prior research from our lab demonstrated that decellularized muscle matrix (DMM) supported regeneration of de novo fibers within the graft. The goal of this study was to determine whether DMM treated with a peripheral nerve using neurotization surgeries would enhance muscle regeneration and innervation. Forty-eight male Sprague Dawley rats were randomized and received a 1.5×1 cm defect treated with no treatment empty defect (ED), DMM, or autograft with a direct peroneal (antagonist) neurotization or tibial via end to side graft (agonist) neurotization. DMM grafts treated with neurotization utilizing either peroneal or tibial nerve axons increased fast twitch fibers within the grafted area compared to untreated DMM or ED. Additionally, the frequency distribution of myofiber size shifted toward a healthier morphology in the tibial nerve axon neurotized DMM compared to the uninjured medial head. Lastly, Nanostring gene results showed DMM treated with a neurotization shifted expression towards a more regenerative phenotype with some myogenic markers returning to sham levels. These data indicate that injured muscle treated with DMM and neurotization becomes pro-regenerative and can contribute to the functionalization of DMM. STATEMENT OF SIGNIFICANCE: Extremity soft tissue trauma like volumetric muscle loss (VML) can result in permanent loss of skeletal muscle mass, denervation, and ischemia posing a significant clinical challenge. VML injuries disrupt normal tissue architecture in addition to intramuscular axons which are critical elements in muscle function and regeneration. The overall objective of this study was to enhance axon growth into a VML injury treated with decellularized muscle matrix (DMM) using neurotization. DMM is an acellular biomaterial capable of regenerating skeletal muscle; however, without bona fide neuromuscular connections, functional gains are small. This study demonstrates that introducing motor axons into an acellular regenerative material using neurotization enhanced muscle regeneration and promoted slow twitch fiber formation.
Collapse
Affiliation(s)
- James T Redden
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA 23284, USA
| | - David J Cohen
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA 23284, USA
| | - Lucas C Olson
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA 23284, USA
| | - Geetanjali Bendale
- Division of Hand Surgery, Department of Orthopaedic Surgery, Virginia Commonwealth University Health System, Richmond, VA 23298, USA
| | - Jonathan E Isaacs
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA 23284, USA; Division of Hand Surgery, Department of Orthopaedic Surgery, Virginia Commonwealth University Health System, Richmond, VA 23298, USA
| | - Zvi Schwartz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA 23284, USA; Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Michael J McClure
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA 23284, USA; Division of Hand Surgery, Department of Orthopaedic Surgery, Virginia Commonwealth University Health System, Richmond, VA 23298, USA.
| |
Collapse
|
2
|
Rieger L, Molina T, Fabre P, Greffard K, Pellerito O, Dort J, Bilodeau JF, Dumont NA. Transcriptomic and lipidomic profiling reveals distinct bioactive lipid signatures in slow and fast muscles and highlights the role of resolvin-D2 in fiber type determination during myogenesis. FASEB J 2024; 38:e70250. [PMID: 39698915 DOI: 10.1096/fj.202401747r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/14/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024]
Abstract
Skeletal muscles are predominantly composed of long, multinucleated muscle fibers, classified according to their metabolic and contractile phenotype. The determination of fiber types is influenced by various factors (e.g., innervation, hormones, physical demand). Our laboratory and others showed that resolvins, lipid mediators derived from omega-3 fatty acids, promote muscle regeneration and function after an injury or in models of muscular dystrophies; however, the effect of resolvins on the determination of muscle phenotype remains unknown. Here, we investigated the impact of lipid mediators on muscle phenotype during myogenesis. Transcriptomics analysis of single-nuclei RNAseq data sets revealed that the enzymes responsible for bioactive lipids biosynthesis are differentially expressed in slow fibers versus fast fibers. Lipidomics analysis of slow-twitch muscle (soleus) versus fast-twitch muscle (tibialis anterior) showed that the levels of lipids derived from arachidonic acid are similar between muscle groups, but lipids derived from alpha-linolenic acid, linoleic acid, eicosapentaenoic acid, and docosahexaenoic acid are enriched in slow-twitch muscle. Screening for different lipids in vitro showed that resolvin-D2 enhances the formation of myotubes expressing the slow myosin heavy chain isoform. In vivo, the administration of resolvin-D2 enhances muscle strength, increases myofiber size, and affects fiber typing in injured muscles but not in uninjured muscles. Resolvin-D2 promoted the transition toward the dominant fiber types in regenerating muscle (i.e., type I in the slow-twitch soleus and type IIB in the fast-twitch tibialis anterior muscle), suggesting its participation in fiber typing in conjunction with other factors. Overall, these findings identified new roles of bioactive lipids in the regulation of fiber typing, which could have therapeutic applicability in muscle injuries or dystrophies.
Collapse
Affiliation(s)
- Lupann Rieger
- CHU Sainte-Justine Research Center, Montreal, Quebec, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Thomas Molina
- CHU Sainte-Justine Research Center, Montreal, Quebec, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Paul Fabre
- CHU Sainte-Justine Research Center, Montreal, Quebec, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Karine Greffard
- Endocrinology and Nephrology Unit, CHU de Québec-Laval University Research Center, Quebec, Quebec, Canada
| | | | - Junio Dort
- School of Pharmaceutical Sciences, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Jean-François Bilodeau
- Endocrinology and Nephrology Unit, CHU de Québec-Laval University Research Center, Quebec, Quebec, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Quebec, Canada
| | - Nicolas A Dumont
- CHU Sainte-Justine Research Center, Montreal, Quebec, Canada
- School of Rehabilitation, Faculty of Medicine, Université de Montréal, Montréal, Quebec, Canada
| |
Collapse
|
3
|
Zhang H, Wague A, Diaz A, Liu M, Sang L, Youn A, Sharma S, Milan N, Kim H, Feeley B, Liu X. Overexpression of PRDM16 improves muscle function after rotator cuff tears. J Shoulder Elbow Surg 2024; 33:2725-2733. [PMID: 39032686 PMCID: PMC12070449 DOI: 10.1016/j.jse.2024.05.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/01/2024] [Accepted: 05/19/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Muscle atrophy, fibrosis, and fatty infiltration are commonly seen in rotator cuff tears (RCTs), which are critical factors that directly determine the clinical outcomes for patients with this injury. Therefore, improving muscle quality after RCT is crucial in improving the clinical outcome of tendon repair. In recent years, it has been discovered that adults have functional beige/brown adipose tissue (BAT) that can secrete batokines to promote muscle growth. PRDM16, a PR-domain-containing protein, was discovered with the ability to determine the brown fat cell fate and stimulate its development. Thus, the goal of this study was to discover the role of PRDM16 in improving muscle function after massive tendon tears using a transgenic mouse model with an elevated level of PRDM16 expression. METHODS Transgenic aP2-driven PRDM16-overexpressing mice and C57BL/6J mice underwent unilateral supraspinatus (SS) tendon transection and suprascapular nerve transection (TTDN) as described previously (n = 8 in each group). DigiGait was performed to evaluate forelimb function at 6 weeks post the TTDN injury. Bilateral SS muscles, interscapular brown fat, epididymal white fat, and inguinal beige fat were harvested for analysis. The expression of PRDM16 in adipose tissue was detected by Western blot. Masson Trichrome staining was conducted to evaluate the muscle fibrosis, and Oil Red O staining was used to determine the fat infiltration. Muscle fiber type was determined by major histocompatibility complex (MHC) expression via immunostaining. All data were presented in the form of mean ± standard deviation. t test and 2-way analysis of variance was performed to determine a statistically significant difference between groups. Significance was considered when P < .05. RESULTS Western blot data showed an increased expression of PRDM16 protein in both white and brown fat in PRDM16-overexpressing mice compared with wild-type (WT) mice. Even though PRDM16 overexpression had no effect on increasing muscle weight, it significantly improved the forelimbs function with longer brake, stance, and stride time and larger stride length and paw area in mice after RCT. Additionally, PRDM16-overexpressing mice showed no difference in the amount of fibrosis when compared to WT mice; however, they had a significantly reduced area of fatty infiltration. These mice also exhibited abundant MHC-IIx fiber percentage in the supraspinatus muscle after TTDN. CONCLUSION Overexpression of PRDM16 significantly improved muscle function and reduced fatty infiltration after rotator cuff tears. Promoting BAT activity is beneficial in improving rotator cuff muscle quality and shoulder function after RCT.
Collapse
Affiliation(s)
- He Zhang
- Department of Physical Education, Central South University, Changsha, Hunan, China; Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Aboubacar Wague
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Agustin Diaz
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Mengyao Liu
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Luke Sang
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Alex Youn
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Sankalp Sharma
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Nesa Milan
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Hubert Kim
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Brian Feeley
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Xuhui Liu
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
4
|
Korb A, Tajbakhsh S, Comai GE. Functional specialisation and coordination of myonuclei. Biol Rev Camb Philos Soc 2024; 99:1164-1195. [PMID: 38477382 DOI: 10.1111/brv.13063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 01/30/2024] [Accepted: 02/02/2024] [Indexed: 03/14/2024]
Abstract
Myofibres serve as the functional unit for locomotion, with the sarcomere as fundamental subunit. Running the entire length of this structure are hundreds of myonuclei, located at the periphery of the myofibre, juxtaposed to the plasma membrane. Myonuclear specialisation and clustering at the centre and ends of the fibre are known to be essential for muscle contraction, yet the molecular basis of this regionalisation has remained unclear. While the 'myonuclear domain hypothesis' helped explain how myonuclei can independently govern large cytoplasmic territories, novel technologies have provided granularity on the diverse transcriptional programs running simultaneously within the syncytia and added a new perspective on how myonuclei communicate. Building upon this, we explore the critical cellular and molecular sources of transcriptional and functional heterogeneity within myofibres, discussing the impact of intrinsic and extrinsic factors on myonuclear programs. This knowledge provides new insights for understanding muscle development, repair, and disease, but also opens avenues for the development of novel and precise therapeutic approaches.
Collapse
Affiliation(s)
- Amaury Korb
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Stem Cells & Development Unit, 25 rue du Dr. Roux, Institut Pasteur, Paris, F-75015, France
| | - Shahragim Tajbakhsh
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Stem Cells & Development Unit, 25 rue du Dr. Roux, Institut Pasteur, Paris, F-75015, France
| | - Glenda E Comai
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Stem Cells & Development Unit, 25 rue du Dr. Roux, Institut Pasteur, Paris, F-75015, France
| |
Collapse
|
5
|
Guilhot C, Catenacci M, Lofaro S, Rudnicki MA. The satellite cell in skeletal muscle: A story of heterogeneity. Curr Top Dev Biol 2024; 158:15-51. [PMID: 38670703 DOI: 10.1016/bs.ctdb.2024.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Skeletal muscle is a highly represented tissue in mammals and is composed of fibers that are extremely adaptable and capable of regeneration. This characteristic of muscle fibers is made possible by a cell type called satellite cells. Adjacent to the fibers, satellite cells are found in a quiescent state and located between the muscle fibers membrane and the basal lamina. These cells are required for the growth and regeneration of skeletal muscle through myogenesis. This process is known to be tightly sequenced from the activation to the differentiation/fusion of myofibers. However, for the past fifteen years, researchers have been interested in examining satellite cell heterogeneity and have identified different subpopulations displaying distinct characteristics based on localization, quiescence state, stemness capacity, cell-cycle progression or gene expression. A small subset of satellite cells appears to represent multipotent long-term self-renewing muscle stem cells (MuSC). All these distinctions led us to the hypothesis that the characteristics of myogenesis might not be linear and therefore may be more permissive based on the evidence that satellite cells are a heterogeneous population. In this review, we discuss the different subpopulations that exist within the satellite cell pool to highlight the heterogeneity and to gain further understanding of the myogenesis progress. Finally, we discuss the long term self-renewing MuSC subpopulation that is capable of dividing asymmetrically and discuss the molecular mechanisms regulating MuSC polarization during health and disease.
Collapse
Affiliation(s)
- Corentin Guilhot
- The Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Marie Catenacci
- The Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Stephanie Lofaro
- The Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Michael A Rudnicki
- The Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
6
|
Hoh JFY. Developmental, physiologic and phylogenetic perspectives on the expression and regulation of myosin heavy chains in mammalian skeletal muscles. J Comp Physiol B 2023:10.1007/s00360-023-01499-0. [PMID: 37277594 DOI: 10.1007/s00360-023-01499-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/05/2023] [Accepted: 05/12/2023] [Indexed: 06/07/2023]
Abstract
The kinetics of myosin controls the speed and power of muscle contraction. Mammalian skeletal muscles express twelve kinetically different myosin heavy chain (MyHC) genes which provides a wide range of muscle speeds to meet different functional demands. Myogenic progenitors from diverse craniofacial and somitic mesoderm specify muscle allotypes with different repertoires for MyHC expression. This review provides a brief synopsis on the historical and current views on how cell lineage, neural impulse patterns, and thyroid hormone influence MyHC gene expression in muscles of the limb allotype during development and in adult life and the molecular mechanisms thereof. During somitic myogenesis, embryonic and foetal myoblast lineages form slow and fast primary and secondary myotube ontotypes which respond differently to postnatal neural and thyroidal influences to generate fully differentiated fibre phenotypes. Fibres of a given phenotype may arise from myotubes of different ontotypes which retain their capacity to respond differently to neural and thyroidal influences during postnatal life. This gives muscles physiological plasticity to adapt to fluctuations in thyroid hormone levels and patterns of use. The kinetics of MyHC isoforms vary inversely with animal body mass. Fast 2b fibres are specifically absent in muscles involved in elastic energy saving in hopping marsupials and generally absent in large eutherian mammals. Changes in MyHC expression are viewed in the context of the physiology of the whole animal. The roles of myoblast lineage and thyroid hormone in regulating MyHC gene expression are phylogenetically the most ancient while that of neural impulse patterns the most recent.
Collapse
Affiliation(s)
- Joseph Foon Yoong Hoh
- Discipline of Physiology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia.
- , PO Box 152, Killara, NSW, 2071, Australia.
| |
Collapse
|
7
|
Angulo M, Vacca A, Rodríguez R, Marin MN, Suárez AL, Jorge G, Nosiglia O, Cambón V, Ríos A, Iglesias M, Seija M, Escande C, Hurtado J, Briva A. Peripheral and respiratory muscle impairment during murine acute lung injury. Physiol Rep 2022; 10:e15449. [PMID: 36065875 PMCID: PMC9446397 DOI: 10.14814/phy2.15449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 08/13/2022] [Indexed: 11/24/2022] Open
Abstract
Acute respiratory distress syndrome is associated with skeletal muscle compromise, which decreases survival and impairs functional capacity. A comparative analysis of peripheral and respiratory muscles' atrophy and dysfunction in acute lung injury (ALI) has not been performed. We aimed to evaluate diaphragmatic and peripheral muscle mass and contractility in an ALI animal model. ALI was induced in C57BL/6 mice by intratracheal lipopolysaccharides instillation. Muscle mass and in vitro contractility were evaluated at different time points in hindlimb soleus (slow-twitch) and extensor digitorum longus (EDL, fast-twitch), as well as in the main respiratory muscle diaphragm. Myogenic precursor satellite cell-specific transcription factor Pax7 expression was determined by Western blot. Lung injury was associated with atrophy of the three studied muscles, although it was more pronounced and persistent in the diaphragm. Specific contractility was reduced during lung injury in EDL muscle but restored by the time lung injury has resolved. Specific force was not affected in soleus and diaphragm. A persistent increase in Pax7 expression was detected in diaphragm and EDL muscles after induction of ALI, but not in soleus muscle. Different peripheral and respiratory skeletal muscles are distinctly affected during the course of ALI. Each of the studied muscles presented a unique pattern in terms of atrophy development, contractile dysfunction and Pax7 expression.
Collapse
Affiliation(s)
- Martín Angulo
- Departamento de Fisiopatología, Hospital de Clínicas, Facultad de MedicinaUniversidad de la RepúblicaMontevideoUruguay
- Laboratorio de Exploración Funcional Respiratoria, Centro de Tratamiento Intensivo, Hospital de Clínicas, Facultad de MedicinaUniversidad de la RepúblicaMontevideoUruguay
| | - Agustina Vacca
- Departamento de Fisiopatología, Hospital de Clínicas, Facultad de MedicinaUniversidad de la RepúblicaMontevideoUruguay
| | - Romina Rodríguez
- Departamento de Fisiopatología, Hospital de Clínicas, Facultad de MedicinaUniversidad de la RepúblicaMontevideoUruguay
| | - María Noel Marin
- Departamento de Fisiopatología, Hospital de Clínicas, Facultad de MedicinaUniversidad de la RepúblicaMontevideoUruguay
| | - Ana Laura Suárez
- Departamento de Fisiopatología, Hospital de Clínicas, Facultad de MedicinaUniversidad de la RepúblicaMontevideoUruguay
| | - Gissel Jorge
- Departamento de Fisiopatología, Hospital de Clínicas, Facultad de MedicinaUniversidad de la RepúblicaMontevideoUruguay
| | - Oscar Nosiglia
- Departamento de Fisiopatología, Hospital de Clínicas, Facultad de MedicinaUniversidad de la RepúblicaMontevideoUruguay
| | - Victoria Cambón
- Departamento de Fisiopatología, Hospital de Clínicas, Facultad de MedicinaUniversidad de la RepúblicaMontevideoUruguay
| | - Anaclara Ríos
- Departamento de Fisiopatología, Hospital de Clínicas, Facultad de MedicinaUniversidad de la RepúblicaMontevideoUruguay
| | - Matías Iglesias
- Departamento de Fisiopatología, Hospital de Clínicas, Facultad de MedicinaUniversidad de la RepúblicaMontevideoUruguay
| | - Mariana Seija
- Departamento de Fisiopatología, Hospital de Clínicas, Facultad de MedicinaUniversidad de la RepúblicaMontevideoUruguay
| | - Carlos Escande
- Laboratorio de Patologías del Metabolismo y EnvejecimientoInstitut PasteurMontevideoUruguay
| | - Javier Hurtado
- Unidad de Medicina IntensivaHospital EspañolMontevideoUruguay
| | - Arturo Briva
- Cátedra de Medicina Intensiva, Hospital de Clínicas, Facultad de MedicinaUniversidad de la RepúblicaMontevideoUruguay
| |
Collapse
|
8
|
Harada A, Goto M, Kato A, Takenaka-Ninagawa N, Tanaka A, Noguchi S, Ikeya M, Sakurai H. Systemic Supplementation of Collagen VI by Neonatal Transplantation of iPSC-Derived MSCs Improves Histological Phenotype and Function of Col6-Deficient Model Mice. Front Cell Dev Biol 2021; 9:790341. [PMID: 34888314 PMCID: PMC8649773 DOI: 10.3389/fcell.2021.790341] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 10/25/2021] [Indexed: 11/25/2022] Open
Abstract
Collagen VI is distributed in the interstitium and is secreted mainly by mesenchymal stromal cells (MSCs) in skeletal muscle. Mutations in COL6A1-3 genes cause a spectrum of COL6-related myopathies. In this study, we performed a systemic transplantation study of human-induced pluripotent stem cell (iPSC)-derived MSCs (iMSCs) into neonatal immunodeficient COL6-related myopathy model (Col6a1KO/NSG) mice to validate the therapeutic potential. Engraftment of the donor cells and the resulting rescued collagen VI were observed at the quadriceps and diaphragm after intraperitoneal iMSC transplantation. Transplanted mice showed improvement in pathophysiological characteristics compared with untreated Col6a1KO/NSG mice. In detail, higher muscle regeneration in the transplanted mice resulted in increased muscle weight and enlarged myofibers. Eight-week-old mice showed increased muscle force and performed better in the grip and rotarod tests. Overall, these findings support the concept that systemic iMSC transplantation can be a therapeutic option for COL6-related myopathies.
Collapse
Affiliation(s)
- Aya Harada
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Megumi Goto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Atsuya Kato
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Nana Takenaka-Ninagawa
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Akito Tanaka
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Satoru Noguchi
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Makoto Ikeya
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Hidetoshi Sakurai
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| |
Collapse
|
9
|
Kamada Y, Toyama S, Arai Y, Inoue H, Nakagawa S, Fujii Y, Kaihara K, Kishida T, Mazda O, Takahashi K. Treadmill running prevents atrophy differently in fast- versus slow-twitch muscles in a rat model of rheumatoid arthritis. J Muscle Res Cell Motil 2021; 42:429-441. [PMID: 34687403 DOI: 10.1007/s10974-021-09610-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 10/13/2021] [Indexed: 10/20/2022]
Abstract
To investigate the effects of treadmill running on two different types of skeletal muscle, we established a rat model of collagen-induced arthritis (CIA). The skeletal muscles studied were the extensor digitorum longus (EDL), which is rich in fast-twitch muscle fibers, and the soleus, which is rich in slow-twitch muscle fibers. The histological and transcriptional changes in these muscles at 14 and 44 days after immunosensitization were compared between rats that were forced to exercise (CIA ex group) and free-reared CIA rats (CIA no group). Change in protein expression was examined on day 14 after a single bout of treadmill running. Treadmill running had different effects on the relative muscle weight and total and fiber cross-sectional areas in each muscle type. In the soleus, it prevented muscle atrophy. Transcriptional analysis revealed increased eukaryotic translation initiation factor 4E (Eif4e) expression on day 14 and increased Atrogin-1 and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) expression on day 44 in the soleus in the CIA ex group, suggesting an interaction between muscle type and exercise. A single bout of treadmill running increased the level of Eif4e and p70S6K and decreased that of Atrogin-1 in the soleus on day 14. Treadmill running prevented muscle atrophy in the soleus in a rat model of rheumatoid arthritis via activation of mitochondrial function, as evidenced by increased PGC-1α expression.
Collapse
Affiliation(s)
- Yoichiro Kamada
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Shogo Toyama
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Yuji Arai
- Department of Sports and Para-Sports Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan.
| | - Hiroaki Inoue
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Shuji Nakagawa
- Department of Sports and Para-Sports Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Yuta Fujii
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Kenta Kaihara
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Tsunao Kishida
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Osam Mazda
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Kenji Takahashi
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| |
Collapse
|
10
|
Momenzadeh S, Zamani S, Pourteymourfard-Tabrizi Z, Barreiro C, Jami MS. Muscles proteome analysis; irisin administration mimics some molecular effects of exercise in quadriceps muscle. Biochimie 2021; 189:144-157. [PMID: 34217820 DOI: 10.1016/j.biochi.2021.06.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/23/2021] [Accepted: 06/29/2021] [Indexed: 11/24/2022]
Abstract
Because of health-promoting effects, the adaptation of skeletal muscles to exercise is considered a therapeutic strategy for metabolic complications and musculoskeletal disabilities. Myokines display many beneficial effects of different exercise modalities. Among them, irisin is known as a systemic effector that positively influences several organs. There are a few studies about the effects of irisin on skeletal muscles, and irisin prosperities need to be well-defined for being an exercise mimetic. To aim this purpose, we assessed the proteome profile of mouse skeletal muscle after eight weeks of irisin injection comparing to resistance and endurance exercise treated groups. In the current study, two-dimensional gel electrophoresis was used to evaluate the protein content of the quadriceps muscle. The results were analyzed with Image Master 2D Platinum V6 software. Differentially expressed proteins were characterized by mass spectrometry (MALDI TOF/TOF) and interpreted using protein data banks and co-expression network. Irisin increases cellular ATP content by driving its overproduction through glycolysis and oxidative phosphorylation similar to two exercise protocols and as a specific property, decreases ATP consumption through creatine kinase downregulation. It also improves the microstructural properties of quadriceps muscle by increasing fiber proteins and might induce cellular proliferation and differentiation. Network analysis of differentially expressed proteins also revealed the co-expression of Irisin precursor with structural and metabolic-related proteins. The protein alterations after irisin administration display the potential of this myokine to mimic some molecular effects of exercise, suggesting it a promising candidate to improve muscle metabolism and structure.
Collapse
Affiliation(s)
- Sedigheh Momenzadeh
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Saeed Zamani
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Pourteymourfard-Tabrizi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran; QIANBIOTEC, Research and Development Center for Biotechnology, Isfahan, Iran
| | - Carlos Barreiro
- INBIOTEC (Instituto de Biotecnología de León), Avda. Real 1 - Parque Científico de León 24006, León, Spain; Departamento de Biología Molecular, Universidad de León, Campus de Ponferrada, Avda. Astorga s/n, 24401, Ponferrada, Spain
| | - Mohammad-Saeid Jami
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran; QIANBIOTEC, Research and Development Center for Biotechnology, Isfahan, Iran; Department of Neurology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA.
| |
Collapse
|
11
|
Brunetti J, Koenig S, Monnier A, Frieden M. Nanopattern surface improves cultured human myotube maturation. Skelet Muscle 2021; 11:12. [PMID: 33952323 PMCID: PMC8097894 DOI: 10.1186/s13395-021-00268-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/19/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In vitro maturation of human primary myoblasts using 2D culture remains a challenging process and leads to immature fibers with poor internal organization and function. This would however represent a valuable system to study muscle physiology or pathophysiology from patient myoblasts, at a single-cell level. METHODS Human primary myoblasts were cultured on 800-nm wide striated surface between two layers of Matrigel, and in a media supplemented with an inhibitor of TGFβ receptor. Gene expression, immunofluorescence, and Ca2+ measurements upon electrical stimulations were performed at various time points during maturation to assess the organization and function of the myotubes. RESULTS We show that after 10 days in culture, myotubes display numerous functional acetylcholine receptor clusters and express the adult isoforms of myosin heavy chain and dihydropyridine receptor. In addition, the myotubes are internally well organized with striations of α-actinin and STIM1, and occasionally ryanodine receptor 1. We also demonstrate that the myotubes present robust Ca2+ responses to repetitive electrical stimulations. CONCLUSION The present method describes a fast and efficient system to obtain well matured and functional myotubes in 2D culture allowing thorough analysis of single-cell Ca2+ signals.
Collapse
Affiliation(s)
- Jessica Brunetti
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Stéphane Koenig
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Arthur Monnier
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Maud Frieden
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
12
|
Khodabukus A. Tissue-Engineered Skeletal Muscle Models to Study Muscle Function, Plasticity, and Disease. Front Physiol 2021; 12:619710. [PMID: 33716768 PMCID: PMC7952620 DOI: 10.3389/fphys.2021.619710] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/25/2021] [Indexed: 12/20/2022] Open
Abstract
Skeletal muscle possesses remarkable plasticity that permits functional adaptations to a wide range of signals such as motor input, exercise, and disease. Small animal models have been pivotal in elucidating the molecular mechanisms regulating skeletal muscle adaptation and plasticity. However, these small animal models fail to accurately model human muscle disease resulting in poor clinical success of therapies. Here, we review the potential of in vitro three-dimensional tissue-engineered skeletal muscle models to study muscle function, plasticity, and disease. First, we discuss the generation and function of in vitro skeletal muscle models. We then discuss the genetic, neural, and hormonal factors regulating skeletal muscle fiber-type in vivo and the ability of current in vitro models to study muscle fiber-type regulation. We also evaluate the potential of these systems to be utilized in a patient-specific manner to accurately model and gain novel insights into diseases such as Duchenne muscular dystrophy (DMD) and volumetric muscle loss. We conclude with a discussion on future developments required for tissue-engineered skeletal muscle models to become more mature, biomimetic, and widely utilized for studying muscle physiology, disease, and clinical use.
Collapse
Affiliation(s)
- Alastair Khodabukus
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
13
|
Hoh JFY. Myosin heavy chains in extraocular muscle fibres: Distribution, regulation and function. Acta Physiol (Oxf) 2021; 231:e13535. [PMID: 32640094 DOI: 10.1111/apha.13535] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022]
Abstract
This review examines kinetic properties and distribution of the 11 isoforms of myosin heavy chain (MyHC) expressed in extraocular muscle (EOM) fibre types and the regulation and function of these MyHCs. Although recruitment and discharge characteristics of ocular motoneurons during fixation and eye movements are well documented, work directly linking these properties with motor unit contractile speed and MyHC composition is lacking. Recruitment of motor units according to Henneman's size principle has some support in EOMs but needs consolidation. Both neurogenic and myogenic mechanisms regulate MyHC expression as in other muscle allotypes. Developmentally, multiply-innervated (MIFs) and singly-innervated fibres (SIFs) are derived presumably from distinct myoblast lineages, ending up expressing MyHCs in the slow and fast ends of the kinetic spectrum respectively. They modulate the synaptic inputs of their motoneurons through different retrogradely transported neurotrophins, thereby specifying their tonic and phasic impulse patterns. Immunohistochemical analyses of EOMs regenerating in situ and in limb muscle beds suggest that the very impulse patterns driving various ocular movements equip effectors with appropriate MyHC compositions and speeds to accomplish their tasks. These experiments also suggest that satellite cells of SIFs and MIFs are distinct lineages expressing different MyHCs during regeneration. MyHC compositions and functional characteristics of orbital fibres show longitudinal variations that facilitate linear ocular rotation during saccades. Palisade endings on global MIFs are postulated to respond to active and passive tensions by triggering axon reflexes that play important roles during fixation, saccades and vergence. How EOMs implement Listings law during ocular rotation is discussed.
Collapse
Affiliation(s)
- Joseph F. Y. Hoh
- Discipline of Physiology and the Bosch Institute School of Medical Sciences Faculty of Medicine and Health The University of Sydney Sydney NSW Australia
| |
Collapse
|
14
|
Li Y, Cheng JX, Yang HH, Chen LP, Liu FJ, Wu Y, Fan M, Wu HT. Transferrin receptor 1 plays an important role in muscle development and denervation-induced muscular atrophy. Neural Regen Res 2021; 16:1308-1316. [PMID: 33318410 PMCID: PMC8284266 DOI: 10.4103/1673-5374.301024] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Previous studies demonstrate an accumulation of transferrin and transferrin receptor 1 (TfR1) in regenerating peripheral nerves. However, the expression and function of transferrin and TfR1 in the denervated skeletal muscle remain poorly understood. In this study, a mouse model of denervation was produced by complete tear of the left brachial plexus nerve. RNA-sequencing revealed that transferrin expression in the denervated skeletal muscle was upregulated, while TfR1 expression was downregulated. We also investigated the function of TfR1 during development and in adult skeletal muscles in mice with inducible deletion or loss of TfR1. The ablation of TfR1 in skeletal muscle in early development caused severe muscular atrophy and early death. In comparison, deletion of TfR1 in adult skeletal muscles did not affect survival or glucose metabolism, but caused skeletal muscle atrophy and motor functional impairment, similar to the muscular atrophy phenotype observed after denervation. These findings suggest that TfR1 plays an important role in muscle development and denervation-induced muscular atrophy. This study was approved by the Institutional Animal Care and Use Committee of Beijing Institute of Basic Medical Sciences, China (approval No. SYXK 2017-C023) on June 1, 2018.
Collapse
Affiliation(s)
- Ying Li
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Juan-Xian Cheng
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Hai-Hong Yang
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing; Department of Anesthesiology, the General Hospital of Western Theater Command, Chengdu, Sichuan Province, China
| | - Li-Ping Chen
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Feng-Jiao Liu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yan Wu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Ming Fan
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences; Chinese Institute for Brain Research (CIBR), Beijing, China
| | - Hai-Tao Wu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences; Chinese Institute for Brain Research (CIBR), Beijing; Key Laboratory of Neuroregeneration, Coinnovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
15
|
Simvastatin Enhances Muscle Regeneration Through Autophagic Defect-Mediated Inflammation and mTOR Activation in G93ASOD1 Mice. Mol Neurobiol 2020; 58:1593-1606. [PMID: 33222146 DOI: 10.1007/s12035-020-02216-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis is a fatal neurodegenerative disease characterised by the selective loss of motor neurons, muscular atrophy, and degeneration. Statins, as 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors, are the most widely prescribed drugs to lower cholesterol levels and used for the treatment of cardiovascular and cerebrovascular diseases. However, statins are seldom used in muscular diseases, primarily because of their rare statin-associated myopathy. Recently, statins have been shown to reduce muscular damage and improve its function. Here, we investigated the role of statins in myopathy using G93ASOD1 mice. Our results indicated that simvastatin significantly increased the autophagic flux defect and increased inflammation in the skeletal muscles of G93ASOD1 mice. We also found that increased inflammation correlated with aggravated muscle atrophy and fibrosis. Nevertheless, long-term simvastatin treatment promoted the regeneration of damaged muscle by activating the mammalian target of rapamycin pathway. However, administration of simvastatin did not impede vast muscle degeneration and movement dysfunction resulting from the enhanced progressive impairment of the neuromuscular junction. Together, our findings highlighted that simvastatin exacerbated skeletal muscle atrophy and denervation in spite of promoting myogenesis in damaged muscle, providing new insights into the selective use of statin-induced myopathy in ALS.
Collapse
|
16
|
Dos Santos M, Backer S, Saintpierre B, Izac B, Andrieu M, Letourneur F, Relaix F, Sotiropoulos A, Maire P. Single-nucleus RNA-seq and FISH identify coordinated transcriptional activity in mammalian myofibers. Nat Commun 2020; 11:5102. [PMID: 33037211 PMCID: PMC7547110 DOI: 10.1038/s41467-020-18789-8] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 09/10/2020] [Indexed: 12/20/2022] Open
Abstract
Skeletal muscle fibers are large syncytia but it is currently unknown whether gene expression is coordinately regulated in their numerous nuclei. Here we show by snRNA-seq and snATAC-seq that slow, fast, myotendinous and neuromuscular junction myonuclei each have different transcriptional programs, associated with distinct chromatin states and combinations of transcription factors. In adult mice, identified myofiber types predominantly express either a slow or one of the three fast isoforms of Myosin heavy chain (MYH) proteins, while a small number of hybrid fibers can express more than one MYH. By snRNA-seq and FISH, we show that the majority of myonuclei within a myofiber are synchronized, coordinately expressing only one fast Myh isoform with a preferential panel of muscle-specific genes. Importantly, this coordination of expression occurs early during post-natal development and depends on innervation. These findings highlight a previously undefined mechanism of coordination of gene expression in a syncytium.
Collapse
Affiliation(s)
| | - Stéphanie Backer
- Université de Paris, Institut Cochin, INSERM, CNRS., 75014, Paris, France
| | | | - Brigitte Izac
- Université de Paris, Institut Cochin, INSERM, CNRS., 75014, Paris, France
| | - Muriel Andrieu
- Université de Paris, Institut Cochin, INSERM, CNRS., 75014, Paris, France
| | - Franck Letourneur
- Université de Paris, Institut Cochin, INSERM, CNRS., 75014, Paris, France
| | - Frederic Relaix
- Université Paris-Est Creteil, INSERM U955 IMRB., 94000, Creteil, France
| | | | - Pascal Maire
- Université de Paris, Institut Cochin, INSERM, CNRS., 75014, Paris, France.
| |
Collapse
|
17
|
Bidirectional myofiber transition through altering the photobiomodulation condition. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2020; 212:112041. [PMID: 33002778 DOI: 10.1016/j.jphotobiol.2020.112041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 08/09/2020] [Accepted: 09/22/2020] [Indexed: 11/20/2022]
Abstract
Despite remarkable advancements in modern medicine, muscular atrophy remains as an unsolved problem. It is well known that pathological characteristics of different atrophy types could vary according to the pathophysiological causes. In fact, the lesion of atrophy is not always homogenously distributed but often predominantly evident in either fast or slow myofibers. As the focalization of the atrophic lesions, the existence and the functional impairment of each fast and slow progenitor/satellite cell (SC) are suspected though there are still controversies about this hypothesis. In this study, we isolated Pax7 positive (Pax7+ve) SCs from the tibia anterior (fast) and soleus (slow) muscles respectively and successfully demonstrated, for the first time, the difference between optimal exposure durations of photobiomodulation (PBM) which was known as low level laser irradiation (LLLI) in promoting proliferation of Pax7+ve SC which were acquired from fast and slow muscles respectively. Moreover, a hypertrophy-accompanied bidirectional change in myofiber composition with neuromuscular junction alteration, either from slow to fast or fast to slow, were achieved by applying different PBM durations. Simultaneously, PBM exhibited a synergistic effect with muscle exercise on the increase in myofiber size. Our data suggested the existence of at least two different populations of Pax7+ve SC which possess distinct sensitivities towards PBM. As our data revealed the capability of PBM in bidirectional changes of skeletal muscle composition and neuromuscular junction constitution thereby strengthen its contractility through altering the irradiation condition, we believe PBM showed the potential to be as a promising clinical treatment for muscular atrophy.
Collapse
|
18
|
Skeletal Muscle Tissue Engineering: Biomaterials-Based Strategies for the Treatment of Volumetric Muscle Loss. Bioengineering (Basel) 2020; 7:bioengineering7030085. [PMID: 32751847 PMCID: PMC7552659 DOI: 10.3390/bioengineering7030085] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/17/2020] [Accepted: 07/28/2020] [Indexed: 12/13/2022] Open
Abstract
Millions of Americans suffer from skeletal muscle injuries annually that can result in volumetric muscle loss (VML), where extensive musculoskeletal damage and tissue loss result in permanent functional deficits. In the case of small-scale injury skeletal muscle is capable of endogenous regeneration through activation of resident satellite cells (SCs). However, this is greatly reduced in VML injuries, which remove native biophysical and biochemical signaling cues and hinder the damaged tissue's ability to direct regeneration. The current clinical treatment for VML is autologous tissue transfer, but graft failure and scar tissue formation leave patients with limited functional recovery. Tissue engineering of instructive biomaterial scaffolds offers a promising approach for treating VML injuries. Herein, we review the strategic engineering of biophysical and biochemical cues in current scaffold designs that aid in restoring function to these preclinical VML injuries. We also discuss the successes and limitations of the three main biomaterial-based strategies to treat VML injuries: acellular scaffolds, cell-delivery scaffolds, and in vitro tissue engineered constructs. Finally, we examine several innovative approaches to enhancing the design of the next generation of engineered scaffolds to improve the functional regeneration of skeletal muscle following VML injuries.
Collapse
|
19
|
Maire P, Dos Santos M, Madani R, Sakakibara I, Viaut C, Wurmser M. Myogenesis control by SIX transcriptional complexes. Semin Cell Dev Biol 2020; 104:51-64. [PMID: 32247726 DOI: 10.1016/j.semcdb.2020.03.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 02/07/2023]
Abstract
SIX homeoproteins were first described in Drosophila, where they participate in the Pax-Six-Eya-Dach (PSED) network with eyeless, eyes absent and dachsund to drive synergistically eye development through genetic and biochemical interactions. The role of the PSED network and SIX proteins in muscle formation in vertebrates was subsequently identified. Evolutionary conserved interactions with EYA and DACH proteins underlie the activity of SIX transcriptional complexes (STC) both during embryogenesis and in adult myofibers. Six genes are expressed throughout muscle development, in embryonic and adult proliferating myogenic stem cells and in fetal and adult post-mitotic myofibers, where SIX proteins regulate the expression of various categories of genes. In vivo, SIX proteins control many steps of muscle development, acting through feedforward mechanisms: in the embryo for myogenic fate acquisition through the direct control of Myogenic Regulatory Factors; in adult myofibers for their contraction/relaxation and fatigability properties through the control of genes involved in metabolism, sarcomeric organization and calcium homeostasis. Furthermore, during development and in the adult, SIX homeoproteins participate in the genesis and the maintenance of myofibers diversity.
Collapse
Affiliation(s)
- Pascal Maire
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014, Paris, France.
| | | | - Rouba Madani
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014, Paris, France
| | - Iori Sakakibara
- Research Center for Advanced Science and Technology, The University of Tokyo, Japan
| | - Camille Viaut
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014, Paris, France
| | - Maud Wurmser
- Department of Integrative Medical Biology (IMB), Umeå universitet, Sweden
| |
Collapse
|
20
|
Militello G, Hosen MR, Ponomareva Y, Gellert P, Weirick T, John D, Hindi SM, Mamchaoui K, Mouly V, Döring C, Zhang L, Nakamura M, Kumar A, Fukada SI, Dimmeler S, Uchida S. A novel long non-coding RNA Myolinc regulates myogenesis through TDP-43 and Filip1. J Mol Cell Biol 2019; 10:102-117. [PMID: 29618024 DOI: 10.1093/jmcb/mjy025] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 03/29/2018] [Indexed: 12/15/2022] Open
Abstract
Myogenesis is a complex process required for skeletal muscle formation during embryonic development and for regeneration and growth of myofibers in adults. Accumulating evidence suggests that long non-coding RNAs (lncRNAs) play key roles in regulating cell fate decision and function in various tissues. However, the role of lncRNAs in the regulation of myogenesis remains poorly understood. In this study, we identified a novel muscle-enriched lncRNA called 'Myolinc (AK142388)', which we functionally characterized in the C2C12 myoblast cell line. Myolinc is predominately localized in the nucleus, and its levels increase upon induction of the differentiation. Knockdown of Myolinc impairs the expression of myogenic regulatory factors and formation of multi-nucleated myotubes in cultured myoblasts. Myolinc also regulates the expression of Filip1 in a cis-manner. Similar to Myolinc, knockdown of Filip1 inhibits myogenic differentiation. Furthermore, Myolinc binds to TAR DNA-binding protein 43 (TDP-43), a DNA/RNA-binding protein that regulates the expression of muscle genes (e.g. Acta1 and MyoD). Knockdown of TDP-43 inhibits myogenic differentiation. We also show that Myolinc-TDP-43 interaction is essential for the binding of TDP-43 to the promoter regions of muscle marker genes. Finally, we show that silencing of Myolinc inhibits skeletal muscle regeneration in adult mice. Altogether, our study identifies a novel lncRNA that controls key regulatory networks of myogenesis.
Collapse
Affiliation(s)
- Giuseppe Militello
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt am Main 60590, Germany.,German Center for Cardiovascular Research, Partner side Rhein-Main, Frankfurt am Main 60590, Germany.,Department of Biosciences, Goethe University Frankfurt, Frankfurt am Main 60438, Germany.,Cardiovascular Innovation Institute, University of Louisville, Louisville, KY 40202, USA
| | - Mohammed Rabiul Hosen
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt am Main 60590, Germany.,German Center for Cardiovascular Research, Partner side Rhein-Main, Frankfurt am Main 60590, Germany.,Department of Biosciences, Goethe University Frankfurt, Frankfurt am Main 60438, Germany
| | - Yuliya Ponomareva
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt am Main 60590, Germany.,German Center for Cardiovascular Research, Partner side Rhein-Main, Frankfurt am Main 60590, Germany.,Department of Biosciences, Goethe University Frankfurt, Frankfurt am Main 60438, Germany
| | - Pascal Gellert
- Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London SW3 6JB, UK
| | - Tyler Weirick
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt am Main 60590, Germany.,German Center for Cardiovascular Research, Partner side Rhein-Main, Frankfurt am Main 60590, Germany.,Department of Biosciences, Goethe University Frankfurt, Frankfurt am Main 60438, Germany.,Cardiovascular Innovation Institute, University of Louisville, Louisville, KY 40202, USA
| | - David John
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt am Main 60590, Germany.,German Center for Cardiovascular Research, Partner side Rhein-Main, Frankfurt am Main 60590, Germany.,Department of Biosciences, Goethe University Frankfurt, Frankfurt am Main 60438, Germany
| | - Sajedah Mahmoud Hindi
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Kamel Mamchaoui
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Paris 75013, France
| | - Vincent Mouly
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Paris 75013, France
| | - Claudia Döring
- Dr. Senckenberg Institute of Pathology, Goethe University Frankfurt, Frankfurt am Main 60590, Germany
| | - Lidan Zhang
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Miki Nakamura
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Ashok Kumar
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - So-Ichiro Fukada
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Stefanie Dimmeler
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt am Main 60590, Germany.,German Center for Cardiovascular Research, Partner side Rhein-Main, Frankfurt am Main 60590, Germany
| | - Shizuka Uchida
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt am Main 60590, Germany.,German Center for Cardiovascular Research, Partner side Rhein-Main, Frankfurt am Main 60590, Germany.,Cardiovascular Innovation Institute, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
21
|
Deficient Skeletal Muscle Regeneration after Injury Induced by a Clostridium perfringens Strain Associated with Gas Gangrene. Infect Immun 2019; 87:IAI.00200-19. [PMID: 31138614 DOI: 10.1128/iai.00200-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 05/17/2019] [Indexed: 02/06/2023] Open
Abstract
Gas gangrene, or clostridial myonecrosis, is usually caused by Clostridium perfringens and may occur spontaneously in association with diabetes mellitus, peripheral vascular disease, or some malignancies but more often after contamination of a deep surgical or traumatic lesion. If not controlled, clostridial myonecrosis results in multiorgan failure, shock, and death, but very little is known about the muscle regeneration process that follows myonecrosis when the infection is controlled. In this study, we characterized the muscle regeneration process after myonecrosis caused in a murine experimental infection with a sublethal inoculum of C. perfringens vegetative cells. The results show that myonecrosis occurs concomitantly with significant vascular injury, which limits the migration of inflammatory cells. A significant increase in cytokines that promote inflammation explains the presence of an inflammatory infiltrate; however, impaired interferon gamma (IFN-γ) expression, a reduced number of M1 macrophages, deficient phagocytic activity, and a prolongation of the permanence of inflammatory cells lead to deficient muscle regeneration. The expression of transforming growth factor β1 (TGF-β1) agrees with the consequent accumulation of collagen in the muscle, i.e., fibrosis observed 30 days after infection. These results provide new information on the pathogenesis of gas gangrene caused by C. perfringens, shed light on the basis of the deficient muscle regenerative activity, and may open new perspectives for the development of novel therapies for patients suffering from this disease.
Collapse
|
22
|
D'Lugos AC, Fry CS, Ormsby JC, Sweeney KR, Brightwell CR, Hale TM, Gonzales RJ, Angadi SS, Carroll CC, Dickinson JM. Chronic doxorubicin administration impacts satellite cell and capillary abundance in a muscle-specific manner. Physiol Rep 2019; 7:e14052. [PMID: 30963722 PMCID: PMC6453819 DOI: 10.14814/phy2.14052] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 03/15/2019] [Accepted: 03/16/2019] [Indexed: 01/23/2023] Open
Abstract
Anthracycline chemotherapies are effective at reducing disease recurrence and mortality in cancer patients. However, these drugs also contribute to skeletal muscle wasting and dysfunction. The purpose of this study was to assess the impact of chronic doxorubicin (DOX) administration on satellite cell and capillary densities in different skeletal muscles. We hypothesized that DOX would reduce satellite cell and capillary densities of the soleus (SOL) and extensor digitorum longus (EDL) muscles, along with muscle fiber size. Ovariectomized female Sprague-Dawley rats were randomized to receive three bi-weekly intraperitoneal injections of DOX (4 mg∙kg-1 ; cumulative dose 12 mg∙kg-1 ) or vehicle (VEH; saline). Animals were euthanized 5d following the last injection and the SOL and EDL were dissected and prepared for immunohistochemical and RT-qPCR analyses. Relative to VEH, CSA of the SOL and EDL fibers were 26% and 33% smaller, respectively, in DOX (P < 0.05). In the SOL, satellite cell and capillary densities were 39% and 35% lower, respectively, in DOX (P < 0.05), whereas in the EDL satellite cell and capillary densities were unaffected by DOX administration (P > 0.05). Proliferating satellite cells were unaffected by DOX in the SOL (P > 0.05). In the SOL, MYF5 mRNA expression was increased in DOX (P < 0.05), while in the EDL MGF mRNA expression was reduced in DOX (P < 0.05). Chronic DOX administration is associated with reduced fiber size in the SOL and EDL; however, DOX appeared to reduce satellite cell and capillary densities only in the SOL. These findings highlight that therapeutic targets to protect skeletal muscle from DOX may vary across muscles.
Collapse
Affiliation(s)
| | - Christopher S. Fry
- Department of Nutrition and MetabolismUniversity of Texas Medical BranchGalvestonTexas
| | - Jordan C. Ormsby
- College of Health SolutionsArizona State UniversityPhoenixArizona
| | | | - Camille R. Brightwell
- Department of Nutrition and MetabolismUniversity of Texas Medical BranchGalvestonTexas
| | - Taben M. Hale
- Department of Basic Medical SciencesCollege of Medicine‐PhoenixUniversity of ArizonaPhoenixArizona
| | - Rayna J. Gonzales
- Department of Basic Medical SciencesCollege of Medicine‐PhoenixUniversity of ArizonaPhoenixArizona
| | | | - Chad C. Carroll
- Department of PhysiologyMidwestern UniversityGlendaleArizona
- Department of Health and KinesiologyPurdue UniversityWest LafayetteIndiana
| | | |
Collapse
|
23
|
Linard C, Brachet M, L’homme B, Strup-Perrot C, Busson E, Bonneau M, Lataillade JJ, Bey E, Benderitter M. Long-term effectiveness of local BM-MSCs for skeletal muscle regeneration: a proof of concept obtained on a pig model of severe radiation burn. Stem Cell Res Ther 2018; 9:299. [PMID: 30409227 PMCID: PMC6225585 DOI: 10.1186/s13287-018-1051-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/10/2018] [Accepted: 10/16/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Medical management of the severe musculocutaneous radiation syndrome involves surgical intervention with debridement of necrotic tissue. Even when skin excision is replaced by specific plastic surgery, treatment of the muscle radiation injury nonetheless remains difficult, for it involves a massive muscle defect in an unpredictable environment, subject to inflammatory waves weeks to months after irradiation, which delay healing and predispose the patient to the development of fibrous scar tissue. In this study, we investigated the long-term effect of local injections of bone marrow-derived mesenchymal stromal cells (BM-MSCs), combined with plastic surgery, to treat muscle necrosis in a large animal model. METHODS Three months after irradiation to the rump, minipigs were treated by excision of necrotic muscle tissue, vascularized flap surgery, and four injections with or without local autologous BM-MSCs, performed weekly. The quality of the muscle wound healing was examined 1 year post-surgery. RESULTS The skeletal muscle surgery without MSC treatment led to permanent deposition of collagen 1 and 3, decreased myofiber diameter, failed muscle fiber regeneration, a reduced number of capillaries, and the accumulation of high calcium and fat. In animals treated by surgery and MSC injections, these indicators were substantially better and demonstrated established regeneration. MSC therapy acts at several levels by stimulating growth factors such as VEGF, which is involved in angiogenesis and satellite cell pool maintenance, and creating a macrophage M1/M2 balance. CONCLUSION Thus, cell therapy using BM-MSCs is an effective and safe way to improve recovery of irradiation-induced skeletal muscle damage without signs of long-term degeneration.
Collapse
Affiliation(s)
- Christine Linard
- Institute of Radiological Protection and Nuclear Safety, B.P. n°17, F-92262 Fontenay-aux-Roses, France
- Unité de Thérapie Tissulaire et Traumatologie de Guerre, Institut de Recherche Biomédicale des Armées, Clamart, France
| | - Michel Brachet
- Department of Plastic Surgery, Military Hospital of Percy, Clamart, France
| | - Bruno L’homme
- Institute of Radiological Protection and Nuclear Safety, B.P. n°17, F-92262 Fontenay-aux-Roses, France
| | - Carine Strup-Perrot
- Institute of Radiological Protection and Nuclear Safety, B.P. n°17, F-92262 Fontenay-aux-Roses, France
| | - Elodie Busson
- Unité des Médicaments de Thérapie Innovante, Centre de Transfusion Sanguine des Armées, Clamart, France
| | - Michel Bonneau
- Centre of Research in Interventional Imaging, National Institut of Agronomic Research, Jouy-en-Josas, France
| | - Jean-Jacques Lataillade
- Unité de Thérapie Tissulaire et Traumatologie de Guerre, Institut de Recherche Biomédicale des Armées, Clamart, France
| | - Eric Bey
- Department of Plastic Surgery, Military Hospital of Percy, Clamart, France
| | - Marc Benderitter
- Institute of Radiological Protection and Nuclear Safety, B.P. n°17, F-92262 Fontenay-aux-Roses, France
| |
Collapse
|
24
|
Schiaffino S. Muscle fiber type diversity revealed by anti-myosin heavy chain antibodies. FEBS J 2018; 285:3688-3694. [PMID: 29761627 DOI: 10.1111/febs.14502] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 04/24/2018] [Accepted: 05/08/2018] [Indexed: 01/02/2023]
Abstract
Different forms of myosin heavy chains (MyHCs), coded by a large family of sarcomeric MYH genes, are expressed in striated muscles. The generation of specific anti-MyHC antibodies has provided a powerful tool to define the fiber types present in skeletal muscles, their functional properties, their response to conditions that affect muscle plasticity and their changes in muscle disorders. Cardiomyocyte heterogeneity has been revealed by the serendipitous observation that different MyHCs are present in atrial and ventricular myocardium and in heart conduction tissue. Developmental MyHCs present in embryonic and fetal/neonatal skeletal muscle are re-expressed during muscle regeneration and can be used to identify regenerating fibers in muscle diseases. MyHC isoforms provide cell type-specific markers to identify the signaling pathways that control muscle cell identity and are an essential reference to interpret the results of single-cell transcriptomics and proteomics.
Collapse
|
25
|
Potential therapeutic targets for ALS: MIR206, MIR208b and MIR499 are modulated during disease progression in the skeletal muscle of patients. Sci Rep 2017; 7:9538. [PMID: 28842714 PMCID: PMC5573384 DOI: 10.1038/s41598-017-10161-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/03/2017] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by the progressive loss of motor neurons followed by muscle weakness, paralysis and death. The disease progression is extremely variable among patients, and reliable prognostic markers have not been identified. The aim of the study was to functionally characterize selected genes and microRNAs acting in the skeletal muscle of ALS patients, taking into account the duration and evolution of the disease, in order to obtain information regarding the muscle response to ALS progression. This prospective, longitudinal study enrolled 14 ALS patients and 24 age- and sex-matched healthy controls. Gene expression and histological analysis indicated an increase of MIR208B and MIR499 levels and the predominance of slow fibres, respectively, in the muscles of patients with a slower disease progression. A decreased expression of MIR206 and increased levels of HDAC4, during the progression of the disease were also observed. Taken together, our data suggest that the molecular signalling that regulates re-innervation and muscle regeneration is hampered during the progression of skeletal muscle impairment in ALS. This could provide precious hints towards defining prognostic protocols, and designing novel tailored therapeutic approaches, to improve ALS patients’ care and delay disease progression.
Collapse
|
26
|
Pini V, Morgan JE, Muntoni F, O’Neill HC. Genome Editing and Muscle Stem Cells as a Therapeutic Tool for Muscular Dystrophies. CURRENT STEM CELL REPORTS 2017; 3:137-148. [PMID: 28616376 PMCID: PMC5445179 DOI: 10.1007/s40778-017-0076-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Purpose of Review Muscular dystrophies are a group of severe degenerative disorders characterized by muscle fiber degeneration and death. Therapies designed to restore muscle homeostasis and to replace dying fibers are being experimented, but none of those in clinical trials are suitable to permanently address individual gene mutation. The purpose of this review is to discuss genome editing tools such as CRISPR/Cas (clustered regularly interspaced short palindromic repeats/CRISPR-associated), which enable direct sequence alteration and could potentially be adopted to correct the genetic defect leading to muscle impairment. Recent Findings Recent findings show that advances in gene therapy, when combined with traditional viral vector-based approaches, are bringing the field of regenerative medicine closer to precision-based medicine. Summary The use of such programmable nucleases is proving beneficial for the creation of more accurate in vitro and in vivo disease models. Several gene and cell-therapy studies have been performed on satellite cells, the primary skeletal muscle stem cells involved in muscle regeneration. However, these have mainly been based on artificial replacement or augmentation of the missing protein. Satellite cells are a particularly appealing target to address these innovative technologies for the treatment of muscular dystrophies.
Collapse
Affiliation(s)
- Veronica Pini
- Molecular and Developmental Neurosciences Program, The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH UK
| | - Jennifer E. Morgan
- Molecular and Developmental Neurosciences Program, The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH UK
| | - Francesco Muntoni
- Molecular and Developmental Neurosciences Program, The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH UK
| | - Helen C. O’Neill
- Embryology, IVF and Reproductive Genetics Group, Institute for Women’s Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX UK
| |
Collapse
|
27
|
Sakakibara I, Wurmser M, Dos Santos M, Santolini M, Ducommun S, Davaze R, Guernec A, Sakamoto K, Maire P. Six1 homeoprotein drives myofiber type IIA specialization in soleus muscle. Skelet Muscle 2016; 6:30. [PMID: 27597886 PMCID: PMC5011358 DOI: 10.1186/s13395-016-0102-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 08/16/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Adult skeletal muscles are composed of slow and fast myofiber subtypes which each express selective genes required for their specific contractile and metabolic activity. Six homeoproteins are transcription factors regulating muscle cell fate through activation of myogenic regulatory factors and driving fast-type gene expression during embryogenesis. RESULTS We show here that Six1 protein accumulates more robustly in the nuclei of adult fast-type muscles than in adult slow-type muscles, this specific enrichment takes place during perinatal growth. Deletion of Six1 in soleus impaired fast-type myofiber specialization during perinatal development, resulting in a slow phenotype and a complete lack of Myosin heavy chain 2A (MyHCIIA) expression. Global transcriptomic analysis of wild-type and Six1 mutant myofibers identified the gene networks controlled by Six1 in adult soleus muscle. This analysis showed that Six1 is required for the expression of numerous genes encoding fast-type sarcomeric proteins, glycolytic enzymes and controlling intracellular calcium homeostasis. Parvalbumin, a key player of calcium buffering, in particular, is a direct target of Six1 in the adult myofiber. CONCLUSIONS This analysis revealed that Six1 controls distinct aspects of adult muscle physiology in vivo, and acts as a main determinant of fast-fiber type acquisition and maintenance.
Collapse
Affiliation(s)
- Iori Sakakibara
- INSERM U1016, Institut Cochin, Paris, 75014 France
- CNRS UMR 8104, Paris, 75014 France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, 75014 France
- Division of Integrative Pathophysiology, Proteo-Science Center, Graduate School of Medicine, Ehime University, Ehime, Japan
| | - Maud Wurmser
- INSERM U1016, Institut Cochin, Paris, 75014 France
- CNRS UMR 8104, Paris, 75014 France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, 75014 France
| | - Matthieu Dos Santos
- INSERM U1016, Institut Cochin, Paris, 75014 France
- CNRS UMR 8104, Paris, 75014 France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, 75014 France
| | - Marc Santolini
- Laboratoire de Physique Statistique, CNRS, Université P. et M. Curie, Université D. Diderot, École Normale Supérieure, Paris, 75005 France
| | - Serge Ducommun
- Nestlé Institute of Health Sciences SA, EPFL Innovation Park, Lausanne, Switzerland
| | - Romain Davaze
- INSERM U1016, Institut Cochin, Paris, 75014 France
- CNRS UMR 8104, Paris, 75014 France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, 75014 France
| | - Anthony Guernec
- INSERM U1016, Institut Cochin, Paris, 75014 France
- CNRS UMR 8104, Paris, 75014 France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, 75014 France
| | - Kei Sakamoto
- Nestlé Institute of Health Sciences SA, EPFL Innovation Park, Lausanne, Switzerland
| | - Pascal Maire
- INSERM U1016, Institut Cochin, Paris, 75014 France
- CNRS UMR 8104, Paris, 75014 France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, 75014 France
| |
Collapse
|
28
|
Qaisar R, Bhaskaran S, Van Remmen H. Muscle fiber type diversification during exercise and regeneration. Free Radic Biol Med 2016; 98:56-67. [PMID: 27032709 DOI: 10.1016/j.freeradbiomed.2016.03.025] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 03/01/2016] [Accepted: 03/24/2016] [Indexed: 01/15/2023]
Abstract
The plasticity of skeletal muscle can be traced down to extensive metabolic, structural and molecular remodeling at the single fiber level. Skeletal muscle is comprised of different fiber types that are the basis of muscle plasticity in response to various functional demands. Resistance and endurance exercises are two external stimuli that differ in their duration and intensity of contraction and elicit markedly different responses in muscles adaptation. Further, eccentric contractions that are associated with exercise-induced injuries, elicit varied muscle adaptation and regenerative responses. Most adaptive changes are fiber type-specific and are highly influenced by diverse structural, metabolic and functional characteristics of individual fiber types. Regulation of signaling pathways by reactive oxygen species (ROS) and oxidative stress also plays an important role in muscle fiber adaptation during exercise. This review focuses on cellular and molecular responses that regulate the adaptation of skeletal muscle to exercise and exercise-related injuries.
Collapse
Affiliation(s)
- Rizwan Qaisar
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA
| | - Shylesh Bhaskaran
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA
| | - Holly Van Remmen
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA.
| |
Collapse
|
29
|
Stölting MNL, Arnold AS, Haralampieva D, Handschin C, Sulser T, Eberli D. Magnetic stimulation supports muscle and nerve regeneration after trauma in mice. Muscle Nerve 2016. [PMID: 26202157 DOI: 10.1002/mus.24780] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Magnetic stimulation (MS) has the ability to induce muscle twitch and has long been proposed as a therapeutic modality for skeletal muscle diseases. However, the molecular mechanisms underlying its means of action have not been defined. METHODS Muscle regeneration after trauma was studied in a standard muscle injury mouse model. The influence of MS on the formation of motor units, posttrauma muscle/nerve regeneration, and vascularization was investigated. RESULTS We found that MS does not cause systemic or muscle damage but improves muscle regeneration by significantly minimizing the presence of inflammatory infiltrate and formation of scars after trauma. It avoids posttrauma muscle atrophy, induces muscle hypertrophy, and increases the metabolism and turnover of muscle. It triples the expression of muscle markers and significantly improves muscle functional recovery after trauma. CONCLUSIONS Our results indicate that MS supports muscle and nerve regeneration by activating muscle-nerve cross-talk and inducing the maturation of neuromuscular junctions.
Collapse
Affiliation(s)
- Meline N L Stölting
- Laboratory for Urologic Tissue Engineering and Stem Cell Therapy, Division of Urology, University of Zurich, Frauenklinikstrasse 10, CH 8091, Zurich, Switzerland
| | - Anne Sophie Arnold
- Biozentrum, Focal Area Growth and Development, University of Basel, Basel, Switzerland
| | - Deana Haralampieva
- Laboratory for Urologic Tissue Engineering and Stem Cell Therapy, Division of Urology, University of Zurich, Frauenklinikstrasse 10, CH 8091, Zurich, Switzerland
| | - Christoph Handschin
- Biozentrum, Focal Area Growth and Development, University of Basel, Basel, Switzerland
| | - Tullio Sulser
- Laboratory for Urologic Tissue Engineering and Stem Cell Therapy, Division of Urology, University of Zurich, Frauenklinikstrasse 10, CH 8091, Zurich, Switzerland
| | - Daniel Eberli
- Laboratory for Urologic Tissue Engineering and Stem Cell Therapy, Division of Urology, University of Zurich, Frauenklinikstrasse 10, CH 8091, Zurich, Switzerland
| |
Collapse
|
30
|
Umezawa T, Higa K, Serikawa M, Yamamoto M, Matsunaga S, Shimazaki J, Abe S. Proliferative activity of skeletal myoblast sheet by paracrine effects of mesenchymal stem cells. J Oral Biosci 2016; 58:158-166. [PMID: 32512684 DOI: 10.1016/j.job.2016.05.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/11/2016] [Accepted: 05/12/2016] [Indexed: 01/09/2023]
Abstract
OBJECTIVES The purpose of this study was to examine the proliferative activity enhancement of skeletal myoblasts in the presence and absence of mesenchymal stem cells (MSC). METHODS We artificially fabricated two types of cell sheets by co-culturing rabbit skeletal myoblast sheets with rabbit MSCs using type IA collagen gel (MC+), and cell sheets without rabbit MSCs (MC-). RESULTS The results of hematoxylin and eosin staining revealed that MC+ was thicker than MC- on day 7. Immunohistochemical staining revealed a low level of desmin expression in both sheets on day 2. Desmin expression increased at days 7 and 12, and desmin localization was consistent with the stratified area. Reverse transcription PCR revealed the presence of MyoD and PAX7 in both sheets on days 2, 7, and 12. The presence of myogenin was confirmed in both sheets on days 7 and 12. Hepatocyte growth factor expression was evident in MC+ on day 2, and in both sheets on day 7. Measurement of cell proliferative activity based on DNA cell cycle analysis indicated that MC+ had significantly higher cell proliferative activity than MC- on day 7. There were no significant differences in cell proliferative activity between MC+ and MC- on day 12. CONCLUSIONS This study demonstrated that the presence of MSCs could transiently enhance the proliferative activity of myoblasts, but that this enhancement is ultimately diminished due to contact inhibition.
Collapse
Affiliation(s)
- Takashi Umezawa
- Department of Anatomy, Tokyo Dental College, 2-9-18, Misaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan.
| | - Kazunari Higa
- Department of Ophthalmology/Cornea Center, Tokyo Dental College, 5-11-13 Sugano Ichikawa-shi, Chiba 272-8513, Japan
| | - Masamitsu Serikawa
- Department of Anatomy, Tokyo Dental College, 2-9-18, Misaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Masahito Yamamoto
- Department of Anatomy, Tokyo Dental College, 2-9-18, Misaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Satoru Matsunaga
- Department of Anatomy, Tokyo Dental College, 2-9-18, Misaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Jun Shimazaki
- Department of Ophthalmology/Cornea Center, Tokyo Dental College, 5-11-13 Sugano Ichikawa-shi, Chiba 272-8513, Japan
| | - Shinichi Abe
- Department of Anatomy, Tokyo Dental College, 2-9-18, Misaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| |
Collapse
|
31
|
Dumont NA, Bentzinger CF, Sincennes MC, Rudnicki MA. Satellite Cells and Skeletal Muscle Regeneration. Compr Physiol 2016; 5:1027-59. [PMID: 26140708 DOI: 10.1002/cphy.c140068] [Citation(s) in RCA: 492] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Skeletal muscles are essential for vital functions such as movement, postural support, breathing, and thermogenesis. Muscle tissue is largely composed of long, postmitotic multinucleated fibers. The life-long maintenance of muscle tissue is mediated by satellite cells, lying in close proximity to the muscle fibers. Muscle satellite cells are a heterogeneous population with a small subset of muscle stem cells, termed satellite stem cells. Under homeostatic conditions all satellite cells are poised for activation by stimuli such as physical trauma or growth signals. After activation, satellite stem cells undergo symmetric divisions to expand their number or asymmetric divisions to give rise to cohorts of committed satellite cells and thus progenitors. Myogenic progenitors proliferate, and eventually differentiate through fusion with each other or to damaged fibers to reconstitute fiber integrity and function. In the recent years, research has begun to unravel the intrinsic and extrinsic mechanisms controlling satellite cell behavior. Nonetheless, an understanding of the complex cellular and molecular interactions of satellite cells with their dynamic microenvironment remains a major challenge, especially in pathological conditions. The goal of this review is to comprehensively summarize the current knowledge on satellite cell characteristics, functions, and behavior in muscle regeneration and in pathological conditions.
Collapse
Affiliation(s)
- Nicolas A Dumont
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - C Florian Bentzinger
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Nestlé Institute of Health Sciences, EPFL Campus, Lausanne, Switzerland
| | - Marie-Claude Sincennes
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Michael A Rudnicki
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Faculty of Medicine, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
32
|
Gorski JP, Huffman NT, Vallejo J, Brotto L, Chittur SV, Breggia A, Stern A, Huang J, Mo C, Seidah NG, Bonewald L, Brotto M. Deletion of Mbtps1 (Pcsk8, S1p, Ski-1) Gene in Osteocytes Stimulates Soleus Muscle Regeneration and Increased Size and Contractile Force with Age. J Biol Chem 2015; 291:4308-22. [PMID: 26719336 DOI: 10.1074/jbc.m115.686626] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Indexed: 12/28/2022] Open
Abstract
Conditional deletion of Mbtps1 (cKO) protease in bone osteocytes leads to an age-related increase in mass (12%) and in contractile force (30%) in adult slow twitch soleus muscles (SOL) with no effect on fast twitch extensor digitorum longus muscles. Surprisingly, bone from 10-12-month-old cKO animals was indistinguishable from controls in size, density, and morphology except for a 25% increase in stiffness. cKO SOL exhibited increased expression of Pax7, Myog, Myod1, Notch, and Myh3 and 6-fold more centralized nuclei, characteristics of postnatal regenerating muscle, but only in type I myosin heavy chain-expressing cells. Increased expression of gene pathways mediating EGF receptor signaling, circadian exercise, striated muscle contraction, and lipid and carbohydrate oxidative metabolism were also observed in cKO SOL. This muscle phenotype was not observed in 3-month-old mice. Although Mbtps1 mRNA and protein expression was reduced in cKO bone osteocytes, no differences in Mbtps1 or cre recombinase expression were observed in cKO SOL, explaining this age-related phenotype. Understanding bone-muscle cross-talk may provide a fresh and novel approach to prevention and treatment of age-related muscle loss.
Collapse
Affiliation(s)
- Jeff P Gorski
- From the Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City Center of Excellence in the Study of Dental and Musculoskeletal Tissues, School of Dentistry,
| | - Nichole T Huffman
- From the Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City Center of Excellence in the Study of Dental and Musculoskeletal Tissues, School of Dentistry
| | - Julian Vallejo
- Muscle Biology Research Group, School of Nursing and Health Studies, and
| | - Leticia Brotto
- Muscle Biology Research Group, School of Nursing and Health Studies, and
| | - Sridar V Chittur
- Center for Functional Genomics, University at Albany, Rensselaer, New York 12144
| | | | - Amber Stern
- School of Computing and Engineering, University of Missouri-Kansas City, Kansas City, Missouri 64108, Engineering Systems, Inc., Charlotte, North Carolina 28277, and
| | - Jian Huang
- Muscle Biology Research Group, School of Nursing and Health Studies, and
| | - Chenglin Mo
- Muscle Biology Research Group, School of Nursing and Health Studies, and
| | - Nabil G Seidah
- Institut de Recherches Cliniques Montreal, Montreal, Quebec H2W IR7, Canada
| | - Lynda Bonewald
- From the Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City Center of Excellence in the Study of Dental and Musculoskeletal Tissues, School of Dentistry
| | - Marco Brotto
- Muscle Biology Research Group, School of Nursing and Health Studies, and
| |
Collapse
|
33
|
Abstract
The Parkes Weber syndrome is a congenital vascular malformation, characterized by varicose veins, arterio-venous fistulas and overgrown limbs. No broadly accepted animal model of Parkes Weber syndrome has been described. We created side-to-side arterio-venous fistula between common femoral vessels with proximal non-absorbable ligature on common femoral vein limiting the enlargement of the vein diameter in Wistar rats. Contralateral limb was sham operated. Invasive blood pressure measurements in both iliac and inferior cava veins were performed in rats 30 days after fistula creation. Tight circumference and femoral bone length were measured. Histopathology and morphology of soleus muscle, extensor digitorum longus muscle, and the common femoral vessel were analyzed. 30 days following arterio-venous fistula creation, a statistically significant elevation of blood pressure in common iliac vein and limb overgrowth was observed. Limb enlargement was caused by muscle overgrowth, varicose veins formation and bone elongation. Arterio-venous fistula with proximal outflow limitation led to significant increase of femoral vein circumference and venous wall thickness. Our study indicates that the described rat model mimics major clinical features characteristic for the human Parkes Weber syndrome: presence of arterio-venous fistula, venous hypertension and dilatation, varicose veins formation, and the limb hypertrophy. We reveal that limb overgrowth is caused by bone elongation, muscle hypertrophy, and venous dilatation. The newly established model will permit detailed studies on the mechanisms underlying the disease and on the efficacy of novel therapeutic strategies for the Parkes Weber syndrome treatment.
Collapse
|
34
|
Khodabukus A, Baar K. Contractile and metabolic properties of engineered skeletal muscle derived from slow and fast phenotype mouse muscle. J Cell Physiol 2015; 230:1750-7. [PMID: 25335966 DOI: 10.1002/jcp.24848] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 10/14/2014] [Indexed: 11/08/2022]
Abstract
Satellite cells derived from fast and slow muscles have been shown to adopt contractile and metabolic properties of their parent muscle. Mouse muscle shows less distinctive fiber-type profiles than rat or rabbit muscle. Therefore, in this study we sought to determine whether three-dimensional muscle constructs engineered from slow soleus (SOL) and fast tibialis anterior (TA) from mice would adopt the contractile and metabolic properties of their parent muscle. Time-to-peak tension (TPT) and half-relaxation time (1/2RT) was significantly slower in SOL constructs. In agreement with TPT, TA constructs contained significantly higher levels of fast myosin heavy chain (MHC) and fast troponin C, I, and T isoforms. Fast SERCA protein, both slow and fast calsequestrin isoforms and parvalbumin were found at higher levels in TA constructs. SOL constructs were more fatigue resistant and contained higher levels of the mitochondrial proteins SDH and ATP synthase and the fatty acid transporter CPT-1. SOL constructs contained lower levels of the glycolytic enzyme phosphofructokinase but higher levels of the β-oxidation enzymes LCAD and VLCAD suggesting greater fat oxidation. Despite no changes in PGC-1α protein, SOL constructs contained higher levels of SIRT1 and PRC. TA constructs contained higher levels of the slow-fiber program repressor SOX6 and the six transcriptional complex (STC) proteins Eya1 and Six4 which may underlie the higher in fast-fiber and lower slow-fiber program proteins. Overall, we have found that muscles engineered from predominantly slow and fast mouse muscle retain contractile and metabolic properties of their native muscle.
Collapse
Affiliation(s)
- Alastair Khodabukus
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, California
| | | |
Collapse
|
35
|
Schiaffino S, Rossi AC, Smerdu V, Leinwand LA, Reggiani C. Developmental myosins: expression patterns and functional significance. Skelet Muscle 2015; 5:22. [PMID: 26180627 PMCID: PMC4502549 DOI: 10.1186/s13395-015-0046-6] [Citation(s) in RCA: 345] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/27/2015] [Indexed: 11/24/2022] Open
Abstract
Developing skeletal muscles express unique myosin isoforms, including embryonic and neonatal myosin heavy chains, coded by the myosin heavy chain 3 (MYH3) and MYH8 genes, respectively, and myosin light chain 1 embryonic/atrial, encoded by the myosin light chain 4 (MYL4) gene. These myosin isoforms are transiently expressed during embryonic and fetal development and disappear shortly after birth when adult fast and slow myosins become prevalent. However, developmental myosins persist throughout adult stages in specialized muscles, such as the extraocular and jaw-closing muscles, and in the intrafusal fibers of the muscle spindles. These myosins are re-expressed during muscle regeneration and provide a specific marker of regenerating fibers in the pathologic skeletal muscle. Mutations in MYH3 or MYH8 are responsible for distal arthrogryposis syndromes, characterized by congenital joint contractures and orofacial dysmorphisms, supporting the importance of muscle contractile activity and body movements in joint development and in shaping the form of the face during fetal development. The biochemical and biophysical properties of developmental myosins have only partially been defined, and their functional significance is not yet clear. One possibility is that these myosins are specialized in contracting against low loads, and thus, they may be adapted to the prenatal environment, when fetal muscles contract against a very low load compared to postnatal muscles.
Collapse
Affiliation(s)
- Stefano Schiaffino
- Venetian Institute of Molecular Medicine (VIMM), Via G. Orus 2, 35129 Padova, Italy
| | - Alberto C Rossi
- Department of Molecular, Cellular and Developmental Biology and BioFrontiers Institute, University of Colorado, Boulder, CO USA
| | - Vika Smerdu
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Leslie A Leinwand
- Department of Molecular, Cellular and Developmental Biology and BioFrontiers Institute, University of Colorado, Boulder, CO USA
| | - Carlo Reggiani
- Department of Biomedical Sciences, University of Padova, Padova, Italy ; CNR Institute of Neuroscience, Padova, Italy
| |
Collapse
|
36
|
Oishi Y, Roy RR, Ogata T, Ohira Y. Heat-Stress effects on the myosin heavy chain phenotype of rat soleus fibers during the early stages of regeneration. Muscle Nerve 2015; 52:1047-56. [DOI: 10.1002/mus.24686] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 04/07/2015] [Accepted: 04/13/2015] [Indexed: 11/10/2022]
Affiliation(s)
- Yasuharu Oishi
- Laboratory of Muscle Physiology; Faculty of Education, Kumamoto University; Kumamoto 860-8555 Japan
| | - Roland R. Roy
- Department of Integrative Biology and Physiology and Brain Research Institute; University of California Los Angeles; Los Angeles California USA
| | - Tomonori Ogata
- Faculty of Human Environmental Studies; Hiroshima-Shudo University; Hiroshima Japan
| | - Yoshinobu Ohira
- Research Center for Adipocyte & Muscle Science; Doshisha University; Kyotanabe City Kyoto Japan
| |
Collapse
|
37
|
Fillmore N, Keung W, Kelly SE, Proctor SD, Lopaschuk GD, Ussher JR. Accumulation of ceramide in slow-twitch muscle contributes to the development of insulin resistance in the obese JCR:LA-cp rat. Exp Physiol 2015; 100:730-41. [DOI: 10.1113/ep085052] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 03/17/2015] [Indexed: 12/30/2022]
Affiliation(s)
- Natasha Fillmore
- Cardiovascular Translational Science Institute; University of Alberta; Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute; University of Alberta; Edmonton Alberta Canada
| | - Wendy Keung
- Mazankowski Alberta Heart Institute; University of Alberta; Edmonton Alberta Canada
| | - Sandra E. Kelly
- Mazankowski Alberta Heart Institute; University of Alberta; Edmonton Alberta Canada
- Alberta Diabetes Institute; University of Alberta; Edmonton Alberta Canada
| | - Spencer D. Proctor
- Mazankowski Alberta Heart Institute; University of Alberta; Edmonton Alberta Canada
- Alberta Diabetes Institute; University of Alberta; Edmonton Alberta Canada
| | - Gary D. Lopaschuk
- Cardiovascular Translational Science Institute; University of Alberta; Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute; University of Alberta; Edmonton Alberta Canada
- Alberta Diabetes Institute; University of Alberta; Edmonton Alberta Canada
| | - John R. Ussher
- Cardiovascular Translational Science Institute; University of Alberta; Edmonton Alberta Canada
- Alberta Diabetes Institute; University of Alberta; Edmonton Alberta Canada
- Faculty of Pharmacy and Pharmaceutical Sciences; University of Alberta; Edmonton Alberta Canada
| |
Collapse
|
38
|
Khodabukus A, Baar K. Glucose Concentration and Streptomycin Alter In Vitro Muscle Function and Metabolism. J Cell Physiol 2015; 230:1226-34. [DOI: 10.1002/jcp.24857] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 10/24/2014] [Indexed: 11/06/2022]
Affiliation(s)
- Alastair Khodabukus
- Division of Neurobiology; Physiology and Behavior; University of California Davis; Davis California
| | - Keith Baar
- Division of Neurobiology; Physiology and Behavior; University of California Davis; Davis California
| |
Collapse
|
39
|
Characterization of excitation–contraction coupling components in human extraocular muscles. Biochem J 2015; 466:29-36. [DOI: 10.1042/bj20140970] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
We show that the expression level of RyR1 in human extraocular muscles (EOMs) is low and that these muscles express different levels of proteins involved in excitation–contraction coupling (ECC) compared with leg muscles (LMs).
Collapse
|
40
|
Genetic Dissection of the Physiological Role of Skeletal Muscle in Metabolic Syndrome. ACTA ACUST UNITED AC 2014. [DOI: 10.1155/2014/635146] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The primary deficiency underlying metabolic syndrome is insulin resistance, in which insulin-responsive peripheral tissues fail to maintain glucose homeostasis. Because skeletal muscle is the major site for insulin-induced glucose uptake, impairments in skeletal muscle’s insulin responsiveness play a major role in the development of insulin resistance and type 2 diabetes. For example, skeletal muscle of type 2 diabetes patients and their offspring exhibit reduced ratios of slow oxidative muscle. These observations suggest the possibility of applying muscle remodeling to recover insulin sensitivity in metabolic syndrome. Skeletal muscle is highly adaptive to external stimulations such as exercise; however, in practice it is often not practical or possible to enforce the necessary intensity to obtain measurable benefits to the metabolic syndrome patient population. Therefore, identifying molecular targets for inducing muscle remodeling would provide new approaches to treat metabolic syndrome. In this review, the physiological properties of skeletal muscle, genetic analysis of metabolic syndrome in human populations and model organisms, and genetically engineered mouse models will be discussed in regard to the prospect of applying skeletal muscle remodeling as possible therapy for metabolic syndrome.
Collapse
|
41
|
Rossi G, Messina G. Comparative myogenesis in teleosts and mammals. Cell Mol Life Sci 2014; 71:3081-99. [PMID: 24664432 PMCID: PMC4111864 DOI: 10.1007/s00018-014-1604-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 02/17/2014] [Accepted: 03/06/2014] [Indexed: 01/02/2023]
Abstract
Skeletal myogenesis has been and is currently under extensive study in both mammals and teleosts, with the latter providing a good model for skeletal myogenesis because of their flexible and conserved genome. Parallel investigations of muscle studies using both these models have strongly accelerated the advances in the field. However, when transferring the knowledge from one model to the other, it is important to take into account both their similarities and differences. The main difficulties in comparing mammals and teleosts arise from their different temporal development. Conserved aspects can be seen for muscle developmental origin and segmentation, and for the presence of multiple myogenic waves. Among the divergences, many fish have an indeterminate growth capacity throughout their entire life span, which is absent in mammals, thus implying different post-natal growth mechanisms. This review covers the current state of the art on myogenesis, with a focus on the most conserved and divergent aspects between mammals and teleosts.
Collapse
Affiliation(s)
- Giuliana Rossi
- Department of Biosciences, University of Milan, 20133, Milan, Italy
| | | |
Collapse
|
42
|
Hetzler KL, Collins BC, Shanely RA, Sue H, Kostek MC. The homoeobox gene SIX1 alters myosin heavy chain isoform expression in mouse skeletal muscle. Acta Physiol (Oxf) 2014; 210:415-28. [PMID: 24102895 DOI: 10.1111/apha.12168] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 08/21/2013] [Accepted: 09/10/2013] [Indexed: 01/06/2023]
Abstract
AIM Six1 is necessary for the genesis of several tissues, but in adults, it is expressed primarily in skeletal muscle where its function is unclear. Overexpression of Six1 with a cofactor in skeletal muscle causes slow-to-fast fibre-type transition. We sought to characterize the effects of a physiologically relevant Six1 knockdown. METHODS The tibialis anterior (TA) muscles of C57BL/6 mice were electroporated with Six1 knockdown vector (siRNA) or empty vector. Muscles were collected at 2 or 14 days after transfection for Six1 and myosin heavy chain (MHC) expression analysis. C2C12 mouse myoblasts were grown in standard conditions. Cells were cotransfected with MHC promoter vectors and Six1 expression vectors. Cells were harvested after 4 days of differentiation. RESULTS In vivo, the Six1 siRNA caused a decreased expression of Six1,1.8-fold (±0.1, P < 0.05). With decreased Six1, MHC IIB expression decreased 2.7-fold (±0.7, P = 0.04). Proportion of muscle fibres expressing MHC IIB decreased (45.3 ± 4.8% vs. 65.1 ± 7.3% in control group, P = 0.04), and total area expressing MHC IIB decreased with decreased Six1 (59.6 ± 4.3% vs. 75.2 ± 5.4% in control group, P < 0.05). Decreased Six1 increased MHC IIA expression 1.9-fold (±0.3, P = 0.04). In vitro, Six1 overexpression increased promoter activation of MHC IIB 2.9-fold (±0.3, P < 0.01). Six1 knockdown repressed MHC IIB promoter 2.9-fold (±0.1, P < 0.05) and MHC IIX 3.7-fold (±0.08, P < 0.01). CONCLUSION Six1 knockdown caused a fast-to-slow shift in MHC isoform, and this was confirmed by promoter activity of MHC genes. Six1 may ultimately control the contractile and metabolic properties that define muscle fibre phenotype.
Collapse
Affiliation(s)
- K. L. Hetzler
- Department of Exercise Science; University of South Carolina; Columbia SC USA
| | - B. C. Collins
- Department of Exercise Science; University of South Carolina; Columbia SC USA
| | - R. A. Shanely
- Appalachian State University-North Carolina Research Campus Human Performance Laboratory; Appalachian State University; Kannapolis NC USA
| | - H. Sue
- Department of Exercise Science; University of South Carolina; Columbia SC USA
| | - M. C. Kostek
- Department of Exercise Science; University of South Carolina; Columbia SC USA
- Department of Physical Therapy; Duquesne University; Pittsburgh PA USA
| |
Collapse
|
43
|
Robison P, Hernández-Ochoa EO, Schneider MF. Atypical behavior of NFATc1 in cultured intercostal myofibers. Skelet Muscle 2014; 4:1. [PMID: 24383888 PMCID: PMC3895734 DOI: 10.1186/2044-5040-4-1] [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: 07/01/2013] [Accepted: 11/28/2013] [Indexed: 12/26/2022] Open
Abstract
Background The NFATc transcription factor family is responsible for coupling cytoplasmic calcium signals to transcription programs in a wide variety of cell types. In skeletal muscle, these transcription factors control the fiber type in response to muscle activity. This excitation-transcription (E-T) coupling permits functional adaptation of muscle according to use. The activity dependence of these transcription programs is sensitive to the firing patterns of the muscle, not merely the period of activity, enabling a nuanced adaptation to various functional tasks. Methods Isolated skeletal muscle fibers expressing exogenous fluorescent NFATc1 were studied by confocal microscopy under stimulation both with and without pharmacological inhibitors. Western blots of whole muscle lysates were also used. Results This study investigates the activity dependent response of NFATc1 skeletal muscle fibers cultured from mice, comparing fibers of respiratory origin to muscles responsible for limb locomotion. Using patterns of stimulation known to strongly activate NFATc1 in the commonly cultured flexor digitorum brevis and soleus muscles, we have observed significant deactivation of NFATc1 in cultured intercostal muscle fibers. This effect is at least partially dependent on the action of JNK and CaMKII in intercostal fibers. Conclusions Our findings highlight the role of lineage in the NFAT pathway, showing that the respiratory intercostal muscle fibers decode similar E-T coupling signals into NFAT transcriptional programs in a different manner from the more commonly studied locomotor muscles of the limbs.
Collapse
Affiliation(s)
| | | | - Martin F Schneider
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N Greene Street, Baltimore, MD 21201, USA.
| |
Collapse
|
44
|
Smith BK, Mathur S, Ye F, Martin AD, Truelson SA, Vandenborne K, Davenport PW. Intrinsic transient tracheal occlusion training and myogenic remodeling of rodent parasternal intercostal fibers. JOURNAL OF REHABILITATION RESEARCH AND DEVELOPMENT 2014; 51:841-854. [PMID: 25509059 PMCID: PMC4269303 DOI: 10.1682/jrrd.2012.12.0232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
It is recognized that diaphragm muscle plasticity occurs with mechanical overloads, yet less is known about synergistic parasternal intercostal muscle fiber remodeling. We conducted overload training with intrinsic transient tracheal occlusion (ITTO) exercises in conscious animals. We hypothesized that ITTO would yield significant fiber hypertrophy and myogenic activation that would parallel diaphragm fiber remodeling. Sprague-Dawley rats underwent placement of a tracheal cuff and were randomly assigned to receive daily 10 min sessions of conscious ITTO or observation (sham) over 2 wk. After training, fiber morphology, myosin heavy chain (MHC) isoform composition, cross-sectional area, proportion of Pax7-positive nuclei, and presence of embryonic MHC (eMHC) were quantified. Type IIx/b fibers were 20% larger after ITTO training than with sham training (ITTO: 4,431 +/– 676 μm2, sham: 3,689 +/– 400 μm2, p < 0.05), and type I fibers were more prevalent after ITTO (p < 0.01). Expression of Pax7 was increased in ITTO parasternals and diaphragm (p < 0.05). In contrast, the proportion of eMHC-positive fibers was increased only in ITTO parasternals (1.2% [3.4%–0.6%], sham: 0% [0.6%–0%], p < 0.05). Although diaphragm and parasternal type II fibers hypertrophy to a similar degree, myogenic remodeling appears to differ between the two muscles.
Collapse
Affiliation(s)
- Barbara K. Smith
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, USA
| | - Sunita Mathur
- Department of Physiotherapy, University of Toronto, Toronto, Ontario, Canada
| | - Fan Ye
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - A. Daniel Martin
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, USA
| | | | - Krista Vandenborne
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, USA
| | - Paul W. Davenport
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
45
|
Klasan GS, Ivanac D, Erzen DJ, Picard A, Takasawa S, Peharec S, Arbanas J, Girotto D, Jerkovic R. Reg3G
gene expression in regenerating skeletal muscle and corresponding nerve. Muscle Nerve 2013; 49:61-8. [DOI: 10.1002/mus.23877] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2013] [Indexed: 12/19/2022]
Affiliation(s)
- Gordana Starcevic Klasan
- Department of Anatomy; School of Medicine; University of Rijeka; Brace Branchetta 20 51000 Rijeka Croatia
| | - Danijel Ivanac
- Department of Anatomy; School of Medicine; University of Rijeka; Brace Branchetta 20 51000 Rijeka Croatia
| | | | - Anne Picard
- Department of Biomedical Science; University of Padua; Padua Italy
| | - Shin Takasawa
- Department of Biochemistry; Nara Medical University; Nara Japan
| | | | - Juraj Arbanas
- Department of Anatomy; School of Medicine; University of Rijeka; Brace Branchetta 20 51000 Rijeka Croatia
| | - Dean Girotto
- Department of Neurosurgery; University Hospital Rijeka; Rijeka Croatia
| | - Romana Jerkovic
- Department of Anatomy; School of Medicine; University of Rijeka; Brace Branchetta 20 51000 Rijeka Croatia
| |
Collapse
|
46
|
Pallafacchina G, Blaauw B, Schiaffino S. Role of satellite cells in muscle growth and maintenance of muscle mass. Nutr Metab Cardiovasc Dis 2013; 23 Suppl 1:S12-S18. [PMID: 22621743 DOI: 10.1016/j.numecd.2012.02.002] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2011] [Revised: 02/01/2012] [Accepted: 02/06/2012] [Indexed: 01/25/2023]
Abstract
Changes in muscle mass may result from changes in protein turnover, reflecting the balance between protein synthesis and protein degradation, and changes in cell turnover, reflecting the balance between myonuclear accretion and myonuclear loss. Myonuclear accretion, i.e. increase in the number of myonuclei within the muscle fibers, takes place via proliferation and fusion of satellite cells, myogenic stem cells associated to skeletal muscle fibers and involved in muscle regeneration. In developing muscle, satellite cells undergo extensive proliferation and most of them fuse with myofibers, thus contributing to the increase in myonuclei during early postnatal stages. A similar process is induced in adult skeletal muscle by functional overload and exercise. In contrast, satellite cells and myonuclei may undergo apoptosis during muscle atrophy, although it is debated whether myonuclear loss occurs in atrophying muscle. An increase in myofiber size can also occur by changes in protein turnover without satellite cell activation, e.g. in late phases of postnatal development or in some models of muscle hypertrophy. The relative role of protein turnover and cell turnover in muscle adaptation and in the establishment of functional muscle hypertrophy remains to be established. The identification of the signaling pathways mediating satellite cell activation may provide therapeutic targets for combating muscle wasting in a variety of pathological conditions, including cancer cachexia, renal and cardiac failure, neuromuscular diseases, as well as aging sarcopenia.
Collapse
Affiliation(s)
- G Pallafacchina
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy; Consiglio Nazionale delle Ricerche (CNR) Institute of Neurosciences, Padova, Italy; Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - B Blaauw
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy; Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - S Schiaffino
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy; Consiglio Nazionale delle Ricerche (CNR) Institute of Neurosciences, Padova, Italy.
| |
Collapse
|
47
|
Establishment of a human skeletal muscle-derived cell line: biochemical, cellular and electrophysiological characterization. Biochem J 2013; 455:169-77. [PMID: 23905709 DOI: 10.1042/bj20130698] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Excitation-contraction coupling is the physiological mechanism occurring in muscle cells whereby an electrical signal sensed by the dihydropyridine receptor located on the transverse tubules is transformed into a chemical gradient (Ca2+ increase) by activation of the ryanodine receptor located on the sarcoplasmic reticulum membrane. In the present study, we characterized for the first time the excitation-contraction coupling machinery of an immortalized human skeletal muscle cell line. Intracellular Ca2+ measurements showed a normal response to pharmacological activation of the ryanodine receptor, whereas 3D-SIM (super-resolution structured illumination microscopy) revealed a low level of structural organization of ryanodine receptors and dihydropyridine receptors. Interestingly, the expression levels of several transcripts of proteins involved in Ca2+ homoeostasis and differentiation indicate that the cell line has a phenotype closer to that of slow-twitch than fast-twitch muscles. These results point to the potential application of such human muscle-derived cell lines to the study of neuromuscular disorders; in addition, they may serve as a platform for the development of therapeutic strategies aimed at correcting defects in Ca2+ homoeostasis due to mutations in genes involved in Ca2+ regulation.
Collapse
|
48
|
Ciciliot S, Rossi AC, Dyar KA, Blaauw B, Schiaffino S. Muscle type and fiber type specificity in muscle wasting. Int J Biochem Cell Biol 2013; 45:2191-9. [DOI: 10.1016/j.biocel.2013.05.016] [Citation(s) in RCA: 433] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 05/13/2013] [Accepted: 05/14/2013] [Indexed: 01/05/2023]
|
49
|
Zhu H, Park S, Scheffler JM, Kuang S, Grant AL, Gerrard DE. Porcine satellite cells are restricted to a phenotype resembling their muscle origin. J Anim Sci 2013; 91:4684-91. [PMID: 23893979 DOI: 10.2527/jas.2012-5804] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Muscles in most domestic animals differ in function and growth potential based largely on muscle fiber type composition. Though much is known about satellite cells (SC), information is limited regarding how populations of SC differ with muscle fiber type, especially in pigs. Therefore, the objective of this study was to isolate and culture SC from red (RST) and white (WST) portions of the semitendinosus muscle of neonatal and adult pigs and determine their capacity to proliferate, differentiate, and express various myosin heavy chain (MyHC) isoforms in vitro. Porcine satellite cells were isolated from RST and WST muscles of 6-wk-old and adult (>6-mo-old) pigs and cultured under standard conditions. Muscle from neonatal pigs yielded nearly 10 times more (P < 0.001) presumptive satellite cells as those from adult pigs, with fusion percentages close to 60% for the former. The RST yielded more (P < 0.001) SC per gram muscle compared to WST, 8.1 ± 0.2 × 10(4) cells versus 6.7 ± 0.1 × 10(4) cells/gram muscle in young pigs, and 9.7 ± 0.4 × 10(3) cells versus 5.5 ± 0.4 × 10(3) cells/gram muscle in adult pigs, respectively. Likewise, satellite cells from RST proliferated faster (P < 0.001) than those from WST across both ages, as indicated by a shorter cell doubling time, 18.6 ± 0.8 h versus 21.3 ± 0.9 h in young pigs, and 23.2 ± 0.7 h versus 26.7 ± 0.9 h in adult pigs, respectively. As a result of shorter times to confluence, satellite cells from RST also formed myotubes earlier than those SC originating from WST. Once induced, however, SC from WST differentiated and fused faster (P < 0.05) as evidenced by fusion percentage within the first 24 h, 41.6% versus 34.3%, respectively; but reached similar ultimate fusion percentages similar to WST by 48 h. Over 90% of MyHC expressed in maximally fused SC cultures from both RST and WST was restricted to the embryonic isoform. Type IIX MyHC mRNA was not detected in any culture. Myotube cultures from RST expressed more (P < 0.01) Type I MyHC isoform mRNA than those from WST, whereas those cultures from WST expressed more (P < 0.05) Type II (including Types IIA and IIB) MyHC transcripts. These data show SC cultures from porcine fast and slow muscles express MyHC profiles largely reflective of their muscle of origin and suggest satellite cells are partially restricted to a particular muscle phenotype in which they are juxtapositioned. Understanding the molecular nature of these intrinsic control mechanisms may lead to improved strategies for augmenting meat animal growth or muscle regeneration.
Collapse
Affiliation(s)
- H Zhu
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg 24060
| | | | | | | | | | | |
Collapse
|
50
|
Chapman MR, Balakrishnan KR, Li J, Conboy MJ, Huang H, Mohanty SK, Jabart E, Hack J, Conboy IM, Sohn LL. Sorting single satellite cells from individual myofibers reveals heterogeneity in cell-surface markers and myogenic capacity. Integr Biol (Camb) 2013; 5:692-702. [PMID: 23407661 DOI: 10.1039/c3ib20290a] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Traditional cell-screening techniques such as FACS and MACS are better suited for large numbers of cells isolated from bulk tissue and cannot easily screen stem or progenitor cells from minute populations found in their physiological niches. Furthermore, these techniques rely upon irreversible antibody binding, potentially altering cell properties, including gene expression and regenerative capacity. To address these challenges, we have developed a novel, label-free stem-cell analysis and sorting platform capable of quantifying cell-surface marker expression of single functional organ stem cells directly isolated from their micro-anatomical niche. Using our unique platform, we have discovered a remarkable heterogeneity in both the regenerative capacity and expression of CXCR4, β1-integrin, Sca-1, M-cadherin, Syndecan-4, and Notch-1 in freshly isolated muscle stem (satellite) cells residing on different, single myofibers and have identified a small population of Sca-1(+)/Myf5(+) myogenic satellite cells. Our results demonstrate the utility of our single-cell platform for uncovering and functionally characterizing stem-cell heterogeneity in the organ microniche.
Collapse
Affiliation(s)
- Matthew R Chapman
- Biophysics Graduate Group, University of California, Berkeley, CA 94720, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|