1
|
Calabrese EJ, Agathokleous E, Dhawan G, Kapoor R, Dhawan V, Manes PK, Calabrese V. Nitric oxide and hormesis. Nitric Oxide 2023; 133:1-17. [PMID: 36764605 DOI: 10.1016/j.niox.2023.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/20/2023] [Accepted: 02/05/2023] [Indexed: 02/10/2023]
Abstract
This present paper provides an assessment of the occurrence of nitric oxide (NO)-induced hormetic-biphasic dose/concentration relationships in biomedical research. A substantial reporting of such NO-induced hormetic effects was identified with particular focus on wound healing, tumor promotion, and sperm biology, including mechanistic assessment and potential for translational applications. Numerous other NO-induced hormetic effects have been reported, but require more development prior to translational applications. The extensive documentation of NO-induced biphasic responses, across numerous organs (e.g., bone, cardiovascular, immune, intestine, and neuronal) and cell types, suggests that NO-induced biological activities are substantially mediated via hormetic processes. These observations are particularly important because broad areas of NO biology are constrained by the quantitative features of the hormetic response. This determines the amplitude and width of the low dose stimulation, affecting numerous biomedical implications, study design features (e.g., number of doses, dose spacing, sample sizes, statistical power), and the potential success of clinical trials.
Collapse
Affiliation(s)
- Edward J Calabrese
- Department of Environmental Health Sciences, Morrill I, N344, University of Massachusetts, Amherst, MA, 01003, USA.
| | - Evgenios Agathokleous
- School of Applied Meteorology, Nanjing University of Information Science & Technology, Nanjing, 210044, China.
| | | | - Rachna Kapoor
- Saint Francis Hospital and Medical Center, Hartford, CT, USA.
| | - Vikas Dhawan
- Department of Surgery, Indian Naval Ship Hospital, Mumbai, India.
| | | | - Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, School of Medicine University of Catania, Via Santa Sofia 97, Catania, 95123, Italy.
| |
Collapse
|
2
|
Calabrese EJ, Calabrese V. Enhancing health span: muscle stem cells and hormesis. Biogerontology 2022; 23:151-167. [PMID: 35254570 DOI: 10.1007/s10522-022-09949-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/04/2022] [Indexed: 12/17/2022]
Abstract
Sarcopenia is a significant public health and medical concern confronting the elderly. Considerable research is being directed to identify ways in which the onset and severity of sarcopenia may be delayed/minimized. This paper provides a detailed identification and assessment of hormetic dose responses in animal model muscle stem cells, with particular emphasis on cell proliferation, differentiation, and enhancing resilience to inflammatory stresses and how this information may be useful in preventing sarcopenia. Hormetic dose responses were observed following administration of a broad range of agents, including dietary supplements (e.g., resveratrol), pharmaceuticals (e.g., dexamethasone), endogenous ligands (e.g., tumor necrosis factor α), environmental contaminants (e.g., cadmium) and physical agents (e.g., low level laser). The paper assesses both putative mechanisms of hormetic responses in muscle stem cells, and potential therapeutic implications and application(s) of hormetic frameworks for slowing muscle loss and reduced functionality during the aging process.
Collapse
Affiliation(s)
- Edward J Calabrese
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Morrill I, N344, Amherst, MA, 01003, USA.
| | - Vittorio Calabrese
- Department of Biomedical & Biotechnological Sciences, School of Medicine, University of Catania, Via Santa Sofia, 97, 95125, Catania, Italy
| |
Collapse
|
3
|
Silva NC, Alvarez AM, DeOcesano-Pereira C, Fortes-Dias CL, Moreira V. Catalytically active phospholipase A 2 myotoxin from Crotalus durissus terrificus induces proliferation and differentiation of myoblasts dependent on prostaglandins produced by both COX-1 and COX-2 pathways. Int J Biol Macromol 2021; 187:603-613. [PMID: 34314795 DOI: 10.1016/j.ijbiomac.2021.07.121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 01/18/2023]
Abstract
Although crotoxin B (CB) is a well-established catalytically active secretory phospholipase A2 group IIA (sPLA2-IIA) myotoxin, we investigated its potential stimulatory effect on myogenesis with the involvement of prostaglandins (PGs) produced by cyclooxygenase (COX)-1 and -2 pathways. Myoblast C2C12 were cultured in proliferation or commitment protocols and incubated with CB followed by lumiracoxib (selective COX-2 inhibitor) or valeryl salicylate (selective COX-1 inhibitor) and subjected to analysis of PG release, cell proliferation and activation of myogenic regulatory factors (MRFs). Our data showed that CB in non-cytotoxic concentrations induces an increase of COX-2 protein expression and stimulates the activity of both COX isoforms to produce PGE2, PGD2 and 15d-PGJ2. CB induced an increase in the proliferation of C2C12 myoblast cells dependent on PGs from both COX-1 and COX-2 pathways. In addition, CB stimulated the activity of Pax7, MyoD, Myf5 and myogenin in proliferated cells. Otherwise, CB increased myogenin activity but not MyoD in committed cells. Our findings evidence the role of COX-1- and COX-2-derived PGs in modulating CB-induced activation of MRFs. This study contributes to the knowledge that CB promote early myogenic events via regulatory mechanisms on PG-dependent COX pathways, showing new concepts about the effect of sPLA2-IIA in skeletal muscle repair.
Collapse
Affiliation(s)
- Nadine C Silva
- Pharmacology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP 04044-020, Brazil
| | - Angela M Alvarez
- Pharmacology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP 04044-020, Brazil; Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo, SP 05503-900, Brazil.
| | - Carlos DeOcesano-Pereira
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo, SP 05503-900, Brazil.
| | | | - Vanessa Moreira
- Pharmacology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP 04044-020, Brazil.
| |
Collapse
|
4
|
Mechanisms regulating myoblast fusion: A multilevel interplay. Semin Cell Dev Biol 2020; 104:81-92. [PMID: 32063453 DOI: 10.1016/j.semcdb.2020.02.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/07/2020] [Accepted: 02/08/2020] [Indexed: 02/07/2023]
Abstract
Myoblast fusion into myotubes is one of the crucial steps of skeletal muscle development (myogenesis). The fusion is preceded by specification of a myogenic lineage (mesodermal progenitors) differentiating into myoblasts and is followed by myofiber-type specification and neuromuscular junction formation. Similarly to other processes of myogenesis, the fusion requires a very precise spatial and temporal regulation occuring both during embryonic development as well as regeneration and repair of the muscle. A plethora of genes and their products is involved in regulation of myoblast fusion and a precise multilevel interplay between them is crucial for myogenic cells to fuse. In this review, we describe both cellular events taking place during myoblast fusion (migration, adhesion, elongation, cell-cell recognition, alignment, and fusion of myoblast membranes enabling formation of myotubes) as well as recent findings on mechanisms regulating this process. Also, we present muscle disorders in humans that have been associated with defects in genes involved in regulation of myoblast fusion.
Collapse
|
5
|
Leng X, Jiang H. Effects of arachidonic acid and its major prostaglandin derivatives on bovine myoblast proliferation, differentiation, and fusion. Domest Anim Endocrinol 2019; 67:28-36. [PMID: 30677541 DOI: 10.1016/j.domaniend.2018.12.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 12/03/2018] [Accepted: 12/13/2018] [Indexed: 12/23/2022]
Abstract
Many studies have shown positive effects of prostaglandins (PGs) on various steps of skeletal muscle formation such as myoblast proliferation and myotube hypertrophy. In animals, PGs are synthesized through the action of the rate-limiting enzymes cyclooxygenase (COX) -1 and COX-2 from arachidonic acid (AA), a conditionally essential fatty acid. As a step toward exploring the possibility of using dietary supplementation of AA to improve skeletal muscle growth in cattle, which are major meat-producing animals, we determined the effects of AA and its major PG derivatives PGE2, PGF2α, and PGI2 on proliferation, differentiation, and fusion of primary bovine myoblasts in vitro. In the proliferation experiment, myoblasts were cultured in a growth medium to which was added 10 μM AA, 1 μM PGE2, 1 μM PGF2α, 1 μM PGI2, or vehicle control for 24 h, and the proliferating cells were identified by 5-ethynyl-2'-deoxyuridine (EdU) labeling. This experiment revealed that AA, PGE2, PGF2α, and PGI2 each increased the number of proliferating cells by 13%, 24%, 16%, and 16%, respectively, compared to the control (n = 7, P < 0.05). In the differentiation and fusion test, myoblasts were induced to differentiate and fuse into myotubes in the presence of the aforementioned treatments for 0, 24, 48, and 72 h. Based on quantitative reverse transcription PCR analyses of mRNAs of myoblast differentiation and fusion markers (myogenin; myosin heavy chain 3; creatine kinase, muscle; myomaker) at 0, 24, and 48 h of differentiation, AA, PGE2, and PGF2α promoted myoblast differentiation (n = 6, P < 0.05). Based on Giemsa staining and counting the number of myotubes at 72 h of differentiation, PGE2 enhanced the number of formed myotubes by 14% (P < 0.05) compared to the control. Treating the myoblasts with AA and either the COX-1 and COX-2 common inhibitor indomethacin or the COX-2-specific inhibitor NS-398 reversed the stimulatory effect of AA on myoblast proliferation (n = 4, P < 0.05). Overall, this study demonstrates that exogenous AA stimulates bovine myoblast proliferation and differentiation in culture. The results of this study suggest that AA stimulates myoblast proliferation through its metabolites PGE2, PGF2α, or PGI2, and that AA stimulates myoblast differentiation through PGE2.
Collapse
Affiliation(s)
- X Leng
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - H Jiang
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
6
|
Murray J, Whitson RH, Itakura K. Reduced prostaglandin I 2 signaling in Arid5b -/- primary skeletal muscle cells attenuates myogenesis. FASEB J 2018; 32:1868-1879. [PMID: 29196500 PMCID: PMC5893174 DOI: 10.1096/fj.201700453rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The AT-rich interaction domain (ARID) family of proteins regulates gene expression, development, and differentiation. Although Arid5b has important functions in adipogenesis and chondrogenesis, the role of Arid5b in skeletal muscle myogenesis has not been investigated. Therefore, we isolated primary skeletal muscle cells from Arid5b+/+ and Arid5b−/− mice and characterized differentiation in these cells. We found that Arid5b−/− primary skeletal muscle cells showed differentiation defects and impaired sarcomeric assembly. Microarray analysis revealed down-regulation of the prostanoid biosynthesis pathway in Arid5b−/− myoblasts, including the genes encoding cyclooxygenase (COX)-1 (Ptgs1) and prostaglandin (PG)I synthase (Ptgis). Down-regulation of COX-1 and PGI synthase was confirmed by real-time PCR and Western blot analyses. Correspondingly, the production of PGI2, as measured by ELISA, was reduced in Arid5b−/− cells relative to Arid5b+/+ cells. Boyden chamber assays showed that migration was increased but chemotaxis was impaired in Arid5b−/− cells. Myoblast fusion was also inhibited in Arid5b−/− cells compared with Arid5b+/+ cells. Treatment with the PGI2 analog iloprost rescued the defects in myotube formation, migration, and fusion. These results demonstrate that Arid5b has a novel and essential role in skeletal muscle differentiation by regulating PGI2 production.—Murray, J., Whitson, R. H., Itakura, K. Reduced prostaglandin I2 signaling in Arid5b−/− primary skeletal muscle cells attenuates myogenesis.
Collapse
Affiliation(s)
- Jennifer Murray
- Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, California, USA
| | - Robert H Whitson
- Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, California, USA
| | - Keiichi Itakura
- Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, California, USA
| |
Collapse
|
7
|
Brewer CB, Bentley JP, Day LB, Waddell DE. Resistance exercise and naproxen sodium: effects on a stable PGF2α metabolite and morphological adaptations of the upper body appendicular skeleton. Inflammopharmacology 2015; 23:319-27. [PMID: 26289996 DOI: 10.1007/s10787-015-0248-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 08/01/2015] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Exercise-induced inflammation has been shown to be necessary for successful skeletal muscle regeneration post-injury. Accordingly, numerous investigations have demonstrated consequences of COX-inhibitors, anti-inflammatory drugs which prevent prostaglandin formation. In addition to its roles in inflammation, prostaglandin F2α (PGF2α) also mediates vital regenerative processes The majority of research to report consequences of suppressing inflammation has utilized acute injury models in combination with acute COX-inhibitor administration. To address the limited research investigating regular consumption of COX-inhibitors over time in exercising humans, the purpose of this study was to determine effects of a non-selective COX-inhibitor on a PGF2α metabolite and morphological adaptations of the upper body appendicular skeleton during periodized resistance training. Twenty-three (N = 23) recreationally trained college-aged males were randomly assigned to receive placebo (n = 11) or naproxen sodium (n = 12). Treatments were prophylactically administered in double-blind fashion with supervised upper body resistance exercise performed twice per week for 6 weeks. Venous blood was sampled pre- and post-exercise and analyzed for 13, 14-dihydro-15-keto PGF2α using enzyme immunoassay. Factorial mixed-design repeated-measures ANOVAs were utilized to examine relative changes in the plasma PGF2α metabolite and upper body appendicular morphology over the training period. RESULTS Naproxen sodium significantly reduced the acute PGF2α metabolite response to exercise (p = 0.013); however, this effect diminished over time (p = 0.02), and both treatment groups exhibited significant increases in dominant arm skeletal muscle tissue (p = 0.037). CONCLUSION Despite acute inhibition of the PGF2α metabolite at early time points, naproxen sodium did not hinder positive morphological adaptations of the upper body in response to resistance training.
Collapse
Affiliation(s)
- Christi B Brewer
- Physical Education, Health, and Recreation, Eastern Washington University, 200 Physical Education Building, Cheney, WA, 99004, USA.
| | - John P Bentley
- Department of Pharmacy Administration, School of Pharmacy, University of Mississippi, Oxford, MS, USA.
| | - Lainy B Day
- Department of Biology, University of Mississippi, Oxford, MS, USA.
| | - Dwight E Waddell
- Department of Electrical Engineering, University of Mississippi, 308 Anderson Hall, Oxford, MS, USA.
| |
Collapse
|
8
|
Totzeck M, Schicho A, Stock P, Kelm M, Rassaf T, Hendgen-Cotta UB. Nitrite circumvents canonical cGMP signaling to enhance proliferation of myocyte precursor cells. Mol Cell Biochem 2014; 401:175-83. [PMID: 25501648 DOI: 10.1007/s11010-014-2305-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 12/10/2014] [Indexed: 01/12/2023]
Abstract
Skeletal muscle tissue has a remarkable high regenerative capacity. The underlying cellular events are governed by complex signaling processes, and the proliferation of skeletal myoblasts is a key initial event. The role of nitric oxide (NO) in cell cycle regulation is well-appreciated. Nitrite, an NO oxidation product, is a stable source for NO-like bioactivity particularly in cases when oxygen shortage compromises NO-synthases activity. Although numerous studies suggest that nitrite effects are largely related to NO-dependent signaling, emerging evidence also implicates that nitrite itself can activate protein pathways albeit under physiological, normoxic conditions. This includes a recently demonstrated cyclic guanosine monophosphate-(cGMP)-independent enhancement of endothelial cell proliferation. Whether nitrite itself has the potential to affect myoblast proliferation and metabolism with or without activation of the canonical NO/cGMP pathway to subsequently support muscle cell regeneration is not known. Here we show that nitrite increases proliferation and metabolic activity of murine cultured myoblasts dose-dependently. This effect is not abolished by the NO scavenger 2-(4-carboxy-phenyl)-4,4,5,5-tetramethylimida-zoline-1-oxyl-3 oxide and does not affect intracellular cGMP levels, implicating a cGMP-independent mechanism. Nitrite circumvents the rapamycin induced attenuation of myoblast proliferation and enhances mTOR activity. Our results provide evidence for a novel potential physiological and therapeutic approach of nitrite in skeletal muscle regeneration processes under normoxia independent of NO and cGMP.
Collapse
Affiliation(s)
- Matthias Totzeck
- Division of Cardiology, Pulmonology and Vascular Medicine, Department of Medicine, Medical Faculty, University Hospital Duesseldorf, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | | | | | | | | | | |
Collapse
|
9
|
Stem cell transplantation for muscular dystrophy: the challenge of immune response. BIOMED RESEARCH INTERNATIONAL 2014; 2014:964010. [PMID: 25054157 PMCID: PMC4098613 DOI: 10.1155/2014/964010] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 06/05/2014] [Indexed: 01/03/2023]
Abstract
Treating muscle disorders poses several challenges to the rapidly evolving field of regenerative medicine. Considerable progress has been made in isolating, characterizing, and expanding myogenic stem cells and, although we are now envisaging strategies to generate very large numbers of transplantable cells (e.g., by differentiating induced pluripotent stem cells), limitations directly linked to the interaction between transplanted cells and the host will continue to hamper a successful outcome. Among these limitations, host inflammatory and immune responses challenge the critical phases after cell delivery, including engraftment, migration, and differentiation. Therefore, it is key to study the mechanisms and dynamics that impair the efficacy of cell transplants in order to develop strategies that can ultimately improve the outcome of allogeneic and autologous stem cell therapies, in particular for severe disease such as muscular dystrophies. In this review we provide an overview of the main players and issues involved in this process and discuss potential approaches that might be beneficial for future regenerative therapies of skeletal muscle.
Collapse
|
10
|
The skeletal muscle arachidonic acid cascade in health and inflammatory disease. Nat Rev Rheumatol 2014; 10:295-303. [PMID: 24468934 DOI: 10.1038/nrrheum.2014.2] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Muscle atrophy and weakness are often observed in patients with chronic inflammatory diseases, and are the major clinical features of the autoimmune myopathies, polymyositis and dermatomyositis. A general understanding of the pathogenesis of muscle atrophy and the impaired muscle function associated with chronic inflammatory diseases has not been clarified. In this context, arachidonic acid metabolites, such as the prostaglandin and leukotriene subfamilies, are of interest because they contribute to immune and nonimmune processes. Accumulating evidence suggests that prostaglandins and leukotrienes are involved in causing muscular pain and inflammation, and also in myogenesis and the repair of muscles. In this Review, we summarize novel findings that implicate prostaglandins and leukotrienes in the muscle atrophy and weakness that occur in inflammatory diseases of the muscles, with a focus on inflammatory myopathies. We discuss the role of the arachidonic acid cascade in skeletal muscle growth and function, and individual metabolites as potential therapeutic targets for the treatment of inflammatory muscle diseases.
Collapse
|
11
|
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) are widely consumed by athletes worldwide, despite growing evidence for a negative influence on the adaptation of skeletal muscle to exercise, at least in young healthy individuals. This review focuses on the potential of NSAIDs to alter the activity of satellite cells, the muscle stem cell responsible for repair and maintenance of skeletal muscle. The signaling pathways that are potentially modified by NSAID exposure are also considered. Growth factors as well as inflammatory cells and connective tissue appear to be key factors in the response of muscle under conditions where cyclooxygenase and prostaglandin activity are blocked through NSAID ingestion or infusion. Discrepancies in the literature regarding the response of young and old individuals are addressed, where it appears that the elderly may benefit from NSAID ingestion, although this clearly requires further study. The long-term implications for the muscle of the apparent inhibitory effect of NSAIDs on satellite cells in younger individuals are not clear, and it is possible these may first become apparent with chronic use in athletes training at a high level or with advancing age. Reports of the potential for NSAIDs to alter prostaglandin and growth factor signaling provide a basis for further study of the mechanism of NSAID action on satellite cells.
Collapse
Affiliation(s)
- Abigail L. Mackey
- Institute of Sports Medicine, Department of Orthopaedic Surgery M, Bispebjerg Hospital, and Centre for Healthy Ageing, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
12
|
Ultrasound Modulates the Inflammatory Response and Promotes Muscle Regeneration in Injured Muscles. Ann Biomed Eng 2013; 41:1095-105. [DOI: 10.1007/s10439-013-0757-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Accepted: 01/29/2013] [Indexed: 11/30/2022]
|
13
|
Abstract
Multiple cellular and molecular processes are rapidly activated following skeletal muscle damage to restore normal muscle structure and function. These processes typically involve an inflammatory response and potentially the consequent occurrence of secondary damage before their resolution and the completion of muscle repair or regeneration. The overall outcome of the inflammatory process is potentially divergent, with the induction of prolonged inflammation and further muscle damage, or its active termination and the promotion of muscle repair and regeneration. The final, detrimental, or beneficial effect of the inflammatory response on muscle repair is influenced by specific interactions between inflammatory and muscle cell-derived cytokines that act as positive and/or negative regulators to coordinate local and systemic inflammatory-related events and modulate muscle repair process. A crucial balance between proinflammatory and anti-inflammatory cytokines appears to attenuate an excessive inflammatory reaction, prevent the development of muscle fibrosis, and adequately promote the regenerative process. In this review, we address the interactive cytokine responses following muscle damage, in the context of induction and progression, or resolution of muscle inflammation and the promotion of muscle repair.
Collapse
|
14
|
Duchesne E, Tremblay MH, Côté CH. Mast cell tryptase stimulates myoblast proliferation; a mechanism relying on protease-activated receptor-2 and cyclooxygenase-2. BMC Musculoskelet Disord 2011; 12:235. [PMID: 21999702 PMCID: PMC3207928 DOI: 10.1186/1471-2474-12-235] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 10/14/2011] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Mast cells contribute to tissue repair in fibrous tissues by stimulating proliferation of fibroblasts through the release of tryptase which activates protease-activated receptor-2 (PAR-2). The possibility that a tryptase/PAR-2 signaling pathway exists in skeletal muscle cell has never been investigated. The aim of this study was to evaluate whether tryptase can stimulate myoblast proliferation and determine the downstream cascade. METHODS Proliferation of L6 rat skeletal myoblasts stimulated with PAR-2 agonists (tryptase, trypsin and SLIGKV) was assessed. The specificity of the tryptase effect was evaluated with a specific inhibitor, APC-366. Western blot analyses were used to evaluate the expression and functionality of PAR-2 receptor and to assess the expression of COX-2. COX-2 activity was evaluated with a commercial activity assay kit and by measurement of PGF2α production. Proliferation assays were also performed in presence of different prostaglandins (PGs). RESULTS Tryptase increased L6 myoblast proliferation by 35% above control group and this effect was completely inhibited by APC-366. We confirmed the expression of PAR-2 receptor in vivo in skeletal muscle cells and in satellite cells and in vitro in L6 cells, where PAR-2 was found to be functional. Trypsin and SLIGKV increased L6 cells proliferation by 76% and 26% above control, respectively. COX-2 activity was increased following stimulation with PAR-2 agonist but its expression remained unchanged. Inhibition of COX-2 activity by NS-398 abolished the stimulation of cell proliferation induced by tryptase and trypsin. Finally, 15-deoxy-Δ-12,14-prostaglandin J2 (15Δ-PGJ2), a product of COX-2-derived prostaglandin D2, stimulated myoblast proliferation, but not PGE2 and PGF2α. CONCLUSIONS Taken together, our data show that tryptase can stimulate myoblast proliferation and this effect is part of a signaling cascade dependent on PAR-2 activation and on the downstream activation of COX-2.
Collapse
Affiliation(s)
- Elise Duchesne
- CHUQ Research Center and Faculty of Medicine, Laval University, 2705 boul, Laurier, Québec, Québec G1V 4G2, Canada
| | | | | |
Collapse
|
15
|
Loell I, Lundberg IE. Can muscle regeneration fail in chronic inflammation: a weakness in inflammatory myopathies? J Intern Med 2011; 269:243-57. [PMID: 21205023 DOI: 10.1111/j.1365-2796.2010.02334.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Idiopathic inflammatory myopathies (IIMs), collectively termed myositis, include three major subgroups: polymyositis, dermatomyositis and inclusion body myositis. IIMs are characterized clinically by muscle weakness and reduced muscle endurance preferentially affecting the proximal skeletal muscle. In typical cases, inflammatory cell infiltrates and proinflammatory cytokines, alarmins and eicosanoids are present in muscle tissue. Treatment with glucocorticoids and other immunosuppressants results in improved performance, but complete recovery is rarely seen. The mechanisms that cause muscle weakness and reduced muscle endurance are multi-factorial, and different mechanisms predominate in different phases of disease. It is likely that a combination of immune-mediated and nonimmune-mediated mechanisms contributes to clinical muscle symptoms. Immune-mediated mechanisms include immune cell-mediated muscle fibre necrosis as well as direct effects of various cytokines on muscle fibre contractility. Among the nonimmune-mediated mechanisms, an acquired metabolic myopathy and so-called endoplasmic reticulum stress may be important. There is also a possibility of defective repair mechanisms, with an influence of both disease-related factors and glucocorticoid treatment. Several proinflammatory molecules observed in muscle tissue of myositis patients, including interleukin (IL)-1, IL-15, tumour necrosis factor, high-mobility group box-1 and eicosanoids, have a role in muscle fibre regeneration, and blocking these molecule may impair muscle repair and recovery. The delicate balance between immunosuppressive treatment to downregulate proinflammatory molecules and an inhibitory effect on muscle fibre regeneration needs to be further understood. This would also be relevant for other chronic inflammatory diseases.
Collapse
Affiliation(s)
- I Loell
- Department of Medicine, Karolinska University Hospital, Solna, Stockholm, Sweden
| | | |
Collapse
|
16
|
Nitric oxide regulates stretch-induced proliferation in C2C12 myoblasts. J Muscle Res Cell Motil 2010; 31:215-25. [DOI: 10.1007/s10974-010-9227-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Accepted: 08/05/2010] [Indexed: 10/19/2022]
|
17
|
Tidball JG, Villalta SA. Regulatory interactions between muscle and the immune system during muscle regeneration. Am J Physiol Regul Integr Comp Physiol 2010; 298:R1173-87. [PMID: 20219869 DOI: 10.1152/ajpregu.00735.2009] [Citation(s) in RCA: 780] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent discoveries reveal complex interactions between skeletal muscle and the immune system that regulate muscle regeneration. In this review, we evaluate evidence that indicates that the response of myeloid cells to muscle injury promotes muscle regeneration and growth. Acute perturbations of muscle activate a sequence of interactions between muscle and inflammatory cells. The initial inflammatory response is a characteristic Th1 inflammatory response, first dominated by neutrophils and subsequently by CD68(+) M1 macrophages. M1 macrophages can propagate the Th1 response by releasing proinflammatory cytokines and cause further tissue damage through the release of nitric oxide. Myeloid cells in the early Th1 response stimulate the proliferative phase of myogenesis through mechanisms mediated by TNF-alpha and IL-6; experimental prolongation of their presence is associated with delayed transition to the early differentiation stage of myogenesis. Subsequent invasion by CD163(+)/CD206(+) M2 macrophages attenuates M1 populations through the release of anti-inflammatory cytokines, including IL-10. M2 macrophages play a major role in promoting growth and regeneration; their absence greatly slows muscle growth following injury or modified use and inhibits muscle differentiation and regeneration. Chronic muscle injury leads to profiles of macrophage invasion and function that differ from acute injuries. For example, mdx muscular dystrophy yields invasion of muscle by M1 macrophages, but their early invasion is accompanied by a subpopulation of M2a macrophages. M2a macrophages are IL-4 receptor(+)/CD206(+) cells that reduce cytotoxicity of M1 macrophages. Subsequent invasion of dystrophic muscle by M2c macrophages is associated with progression of the regenerative phase in pathophysiology. Together, these findings show that transitions in macrophage phenotype are an essential component of muscle regeneration in vivo following acute or chronic muscle damage.
Collapse
Affiliation(s)
- James G Tidball
- Molecular, Cellular and Integrative Physiology Program, Department of Integrative Biology and Physiology, University of California-Los Angeles, CA 90095-1606, USA.
| | | |
Collapse
|
18
|
Monda M, Vicidomini C, Viggiano A, Sampaolo S, Di Iorio G, Viggiano A, Viggiano E, De Luca B. Inhibition of prostaglandin synthesis reduces the induction of MyoD expression in rat soleus muscle. J Muscle Res Cell Motil 2009; 30:139-44. [DOI: 10.1007/s10974-009-9182-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Accepted: 05/19/2009] [Indexed: 10/20/2022]
|
19
|
Korotkova M, Helmers SB, Loell I, Alexanderson H, Grundtman C, Dorph C, Lundberg IE, Jakobsson PJ. Effects of immunosuppressive treatment on microsomal prostaglandin E synthase 1 and cyclooxygenases expression in muscle tissue of patients with polymyositis or dermatomyositis. Ann Rheum Dis 2007; 67:1596-602. [PMID: 18094001 PMCID: PMC2582339 DOI: 10.1136/ard.2007.079525] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVES To investigate the expression of microsomal prostaglandin E (PGE) synthase 1 (mPGES-1) and cyclooxygenase (COX) in muscle biopsies from patients with polymyositis or dermatomyositis before and after conventional immunosuppressive treatment. METHODS mPGES-1 and COX expression was evaluated by immunohistochemistry in muscle tissue from healthy individuals and from patients with polymyositis or dermatomyositis before and after conventional immunosuppressive treatment. The number of inflammatory cell infiltrates, T lymphocytes and macrophages was estimated before and after treatment. To localise the mPGES-1 expression double immunofluorescence was performed with antibodies against mPGES-1, CD3, CD68, CD163 and a fibroblast marker. A functional index was used to assess muscle function. RESULTS In patients with myositis, mPGES-1, COX-2 and COX-1 expression was significantly higher compared to healthy individuals and associated with inflammatory cells. Double immunofluorescence demonstrated a predominant expression of mPGES-1 in macrophages. Conventional immunosuppressive treatment resulted in improved but still lower muscle function than normal. A decreased number of CD68-positive macrophages and reduced COX-2 expression in muscle tissue was also seen. By contrast, following the same treatment no significant changes were observed in muscle tissue regarding number of infiltrates, T lymphocytes, CD163-positive macrophages or mPGES-1 protein levels. CONCLUSIONS Increased expression of mPGES-1, COX-1 and COX-2 at protein level was observed in muscle tissue from patients with myositis compared to healthy individuals. Conventional immunosuppressive treatment led to a significant downregulation of COX-2 in myositis muscle tissue. However, the expression of mPGES-1 and COX-1 remained unchanged indicating a role of these enzymes in the chronicity of these diseases.
Collapse
Affiliation(s)
- M Korotkova
- Department of Medicine, Rheumatology Unit, Karolinska Institutet/Karolinska University Hospital, Solna, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Mackey AL, Kjaer M, Dandanell S, Mikkelsen KH, Holm L, Døssing S, Kadi F, Koskinen SO, Jensen CH, Schrøder HD, Langberg H. The influence of anti-inflammatory medication on exercise-induced myogenic precursor cell responses in humans. J Appl Physiol (1985) 2007; 103:425-31. [PMID: 17463304 DOI: 10.1152/japplphysiol.00157.2007] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The consumption of nonsteroidal anti-inflammatory drugs (NSAIDs) is widespread among athletes when faced with muscle soreness or injury, but the effects of NSAIDs on satellite cell activity in humans are unknown. To investigate this, 14 healthy male endurance athletes (mean peak oxygen consumption 62 ml·kg−1·min−1) volunteered for the study, which involved running 36 km. They were divided into two groups and received either 100 mg indomethacin per day or placebo. Muscle biopsies collected before the run and on days 1, 3, and 8 afterward were analyzed for satellite cells by immunohistochemistry with the aid of neural cell adhesion molecule (NCAM) and fetal antigen-1 (FA1) antibodies. Muscle biopsies were also collected from untrained individuals for comparison. Compared with preexercise levels, a 27% increase in the number of NCAM+ cells was observed on day 8 postexercise in the placebo group ( P < 0.05), while levels remained similar at all time points in the NSAID group. No change was seen in the proportion of FA1+ cells, although lower levels were found in the muscle of endurance-trained athletes compared with untrained individuals ( P < 0.05). These results suggest that ingestion of anti-inflammatory drugs attenuates the exercise-induced increase in satellite cell number, supporting the role of the cyclooxygenase pathway in satellite cell activity.
Collapse
Affiliation(s)
- Abigail L Mackey
- Institute of Sports Medicine Copenhagen, Bispebjerg Hospital, Bldg. 8, Bispebjerg Bakke 23, 2400 Copenhagen NV, Denmark.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Bondesen BA, Jones KA, Glasgow WC, Pavlath GK. Inhibition of myoblast migration by prostacyclin is associated with enhanced cell fusion. FASEB J 2007; 21:3338-45. [PMID: 17488951 DOI: 10.1096/fj.06-7070com] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Satellite cells are stem cells that are critical for the formation and growth of skeletal muscle during myogenesis. To differentiate and fuse, proliferating satellite cells or myoblasts must migrate and establish stable cell-cell contacts. However, the factors that regulate myoblast migration and fusion are not understood completely. We have identified PGI2 as a novel regulator of myogenesis in vitro. PGI2 is a member of the family of prostaglandins (PG), autocrine/paracrine signaling molecules synthesized via the cyclooxygenase-1 and -2 pathways. Primary mouse muscle cells both secrete PGI2 and express the PGI2 receptor, IP, at various stages of myogenesis. Using genetic and pharmacological approaches, we show that PGI2 is a negative regulator of myoblast migration that also enhances cell fusion. Thus, PGI2 may act as a "brake" on migrating cells to facilitate cell-cell contact and fusion. Together, our results highlight the importance of the balance between positive and negative regulators in cell migration and myogenesis. This work may have implications for migration of other populations of adult stem cells and/or cells that undergo fusion.
Collapse
Affiliation(s)
- Brenda A Bondesen
- Emory University School of Medicine, Department of Pharmacology, Atlanta, GA 30322, USA
| | | | | | | |
Collapse
|
22
|
Bondesen BA, Mills ST, Pavlath GK. The COX-2 pathway regulates growth of atrophied muscle via multiple mechanisms. Am J Physiol Cell Physiol 2006; 290:C1651-9. [PMID: 16467402 DOI: 10.1152/ajpcell.00518.2005] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Loss of muscle mass occurs with disease, injury, aging, and inactivity. Restoration of normal muscle mass depends on myofiber growth, the regulation of which is incompletely understood. Cyclooxygenase (COX)-2 is one of two isoforms of COX that catalyzes the synthesis of prostaglandins, paracrine hormones that regulate diverse physiological and pathophysiological processes. Previously, we demonstrated that the COX-2 pathway regulates early stages of myofiber growth during muscle regeneration. However, whether the COX-2 pathway plays a common role in adult myofiber growth or functions specifically during muscle regeneration is unknown. Therefore, we examined the role of COX-2 during myofiber growth following atrophy in mice. Muscle atrophy was induced by hindlimb suspension (HS) for 2 wk, followed by a reloading period, during which mice were treated with either the COX-2-selective inhibitor SC-236 (6 mg x kg(-1) x day(-1)) or vehicle. COX-2 protein was expressed and SC-236 attenuated myofiber growth during reloading in both soleus and plantaris muscles. Attenuated myofiber growth in the soleus was associated with both decreased myonuclear addition and decreased inflammation, whereas neither of these processes mediated the effects of SC-236 on plantaris growth. In addition, COX-2(-/-) satellite cells exhibited impaired activation/proliferation in vitro, suggesting direct regulation of muscle cell activity by COX-2. Together, these data suggest that the COX-2 pathway plays a common regulatory role during various types of muscle growth via multiple mechanisms.
Collapse
Affiliation(s)
- Brenda A Bondesen
- Emory Univ. School of Medicine, Dept. of Pharmacology, O. W. Rollins Research Bldg., Atlanta, GA 30322, USA
| | | | | |
Collapse
|
23
|
Dupouy VM, Ferre PJ, Uro-Coste E, Lefebvre HP. Time course of COX-1 and COX-2 expression during ischemia-reperfusion in rat skeletal muscle. J Appl Physiol (1985) 2006; 100:233-9. [PMID: 16357083 DOI: 10.1152/japplphysiol.00673.2004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The aim of this study was to assess cyclooxygenase (COX)-1 and COX-2 expression in skeletal muscle after an ischemia-reperfusion (I/R). Male Sprague-Dawley rats were subjected to unilateral hindlimb ischemia for 2 h and then euthanized after 0, 1, 2, 4, 6, 10, 24, and 72 h of reperfusion. The COX protein and mRNA were assessed in control and injured gastrocnemius muscle. Muscle damage was indirectly determined by plasma creatine kinase activity and edema by weighing wet muscle. Creatine kinase activity in plasma increased as early as 1 h after reperfusion and returned to control levels by 72 h of reperfusion. Edema was observed at 6 and 10 h of reperfusion, but histological investigations showed an absence of tissular inflammatory cell infiltration. COX-1 mRNA was expressed in control muscle and was increased at 72 h of reperfusion, but the levels of associated COX-1 protein detected in control and injured gastrocnemius muscle were similar. COX-2 mRNA was not, or only slightly, detectable in control muscle and after I/R. In contrast, I/R induced major overexpression of COX-2 immunoreactivity at 6 and 10 h of reperfusion with a maximum at 10 h, whereas COX-2 protein was undetectable in control muscle. In conclusion, hindlimb I/R induced a large overexpression of COX-2 but not COX-1 protein between 6 and 10 h after injury. These results suggest a role for COX-2 enzyme in such pathophysiological conditions of the skeletal muscle.
Collapse
Affiliation(s)
- V M Dupouy
- Unité Mixte de Recherche 181 de Physiopathologie et Toxicologic Expérimentales, Institut National de la Recherche Agronomique/Ecole Nationale Vétérinaire de Toulouse, France.
| | | | | | | |
Collapse
|
24
|
Otis JS, Burkholder TJ, Pavlath GK. Stretch-induced myoblast proliferation is dependent on the COX2 pathway. Exp Cell Res 2005; 310:417-25. [PMID: 16168411 DOI: 10.1016/j.yexcr.2005.08.009] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2005] [Revised: 08/11/2005] [Accepted: 08/12/2005] [Indexed: 12/15/2022]
Abstract
Skeletal muscle increases in size due to weight bearing loads or passive stretch. This growth response is dependent in part upon myoblast proliferation. Although skeletal muscles are responsive to mechanical forces, the effect on myoblast proliferation remains unknown. To investigate the effects of mechanical stretch on myoblast proliferation, primary myoblasts isolated from Balb/c mice were subjected to 25% cyclical uniaxial stretch for 5 h at 0.5 Hz. Stretch stimulated myoblast proliferation by 32% and increased cell number by 41% 24 and 48 h after stretch, respectively. COX2 mRNA increased 3.5-fold immediately poststretch. Prostaglandin E2 and F2alpha increased 2.4- and 1.6-fold 6 h after stretch, respectively. Because COX2 has been implicated in regulating muscle growth and regeneration, we hypothesized that stretched myoblasts may proliferate via a COX2-dependent mechanism. We employed two different models to disrupt COX2 activity: (1) treatment with a COX2-selective drug, and (2) transgenic mice null for COX2. Treating myoblasts with a COX2-specific inhibitor blocked stretch-induced proliferation. Likewise, stretched COX2-/- myoblasts failed to proliferate compared to controls. However, supplementing stretched, COX2-/- myoblasts with prostaglandin E2 or fluprostenol increased proliferation. These data suggest that the COX2 pathway is critical for myoblast proliferation in response to stretch.
Collapse
Affiliation(s)
- Jeffrey S Otis
- Emory University School of Medicine, Department of Pharmacology, O.W. Rollins Research Building, Room 5027, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
25
|
Mendias CL, Tatsumi R, Allen RE. Role of cyclooxygenase-1 and -2 in satellite cell proliferation, differentiation, and fusion. Muscle Nerve 2004; 30:497-500. [PMID: 15372441 DOI: 10.1002/mus.20102] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Skeletal muscle satellite cells play an important role in muscle regeneration. Previous work has suggested that nonsteroidal anti-inflammatory drugs may inhibit their activity. We cultured skeletal muscle satellite cells from 9-month-old Sprague-Dawley rats and exposed them to naproxen sodium (a nonselective cyclooxygenase inhibitor), NS-398 (a selective cyclooxygenase-2 inhibitor), and SC-560 (a selective cyclooxygenase-1 inhibitor) for 96 h. Cyclooxygenase-2 inhibition alone resulted in decreased satellite cell proliferation, and inhibition of both cyclooxygenase-1 and cyclooxygenase-2 resulted in decreased satellite cell differentiation and fusion. This study suggests that the cyclooxygenase enzymes appear to play an important part in satellite cell proliferation, differentiation, and fusion and that nonsteroidal anti-inflammatory medication may have an adverse effect on muscle regeneration following injury. The use of a selective cyclooxygenase-2 inhibitor over nonspecific cyclooxygenase inhibitors in the treatment of muscle injuries is not supported.
Collapse
|
26
|
Bondesen BA, Mills ST, Kegley KM, Pavlath GK. The COX-2 pathway is essential during early stages of skeletal muscle regeneration. Am J Physiol Cell Physiol 2004; 287:C475-83. [PMID: 15084473 DOI: 10.1152/ajpcell.00088.2004] [Citation(s) in RCA: 212] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Skeletal muscle regeneration comprises several overlapping cellular processes, including inflammation and myogenesis. Prostaglandins (PGs) may regulate muscle regeneration, because they modulate inflammation and are involved in various stages of myogenesis in vitro. PG synthesis is catalyzed by different isoforms of cyclooxygenase (COX), which are inhibited by nonsteroidal anti-inflammatory drugs. Although experiments employing nonsteroidal anti-inflammatory drugs have implicated PGs in tissue repair, how PGs regulate muscle regeneration remains unclear, and the potentially distinct roles of different COX isoforms have not been investigated. To address these questions, a localized freeze injury was induced in the tibialis anterior muscles of mice chronically treated with either a COX-1- or COX-2-selective inhibitor (SC-560 and SC-236, respectively), starting before injury. The size of regenerating myofibers was analyzed at time points up to 5 wk after injury and found to be decreased by SC-236 and in COX-2−/−muscles, but unaffected by SC-560. In contrast, SC-236 had no effect on myofiber growth when administered starting 7 days after injury. The attenuation of myofiber growth by SC-236 treatment and in COX-2−/−muscles is associated with decreases in the number of myoblasts and intramuscular inflammatory cells at early times after injury. Together, these data suggest that COX-2-dependent PG synthesis is required during early stages of muscle regeneration and thus raise caution about the use of COX-2-selective inhibitors in patients with muscle injury or disease.
Collapse
Affiliation(s)
- Brenda A Bondesen
- Dept. of Pharmacology, Emory University School of Medicine, Rm. 5024, O.W. Rollins Research Bldg., Atlanta, GA 30322, USA
| | | | | | | |
Collapse
|
27
|
Abmayr SM, Balagopalan L, Galletta BJ, Hong SJ. Cell and molecular biology of myoblast fusion. INTERNATIONAL REVIEW OF CYTOLOGY 2003; 225:33-89. [PMID: 12696590 DOI: 10.1016/s0074-7696(05)25002-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In organisms from Drosophila to mammals, the musculature is comprised of an elaborate array of distinct fibers that are generated by the fusion of committed myoblasts. These muscle fibers differ from each other in features that include location, pattern of innervation, site of attachment, and size. The sizes of the newly formed muscles of an embryo are controlled in large part by the number of cells that form the syncitial fiber. Over the past few decades, an extensive body of literature has described the process of myoblast fusion in vertebrates, relying primarily on the strengths of tissue culture model systems. More recently, genetic studies in Drosophila embryos have provided new insights into the process. Together, these studies define the steps necessary for myoblast differentiation, the acquisition of fusion competence, the recognition and adhesion between myoblasts, and the fusion of two lipid bilayers into one. In this review, we have attempted to combine insights from both Drosophila and vertebrate studies to trace the processes and molecules involved in myoblast fusion. Implicit in this approach is the assumption that fundamental aspects of myoblast fusion will be similar, independent of the organism in which it is occurring.
Collapse
MESH Headings
- Animals
- Cell Adhesion/physiology
- Cell Differentiation/physiology
- Cell Membrane/metabolism
- Drosophila melanogaster/embryology
- Drosophila melanogaster/metabolism
- Drosophila melanogaster/ultrastructure
- Embryo, Nonmammalian/embryology
- Embryo, Nonmammalian/metabolism
- Embryo, Nonmammalian/ultrastructure
- Humans
- Membrane Fusion/physiology
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/ultrastructure
- Muscle, Skeletal/embryology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/ultrastructure
- Myoblasts, Skeletal/metabolism
- Myoblasts, Skeletal/ultrastructure
Collapse
Affiliation(s)
- Susan M Abmayr
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | | | | | | |
Collapse
|
28
|
Naro F, De Arcangelis V, Sette C, Ambrosio C, Komati H, Molinaro M, Adamo S, Nemoz G. A bimodal modulation of the cAMP pathway is involved in the control of myogenic differentiation in l6 cells. J Biol Chem 2003; 278:49308-15. [PMID: 14506285 DOI: 10.1074/jbc.m306941200] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have previously shown that myogenesis induction by Arg8-vasopressin (AVP) in L6 rat myoblasts involves a sustained stimulation of type 4 cAMP-phosphodiesterase. In this model, we observed that a transient cAMP generation occurs in the minutes following AVP addition. Evidence suggests that cAMP generation is due to the prostaglandins produced in response to AVP binding to V1a receptors and subsequent activation of phospholipase A2. The early cAMP increase was effective in activating cAMP-dependent protein kinase (PKA) and increasing phosphorylation of CREB transcription factor. Inhibition of PKA by compound H89 prior to AVP addition led to a significant reduction of expression of the differentiation marker creatine kinase, whereas H89 added 1-5 h after AVP had no significant effect. Furthermore, PKA inhibition 24 h after the beginning of AVP treatment potentiated differentiation. This shows that both an early activation and a later down-regulation of the cAMP pathway are required for AVP induction of myogenesis. Because phosphodiesterase PDE4D3 overexpressed in L6 cells lost its ability to potentiate AVP-induced differentiation when mutated and rendered insensitive to PKA phosphorylation and activation, we hypothesize that the early cAMP increase is required to trigger the down-regulation of cAMP pathway through stimulation of phosphodiesterase.
Collapse
Affiliation(s)
- Fabio Naro
- Dipartimento di Istologia ed Embriologia Medica, Università "La Sapienza," 00161 Rome, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Horsley V, Pavlath GK. Prostaglandin F2(alpha) stimulates growth of skeletal muscle cells via an NFATC2-dependent pathway. J Cell Biol 2003; 161:111-8. [PMID: 12695501 PMCID: PMC2172881 DOI: 10.1083/jcb.200208085] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Skeletal muscle growth requires multiple steps to form large multinucleated muscle cells. Molecules that stimulate muscle growth may be therapeutic for muscle loss associated with aging, injury, or disease. However, few factors are known to increase muscle cell size. We demonstrate that prostaglandin F2alpha (PGF2alpha) as well as two analogues augment muscle cell size in vitro. This increased myotube size is not due to PGF2alpha-enhancing cell fusion that initially forms myotubes, but rather to PGF2alpha recruiting the fusion of cells with preexisting multinucleated cells. This growth is mediated through the PGF2alpha receptor (FP receptor). As the FP receptor can increase levels of intracellular calcium, the involvement of the calcium-regulated transcription factor nuclear factor of activated T cells (NFAT) in mediating PGF2alpha-enhanced cell growth was examined. We show that NFAT is activated by PGF2alpha, and the isoform NFATC2 is required for PGF2alpha-induced muscle cell growth and nuclear accretion, demonstrating the first intersection between prostaglandin receptor activation and NFAT signaling. Given this novel role for PGF2alpha in skeletal muscle cell growth, these studies raise caution that extended use of drugs that inhibit PG production, such as nonsteroidal antiinflammatory drugs, may be deleterious for muscle growth.
Collapse
MESH Headings
- Active Transport, Cell Nucleus/drug effects
- Active Transport, Cell Nucleus/genetics
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/adverse effects
- Calcium/metabolism
- Calcium Signaling/drug effects
- Calcium Signaling/genetics
- Cell Differentiation/drug effects
- Cell Differentiation/physiology
- Cell Size/drug effects
- Cell Size/genetics
- Cells, Cultured
- DNA-Binding Proteins/deficiency
- DNA-Binding Proteins/genetics
- Dinoprost/analogs & derivatives
- Dinoprost/metabolism
- Dinoprost/pharmacology
- Growth Substances/metabolism
- Growth Substances/pharmacology
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/growth & development
- Muscle, Skeletal/metabolism
- NFATC Transcription Factors
- Nuclear Proteins
- Protein Isoforms/deficiency
- Protein Isoforms/genetics
- Receptors, Prostaglandin/drug effects
- Receptors, Prostaglandin/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Transcription Factors/deficiency
- Transcription Factors/genetics
- Transcription, Genetic/drug effects
- Transcription, Genetic/genetics
Collapse
Affiliation(s)
- Valerie Horsley
- Cell and Developmental Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | |
Collapse
|
30
|
Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, Benhaim P, Lorenz HP, Hedrick MH. Multilineage cells from human adipose tissue: implications for cell-based therapies. TISSUE ENGINEERING 2001; 7:211-28. [PMID: 11304456 DOI: 10.1089/107632701300062859] [Citation(s) in RCA: 5650] [Impact Index Per Article: 245.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Future cell-based therapies such as tissue engineering will benefit from a source of autologous pluripotent stem cells. For mesodermal tissue engineering, one such source of cells is the bone marrow stroma. The bone marrow compartment contains several cell populations, including mesenchymal stem cells (MSCs) that are capable of differentiating into adipogenic, osteogenic, chondrogenic, and myogenic cells. However, autologous bone marrow procurement has potential limitations. An alternate source of autologous adult stem cells that is obtainable in large quantities, under local anesthesia, with minimal discomfort would be advantageous. In this study, we determined if a population of stem cells could be isolated from human adipose tissue. Human adipose tissue, obtained by suction-assisted lipectomy (i.e., liposuction), was processed to obtain a fibroblast-like population of cells or a processed lipoaspirate (PLA). These PLA cells can be maintained in vitro for extended periods with stable population doubling and low levels of senescence. Immunofluorescence and flow cytometry show that the majority of PLA cells are of mesodermal or mesenchymal origin with low levels of contaminating pericytes, endothelial cells, and smooth muscle cells. Finally, PLA cells differentiate in vitro into adipogenic, chondrogenic, myogenic, and osteogenic cells in the presence of lineage-specific induction factors. In conclusion, the data support the hypothesis that a human lipoaspirate contains multipotent cells and may represent an alternative stem cell source to bone marrow-derived MSCs.
Collapse
Affiliation(s)
- P A Zuk
- Laboratory for Regenerative Bioengineering and Repair, UCLA School of Medicine, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Gentili C, Cermelli S, Tacchetti C, Cossu G, Cancedda R, Descalzi Cancedda F. Expression of the extracellular fatty acid binding protein (Ex-FABP) during muscle fiber formation in vivo and in vitro. Exp Cell Res 1998; 242:410-8. [PMID: 9683528 DOI: 10.1006/excr.1998.4098] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We report that Ex-FABP, an extracellular protein belonging to the lipocalin family and involved in the extracellular transport of long-chain fatty acids, is expressed in the forming myotubes both in vivo and in vitro. The presence of the protein and of the mRNA was observed in newly formed myotubes at early stages of chick embryo development by immunohistochemistry and by in situ hybridization. At later stages of development myofibers still expressed both the mRNA and the protein. Ex-FABP expression was observed also in the developing myocardium and the muscular layer of large blood vessels. In agreement with these findings, an initial expression of the mRNA and protein secretion by cultured chicken myoblasts were observed only after the onset of myoblast fusion. Double-immunofluorescence staining of these cultured cells revealed that multinucleate myotubes were stained by antibodies directed against both the Ex-FABP and the sarcomeric myosin, whereas immature myotubes and single myoblasts were not. When added to cultured myoblasts, antibodies against the Ex-FABP induced a strong enhancement of the production of the same protein. In all experiments some cell sufferance and a transient impairment of myotube formation were also observed. The finding that the continuous removal of the Ex-FABP from the culture medium of myoblasts, due to the formation of immune complexes, resulted in an overproduction of the protein suggests a feedback (autocrine) control during myotube differentiation and maturation. We propose that the requirement for increased transport and metabolism of free fatty acid released from the membrane phospholipids and storage lipids, mediated by Ex-FABP, may be essential during differentiation of multinucleated myotubes or that an increased local demand of fatty acids and metabolites may act as a local hormone in tissues differentiating and undergoing morphogenesis.
Collapse
Affiliation(s)
- C Gentili
- Istituto Nazionale per la Ricerca sul Cancro, Centro di Biotecnologie Avanzate, Genoa, Italy
| | | | | | | | | | | |
Collapse
|
32
|
Hardiman O, Sweeney DJ, Keenan AK. Adenylyl cyclase activity in clonally derived human myoblast cultures: evidence for myoblast heterogeneity. Neuromuscul Disord 1996; 6:283-91. [PMID: 8887958 DOI: 10.1016/0960-8966(96)00355-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In vitro myogenesis recapitulates the programme of myogenesis in vivo. During the process of muscle differentiation, cAMP plays an important role in the control of gene expression and in the integration of metabolic functions. cAMP generation may be affected by drugs or hormones that interact with the membrane-bound enzyme adenylyl cyclase, including adrenergic agents and glucocorticoids. In this study, adenylyl cyclase activity was evaluated in membranes prepared from human clonally derived muscle cultures. In control cultures, there was considerable inter-clonal variation in basal, sodium-fluoride and forskolin-stimulated adenylyl cyclase activity. Cultures differed in their response to steroids: adenylyl cyclase activity was markedly enhanced in some clones, and was significantly inhibited in other clones. Pre-treatment of cultures with pertussis toxin indicated that the effects of steroids are mediated in part by modulation of G-protein activity. These findings indicate a substantial heterogeneity among myoblast clones with respect to the modulating effect of steroids on adenylyl cyclase activity. This observation may account for the conflicting reports of steroid effects on muscle in vitro, and may be of relevance to the understanding of possible transmembrane signalling alterations in muscle disease.
Collapse
Affiliation(s)
- O Hardiman
- Dept. Human Anatomy & Physiology, University college Dublin, Ireland
| | | | | |
Collapse
|
33
|
Rossi A, Santoro MG. Induction by prostaglandin A1 of haem oxygenase in myoblastic cells: an effect independent of expression of the 70 kDa heat shock protein. Biochem J 1995; 308 ( Pt 2):455-63. [PMID: 7772027 PMCID: PMC1136947 DOI: 10.1042/bj3080455] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Prostaglandins of the A type (PGA) induce the synthesis of 70 kDa heat shock proteins (hsp70) in a large variety of mammalian cells. Induction of hsp70 has been associated with a cytoprotective effect of PGA1 after virus infection or thermal injury. In the present report we provide evidence that, in murine myoblasts, PGA1 is not able to induce hsp70 expression, whereas it increases the synthesis of the constitutive protein, hsc70, and dramatically induces the synthesis of a 32 kDa protein (p32). The p32 protein has been identified as haem oxygenase. PGA1 acts at the transcriptional level by inducing haem oxygenase mRNA synthesis, and the signal for induction appears to be associated with decreased intracellular GSH levels. Haem oxygenase, a low-molecular-mass stress protein induced in mammalian cells by oxidant stress, is known to be part of a general inducible antioxidant defence pathway. The fact that prostaglandin synthesis is stimulated in muscle during contraction and in the heart in response to ischaemia raises the possibility that induction of haem oxygenase by PGA in myoblasts could be part of a protective mechanisms in operation during stress and hypoxia.
Collapse
Affiliation(s)
- A Rossi
- Institute of Experimental Medicine, CNR, Rome, Italy
| | | |
Collapse
|
34
|
Hume R, Bell J, Gourlay M, Giles M, Hallas A, Cossar D, Kelly R. Prostaglandin production and metabolism in self-differentiating human fetal lung organ culture. Exp Lung Res 1993; 19:361-76. [PMID: 8319605 DOI: 10.3109/01902149309064352] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
PGE2 and PGF2 alpha are released into the media of human fetal lung organ cultures in decreasing amounts with time. This decline in PGs is not due to culture failure or loss of synthetic capacity, which can be stimulated by fetal bovine serum, nor is it due to increased catabolism of PGE2 to 13,14-dihydro-15-keto-PGE2 (PGEM) or of PGF2 alpha to 13,14-dihydro-15-keto-PGF2 alpha (PGFM). Immunohistochemically reactive PGs are not retained within lung cells. Antisera against methyl-moximated derivatives of PGEM or PGFM and preceded by derivatization on tissue sections of PGs by methyl-moximation not only demonstrate the localization of PGEM and PGFM in epithelial cells and blood vessels, but also show an overall decline in immunoreactivity with time. In addition electron microscopy of uncultured fetal lung removed directly after termination reveals various degrees of mitochondrial damage and in some cases plasma membrane blebs which resolve during the period in culture and as fetal lung self-differentiates. It is proposed that oxidative and mechanical stresses, occurring during termination of pregnancy or tissue preparation, result in cell damage and increased lung prostaglandin production, which, although decreasing during culture as cells recover, is sufficient to trigger terminal self-differentiation.
Collapse
Affiliation(s)
- R Hume
- Department of Child Health, University of Dundee, Scotland
| | | | | | | | | | | | | |
Collapse
|
35
|
Grounds MD, Yablonka-Reuveni Z. Molecular and cell biology of skeletal muscle regeneration. MOLECULAR AND CELL BIOLOGY OF HUMAN DISEASES SERIES 1993; 3:210-56. [PMID: 8111541 DOI: 10.1007/978-94-011-1528-5_9] [Citation(s) in RCA: 84] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- M D Grounds
- Department of Pathology, University of Western Australia, Queen Elizabeth II Medical Centre, Perth
| | | |
Collapse
|
36
|
Hume R, Cossar D, Kelly R, Giles M, Hallas A, Gourlay M, Bell J. Prostaglandins PGE2 and PGF2 alpha in human fetal lung: immunohistochemistry and release from organ culture. Exp Lung Res 1992; 18:259-73. [PMID: 1374024 DOI: 10.3109/01902149209031684] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Immunohistochemical studies in human fetal lung have shown that epithelial and endothelial cells are both strongly and equally reactive for PGE2. In contrast, epithelial PGF2 alpha reactivity varied between fetuses, in some as intense as endothelial staining and in others very much less. As lung organ cultures differentiated, the intensity of PGE2 staining declined in airways and blood vessels, although it was still weakly positive at 10 days. In contrast, epithelial cells rapidly became negative for PGF2 alpha, whereas PGF2 alpha positivity was retained in blood vessels, albeit less obviously. PGF2 alpha and PGE2 were released into the media of organ cultures in decreasing amounts as cultures progressed. Amounts of released PGF2 alpha were greater by 2- to 10-fold than PGE2. Our findings suggest that the endogenous production of prostaglandins by human fetal lung in organ culture has a key role in the self-differentiation process that occurs in the absence of sera or added growth factors or hormones.
Collapse
Affiliation(s)
- R Hume
- Department of Child Life and Health, University of Edinburgh, Scotland
| | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Prostaglandins are known to affect myoblast proliferation and fusion in vitro and are putative regulators of in vivo myogenesis. The levels of E and F alpha series prostaglandins in the thigh muscles of chicken embryos were measured by radioimmunoassays and correlated with indicators of muscle development. Just prior to the onset of secondary myogenesis, the amounts of PGE1, PGE2 and PGF1 alpha plus PGF2 alpha per mg of protein were high. In temporal association with myotube formation, the amount of PGE1 and PGE2 per mg of protein decreased. PGF alpha levels also fell, but at a slower rate than observed with the E series prostaglandins. The decreases in the amounts of prostaglandins per mg protein appeared to be due to a decline in the total amount of prostaglandin within each muscle. These observations are consistent with prostaglandins being one of the factors that controls in vivo muscle formation.
Collapse
Affiliation(s)
- I S McLennan
- Department of Anatomy, University of Otago Medical School, Dunedin, New Zealand
| |
Collapse
|
38
|
Hume R, Kelly R, Cossar D, Giles M, Hallas A, Gourlay M, Bell J. Self-differentiation of human fetal lung organ culture: the role of prostaglandins PGE2 and PGF2 alpha. Exp Cell Res 1991; 194:111-7. [PMID: 2015844 DOI: 10.1016/0014-4827(91)90138-k] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Addition of PGE2, but not PGF2 alpha, to fetal lung organ cultures accelerates the process of self-differentiation with increased dilatation of terminal airsacs and differentiation of the epithelial lining. Indomethacin reduces the endogenous production by organ cultures of PGE2, PGF2 alpha, 13,14-dihydro-15-keto-PGE2, and 13,14-dihydro-15-keto-PGF2 alpha and retards the process of self-differentiation. Prolonged exposure of cultures to indomethacin results in cell necrosis. Indomethacin inhibition of self-differentiation can be reversed and accelerated by the addition of PGE2. Addition of PGF2 alpha in the presence of indomethacin prevents indomethacin-associated cell necrosis but does not accelerate dilatation or differentiation beyond that of cultures in sera-free media without additions. We propose that the endogenous production of PGE2 is a key process in the mechanism of self-differentiation of human fetal lung in organ culture.
Collapse
Affiliation(s)
- R Hume
- Department of Child Life and Health, University of Edinburgh, Scotland, U.K
| | | | | | | | | | | | | |
Collapse
|
39
|
Gaillard D, Wabitsch M, Pipy B, Négrel R. Control of terminal differentiation of adipose precursor cells by glucocorticoids. J Lipid Res 1991. [DOI: 10.1016/s0022-2275(20)42044-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
40
|
Kimura I, Nakagawa M, Kobayashi S, Kimura M. Effects of prostaglandin E2 and cyclooxygenase inhibitors on clustering and level of nicotinic acetylcholine receptor in mouse myotubes co-cultured with spinal cord explant. Int J Dev Neurosci 1991; 9:357-63. [PMID: 1950650 DOI: 10.1016/0736-5748(91)90057-s] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The clustering and level of nicotinic acetylcholine receptor (n-AChR) in cultured mouse myotubes are negatively controlled by endogenous phospholipase A2 (PLA2) (Kimura et al., Int. J. Devl. Neurosci. 5, 127-133, 1987). The effects of PLA2-related metabolites, prostaglandins, leukotrienes and platelet-activating factor (PAF) were investigated using fluorescein isothiocyanate-alpha-bungarotoxin. Peak and total fluorescence within a cluster were used as indices of clustering and level of n-AChR, respectively. Prostaglandin E2 (PGE2, 1-10 microM) decreased both indices in a concentration-dependent manner. Aspirin and indomethacin, cyclooxygenase inhibitors, increased the indices at 1.0 microM and 10-30 nM, and decreased them at higher concentrations of 10-30 microM and 0.1-1 microM, respectively. Prostaglandin F2 alpha (PGF2 alpha, 1-10 microM), nordihydroguaiaretic acid (30 microM), a lipoxygenase inhibitor, and PAF (10 microM) had no effect. These results suggest that the control of endogenous PLA2 on the clustering and level of n-AChR is due to PGE2, but not to PGF2 alpha, leukotrienes or PAF.
Collapse
Affiliation(s)
- I Kimura
- Department of Chemical Pharmacology, Faculty of Pharmaceutical Sciences, Toyama Medical and Pharmaceutical University, Japan
| | | | | | | |
Collapse
|
41
|
Abstract
The involvement of Ca2+ and PGE1 in myoblast fusion has been well documented. Extracellular Ca2+ is essential for myoblast adhesion, alignment, and fusion. There is an obligatory increase in Ca2+ influx immediately preceding fusion and the Ca2+ ionophore A23187 promotes precocious fusion. PGE1 receptors appear just prior to fusion, and an antagonist of PGE1 binding to cell surface receptors blocks fusion when added prior to Ca2+ influx. Finally, exogenous PGE1 induces precocious fusion. The present study was an initial test of the hypothesis that membrane protein phosphorylation by protein kinase C (PKC) links PGE1 receptor occupancy and the increase in Ca2+ influx. Our conclusion that PKC is an essential component in the regulation of myoblast fusion is based in part on the following evidence: (1) an activator of PKC, the tumor promoter 12-O-tetradecanoylphorbol-13-acetate (TPA), at low concentration and for a brief exposure period, induces precocious fusion and stimulates Ca2+ influx; (2) 4 alpha-phorbol-12,13-didecanoate, an inactive analog of TPA, has no discernible effect on fusion or Ca2+ influx; (3) 1-oleoyl-2-acetyl glycerol, an analog of endogenous diacylglycerol (DAG) which activates PKC, promotes precocious fusion, as does the DAG kinase inhibitor R59022 (6-[2-[4-[(4-fluorophenyl)phenylmethylene]-1-piperidinyl]ethyl]-7- methyl-5H-thiazole-[3,2 alpha]-pyrimidin-5-one) which raises the level of endogenous DAG by inhibiting its catabolism; (4) 1-(5-isoquinolinesulfonyl)-2-methylpiperazine (H-7), a highly potent PKC inhibitor, reversibly blocks myogenesis at a point between alignment and fusion; and (5) H-7 also blocks the normal increase in Ca2+ influx preceding fusion.
Collapse
Affiliation(s)
- J D David
- Division of Biological Sciences, University of Missouri, Columbia 65211
| | | | | |
Collapse
|
42
|
Ralph RK, Wojcik S. Inhibitors of lipoxygenase have antiproliferative effects on P815 murine mastocytoma cells. Cancer Lett 1990; 49:181-5. [PMID: 2107964 DOI: 10.1016/0304-3835(90)90156-r] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The effect of a variety of inhibitors of prostaglandin synthase, lipoxygenase and phospholipase on the growth of P815 murine mastocytoma cells was examined. Only lipoxygenase inhibitors substantially reduced growth, presumably by inhibiting the production of arachidonic acid metabolites rather than causing arachidonate accumulation since excess arachidonic acid did not reverse growth inhibition. Evidence is presented that production of leukotrienes B4, C4, D4 or E4 was not involved. Other metabolites of arachidonic acid were not excluded. A role for lipoxygenase in growth signal transduction in these and other cells is suggested.
Collapse
Affiliation(s)
- R K Ralph
- Department of Cellular and Molecular Biology, University of Auckland, New Zealand
| | | |
Collapse
|
43
|
Palmer RM. Prostaglandins and the control of muscle protein synthesis and degradation. Prostaglandins Leukot Essent Fatty Acids 1990; 39:95-104. [PMID: 2188265 DOI: 10.1016/0952-3278(90)90017-f] [Citation(s) in RCA: 59] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- R M Palmer
- Division of Biochemistry, Rowett Research Institute, Bucksburn, Aberdeen, UK
| |
Collapse
|
44
|
Palmer RM, Bain PA, Southorn BG. Dexamethasone-induced catabolism and insulin resistance in L6 myoblasts are reversed by the removal of serum. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. C, COMPARATIVE PHARMACOLOGY AND TOXICOLOGY 1990; 97:369-72. [PMID: 1710557 DOI: 10.1016/0742-8413(90)90156-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
1. One hundred nanomolar dexamethasone reduced protein synthesis by 16% and also decreased the accretion of protein and RNA in L6 myoblasts when foetal calf serum was present; these effects were reversed when serum was omitted from the medium. 2. Insulin (100 microU/ml) increased protein synthesis, protein accretion and RNA accretion both in the presence and the absence of serum. 3. Dexamethasone inhibited the effects of 100 microU insulin/ml in the presence of serum and induced insulin resistance; in the presence of 25 or 100 nM dexamethasone insulin was ineffective at concentrations below 250 microU and 1 mU/ml respectively.
Collapse
Affiliation(s)
- R M Palmer
- Rowett Research Institute, Bucksburn, Aberdeen, Scotland
| | | | | |
Collapse
|
45
|
Peplow P, Jeremy JY. Antiestrogens: structure, mode of action and relationship to eicosanoids. Prostaglandins Leukot Essent Fatty Acids 1989; 37:241-54. [PMID: 2682672 DOI: 10.1016/0952-3278(89)90035-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- P Peplow
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | | |
Collapse
|
46
|
Palmer RM, Bain PA. Indomethacin inhibits the insulin-induced increases in RNA and protein synthesis in L6 skeletal muscle myoblasts. PROSTAGLANDINS 1989; 37:193-203. [PMID: 2471230 DOI: 10.1016/0090-6980(89)90056-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Rates of accretion of RNA and protein and rates of protein synthesis were measured in sub-confluent cultures of L6 myoblasts. Insulin (100 microU/ml) stimulated protein synthesis by 15% within 30 min and by 40% at two and six hours. By six hours insulin also increased the accretion of RNA (+15%). The cyclo-oxygenase inhibitor indomethacin did not reduce the basal rate of RNA or protein accretion in L6 cells but reduced the rate of protein synthesis by 16%. When added together with insulin, indomethacin inhibited the hormonally-stimulated rate of protein synthesis and also significantly reduced the accretion of RNA. Indomethacin still reduced the effects of insulin on protein synthesis when added to the cells two hours after the hormone. Synthesis of RNA measured by the incorporation of [3H]-uridine was also stimulated by insulin but was inhibited by indomethacin only when the drug was present throughout the incubation. Inhibition of protein synthesis by cyclo-oxygenase inhibitors may be the result of both a direct action on translational efficiency and an effect on RNA synthesis.
Collapse
Affiliation(s)
- R M Palmer
- Rowett Research Institute, Aberdeen, Scotland
| | | |
Collapse
|
47
|
Grigoriadis AE, Heersche JN, Aubin JE. Differentiation of muscle, fat, cartilage, and bone from progenitor cells present in a bone-derived clonal cell population: effect of dexamethasone. J Cell Biol 1988; 106:2139-51. [PMID: 3384856 PMCID: PMC2115137 DOI: 10.1083/jcb.106.6.2139] [Citation(s) in RCA: 452] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
RCJ 3.1, a clonally derived cell population isolated from 21-d fetal rat calvaria, expresses the osteoblast-associated characteristics of polygonal morphology, a cAMP response to parathyroid hormone, synthesis of predominantly type I collagen, and the presence of 1,25-dihydroxyvitamin D3-regulated alkaline phosphatase activity. When cultured in the presence of ascorbic acid, sodium beta-glycerophosphate, and the synthetic glucocorticoid dexamethasone, this clone differentiated in a time-dependent manner into four morphologically distinct phenotypes of known mesenchymal origin. Multinucleated muscle cells were observed as early as 9-10 d in culture, lipid-containing adipocytes formed after 12 d, chondrocyte nodules were observed after 16 d, and mineralized bone nodules formed after 21 d in culture. The differentiated cell types were characterized morphologically, histochemically, and immunohistochemically. The formation of adipocytes and chondrocytes was dependent upon the addition of dexamethasone; the muscle and bone phenotypes were also expressed at low frequency in the absence of dexamethasone. The sex steroid hormones progesterone and 17 beta-estradiol had no effect on differentiation in this system, suggesting that the effects of dexamethasone represent effects specific for glucocorticosteroids. Increasing concentrations of dexamethasone (10(-9)-10(-6) M) increased the numbers of myotubes, adipocytes, and chondrocytes; however, when present continuously for 35 d, the lower concentrations appeared to better maintain the muscle and adipocyte phenotypes. Bone nodules were not quantitated because the frequency of bone nodule formation was too low. Single cells obtained by plating RCJ 3.1 cells at limiting dilutions in the presence of dexamethasone, were shown to give rise to subclones that could differentiate into either single or multiple phenotypes. Thus, the data suggest that this clonal cell line contains subpopulations of mesenchymal progenitor cells which can, under the influence of glucocorticoid hormones, differentiate in vitro into four distinct cell types. It is, therefore, a unique cell line which will be of great use in the study of the regulation of mesenchymal stem cell differentiation.
Collapse
Affiliation(s)
- A E Grigoriadis
- Medical Research Council Group in Periodontal Physiology, University of Toronto, Ontario, Canada
| | | | | |
Collapse
|
48
|
Entwistle A, Zalin RJ, Warner AE, Bevan S. A role for acetylcholine receptors in the fusion of chick myoblasts. J Biophys Biochem Cytol 1988; 106:1703-12. [PMID: 3372592 PMCID: PMC2115056 DOI: 10.1083/jcb.106.5.1703] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The role of acetylcholine receptors in the control of chick myoblast fusion in culture has been explored. Spontaneous fusion of myoblasts was inhibited by the nicotinic acetylcholine receptor antagonists alpha-bungarotoxin, Naja naja toxin and monoclonal antibody mcAb 5.5. The muscarinic antagonists QNB and n-methyl scopolamine were without effect. Atropine had no effect below 1 microM, where it blocks muscarinic receptors; at higher concentrations, when it blocks nicotinic receptors also, atropine inhibited myoblast fusion. The inhibitions imposed by acetylcholine receptor antagonists lasted for approximately 12 h; fusion stimulated by other endogenous substances then took over. The inhibition was limited to myoblast fusion. The increases in cell number, DNA content, the level of creatine phosphokinase activity (both total and muscle-specific isozyme) and the appearance of heavy chain myosin, which accompany muscle differentiation, followed a normal time course. Pre-fusion myoblasts, fusing myoblasts, and young myotubes specifically bound labeled alpha-bungarotoxin, indicating the presence of acetylcholine receptors. The nicotinic acetylcholine receptor agonist, carbachol, induced uptake of [14C]Guanidinium through the acetylcholine receptor. Myoblasts, aligned myoblasts and young myotubes expressed the synthetic enzyme Choline acetyltransferase and stained positively with antibodies against acetylcholine. The appearance of ChAT activity in myogenic cultures was prevented by treatment with BUDR; nonmyogenic cells in the cultures expressed ChAT at a level which was too low to account for the activity in myogenic cultures. We conclude that activation of the nicotinic acetylcholine receptor is part of the mechanism controlling spontaneous myoblast fusion and that myoblasts synthesize an endogenous, fusion-inducing agent that activates the nicotinic ACh receptor.
Collapse
Affiliation(s)
- A Entwistle
- Department of Anatomy and Developmental Biology, University College London, United Kingdom
| | | | | | | |
Collapse
|