101
|
Peviani SM, Gomes ARS, Moreira RFC, Moriscot AS, Salvini TF. Short bouts of stretching increase myo-D, myostatin and atrogin-1 in rat soleus muscle. Muscle Nerve 2007; 35:363-70. [PMID: 17143883 DOI: 10.1002/mus.20695] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Stretching is widely used in rehabilitation and sports activities to improve joint range-of-motion and flexibility in humans, but the effect of stretching on the gene expression of skeletal muscle is poorly understood. We evaluated the effect of short bouts of passive stretching of rat soleus muscle on myo-D, myostatin, and atrogin-1 gene expressions. Six groups of animals were submitted to a single session of stretching (10 stretches of 1 minute with 30 seconds of rest between them, performed manually) and were evaluated immediately (I), and 8, 24, 48, 72, and 168 hours after the session. To evaluate the effect of repetitive sessions of stretching on the soleus muscle over 1 week, three groups of animals received a single session per day of stretching and the muscle was evaluated immediately after 2, 3, and 7 sessions. The mRNA levels of myo-D, myostatin, and atrogin-1 were determined by real-time polymerase chain reaction. A single session of stretching increased the mRNA levels of myo-D (after 24 h), myostatin (I, and 168 h later), and atrogin-1 (after 48 h). Repeated daily session of stretching over 1 week increased myostatin (after 7 sessions) and atrogin-1 expression (after 2, 3, and 7 sessions). Thus, short bouts of passive stretching are able to increase the gene expression of factors associated with muscle growth (myo-D), negative regulation of muscle mass (myostatin), and atrophy (atrogin-1), indicating muscle remodeling through different pathways.
Collapse
Affiliation(s)
- Sabrina Messa Peviani
- Unit of Skeletal Muscle Plasticity, Department of Physical Therapy, Federal University of São Carlos, Rodovia Washington Luís, Km 235, CEP 13565-905, São Carlos, SP, Brazil
| | | | | | | | | |
Collapse
|
102
|
|
103
|
Brzóska E, Grabowska I, Hoser G, Stremińska W, Wasilewska D, Machaj EK, Pojda Z, Moraczewski J, Kawiak J. Participation of stem cells from human cord blood in skeletal muscle regeneration of SCID mice. Exp Hematol 2006; 34:1262-70. [PMID: 16939819 DOI: 10.1016/j.exphem.2006.05.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2005] [Revised: 04/28/2006] [Accepted: 05/10/2006] [Indexed: 10/24/2022]
Abstract
OBJECTIVE In this report, we demonstrate the participation of human cord blood (HUCB) stem cells in the skeletal muscle regeneration of SCID (severe combined immunodeficient) mice. MATERIALS AND METHODS The HUCB cells were labeled with the PKH26 fluorescent marker or recognized by an anti-HLA-ABC or anti-beta-2-microglobulin antibody. The HUCB cells were implanted directly into the damaged mouse muscle. The regeneration process and the implanted HUCB cells were traced each day after the damage, throughout a period of 7 days, and additionally at day 30 with the use of flow cytometry and confocal microscopy. RESULTS The PKH26-labeled cells isolated from the regenerating muscle were positive for the anti-HLA-ABC antibody. The percentage of the PKH26(+) and HLA-ABC(+) cells decreased from day 1 to day 5. In the regenerating muscle, the percentage of the HLA-ABC(+) cells increased, as measured on days 7 and 30. Moreover, myofibers containing fragments of the PKH26-labeled sarcolemma were noticed. Labeling with the anti-human beta(2)-microglobulin antibody showed the presence of positive cells and myofibers at day 7 of the regeneration, suggesting fusion of human and mouse cells. CONCLUSIONS We suggest that the HUCB cells implanted into the damaged muscle are present there for at least 30 days and that they participate in the muscle regeneration. Moreover, our study shows that the implanted HUCB cells form human muscle precursor cells residing in the repaired mouse muscle. We suggest that the HUCB cell circulation after transplantation depends on SDF-1 (stromal-derived factor-1) expression in regenerating muscle.
Collapse
Affiliation(s)
- Edyta Brzóska
- Department of Clinical Cytology, Medical Center of Postgraduate Education, Warsaw, Poland
| | | | | | | | | | | | | | | | | |
Collapse
|
104
|
Zammit PS, Partridge TA, Yablonka-Reuveni Z. The skeletal muscle satellite cell: the stem cell that came in from the cold. J Histochem Cytochem 2006; 54:1177-91. [PMID: 16899758 DOI: 10.1369/jhc.6r6995.2006] [Citation(s) in RCA: 462] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The muscle satellite cell was first described and actually named on the basis of its anatomic location under the basement membrane surrounding each myofiber. For many years following its discovery, electron microscopy provided the only definitive method of identification. More recently, several molecular markers have been described that can be used to detect satellite cells, making them more accessible for study at the light microscope level. Satellite cells supply myonuclei to growing myofibers before becoming mitotically quiescent in muscle as it matures. They are then activated from this quiescent state to fulfill their roles in routine maintenance, hypertrophy, and repair of adult muscle. Because muscle is able to efficiently regenerate after repeated bouts of damage, systems must be in place to maintain a viable satellite cell pool, and it was proposed over 30 years ago that self-renewal was the primary mechanism. Self-renewal entails either a stochastic event or an asymmetrical cell division, where one daughter cell is committed to differentiation whereas the second continues to proliferate or becomes quiescent. This classic model of satellite cell self-renewal and the importance of satellite cells in muscle maintenance and repair have been challenged during the past few years as bone marrow-derived cells and various intramuscular populations were shown to be able to contribute myonuclei and occupy the satellite cell niche. This is a fast-moving and dynamic field, however, and in this review we discuss the evidence that we think puts this enigmatic cell firmly back at the center of adult myogenesis.
Collapse
Affiliation(s)
- Peter S Zammit
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL England.
| | | | | |
Collapse
|
105
|
Abstract
Somatic stem cell populations participate in the development and regeneration of their host tissues. Skeletal muscle is capable of complete regeneration due to stem cells that reside in skeletal muscle and nonmuscle stem cell populations. However, in severe myopathic diseases such as Duchenne Muscular Dystrophy, this regenerative capacity is exhausted. In the present review, studies will be examined that focus on the origin, gene expression, and coordinated regulation of stem cell populations to highlight the regenerative capacity of skeletal muscle and emphasize the challenges for this field. Intense interest has focused on cell-based therapies for chronic, debilitating myopathic diseases. Future studies that enhance our understanding of stem cell biology and repair mechanisms will provide a platform for therapeutic applications directed toward these chronic, life-threatening diseases.
Collapse
Affiliation(s)
- Xiaozhong Shi
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | |
Collapse
|
106
|
Gomes AR, Soares AG, Peviani S, Nascimento RB, Moriscot AS, Salvini TF. The effect of 30 minutes of passive stretch of the rat soleus muscle on the myogenic differentiation, myostatin, and atrogin-1 gene expressions. Arch Phys Med Rehabil 2006; 87:241-6. [PMID: 16442979 DOI: 10.1016/j.apmr.2005.08.126] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2005] [Accepted: 08/23/2005] [Indexed: 10/25/2022]
Abstract
UNLABELLED The effect of 30 minutes of passive stretch of the rat soleus muscle on the myogenic differentiation, myostatin, and atrogin-1 gene expressions. OBJECTIVE To evaluate the effect of passive stretch, applied for 30 minutes to the rat soleus muscle, on the myogenic differentiation (myoD), myostatin, and atrogin-1 gene expressions. DESIGN Case-controlled study. SETTING University laboratory. ANIMALS Fifty 12-week-old male Wistar rats. INTERVENTIONS Six groups of animals were given a single stretch bout and were evaluated immediately and 8, 24, 48, 72, and 168 hours later. Another 3 groups were evaluated immediately after 2, 3, and 7 stretches. An intact control group was also analyzed. MAIN OUTCOME MEASURES The messenger ribonucleic acid (mRNA) levels of myoD, myostatin, and atrogin-1 were assessed by real-time polymerase chain reaction. RESULTS Twenty-four hours after a single session of stretch only, the myoD mRNA levels had increased compared with the control group, whereas an increase in the atrogin-1 expression was observed after 2, 3, and 7 stretches. CONCLUSIONS A single session of passive stretch increased the myoD gene expression, a factor related to muscle growth. Interestingly, daily stretches increased the atrogin-1 gene expression, a gene primarily associated with muscle atrophy. The results indicated that gene expression was responsive to the number of stretch sessions.
Collapse
Affiliation(s)
- Anna R Gomes
- Unit of Skeletal Muscle Plasticity, Department of Physical Therapy, Federal University of São Carlos, Brazil
| | | | | | | | | | | |
Collapse
|
107
|
Archer JD, Vargas CC, Anderson JE. Persistent and improved functional gain in mdx dystrophic mice after treatment with L-arginine and deflazacort. FASEB J 2006; 20:738-40. [PMID: 16464957 DOI: 10.1096/fj.05-4821fje] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Although an increase in nitric oxide (NO) in muscle is reported to improve the outcome of deflazacort treatment for mdx mouse muscular dystrophy, the genetic homologue of Duchenne muscular dystrophy (DMD), the impact such treatment on the functional outcomes of the disease, including fiber susceptibility to exercise-induced injury, is not established. Experiments were designed to test whether treatment with deflazacort and L-arginine (a substrate for NO synthase, NOS) would change the extent of fiber injury induced by 24 h of voluntary exercise. The impact of exercise-related injury to induce a secondary regenerative response by muscle was also examined as corroborating evidence of muscle damage. Dystrophic mdx mice were treated for 3 wk with placebo, deflazacort, or deflazacort plus either L-arginine or N(G)-nitro-L-arginine methyl ester (a NOS inhibitor). Deflazacort, especially combined with L-arginine, spared quadriceps muscle from injury-induced regeneration (myf5 expression) compared with placebo treatment, despite an increase in membrane permeability immediately after exercise (assessed by Evans blue dye infiltration). Deflazacort alone prevented the typical progressive loss of function (measured as voluntary distance run over 24 h) that was observed 3 months later in placebo-treated mice. Therefore, combined deflazacort plus L-arginine treatment spared mdx dystrophic limb muscle from exercise-induced damage and the need for regeneration and induced a persistent functional improvement in distance run. Results suggest a potential new treatment option for improving the quality of life for boys with DMD.
Collapse
Affiliation(s)
- Jonathan D Archer
- Department of Human Anatomy and Cell Science, Faculty of Medicine, University of Manitoba, Winnipeg, Canada
| | | | | |
Collapse
|
108
|
Blazevich AJ, Sharp NCC. Understanding Muscle Architectural Adaptation: Macro- and Micro-Level Research. Cells Tissues Organs 2006; 181:1-10. [PMID: 16439814 DOI: 10.1159/000089964] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2005] [Indexed: 12/28/2022] Open
Abstract
Recent research using muscle-imaging techniques has revealed a remarkable plasticity of human muscle architecture where significant changes in fascicle lengths and angles have resulted from the chronic performance, or cessation, of strong muscle contractions. However, there is a paucity of data describing architectural adaptations to chronic stretching, disuse and immobilization, illness, and aging, and those data that are available are equivocal. Understanding their impact is important in order that effective interventions for illness/injury management and rehabilitation, and programs to improve the physical capacity of workers, the aged and athletes can be determined. Nonetheless, recent advances in myocellular research could provide a framework allowing the prediction of architectural changes in these understudied areas. Examination of the site-specific response to mechanical stress of calpain-dependent ubiquitin-proteasome proteolysis, or of the cellular response to stress after the knockout (or incapacitation) of sarcomeric and cytoskeletal proteins involved in cellular signal transduction, provides an exciting paradigm by which myocellular adaptation can be described. Such research might contribute to the understanding of macro-level changes in muscle architecture.
Collapse
Affiliation(s)
- Anthony J Blazevich
- Centre for Sports Medicine and Human Performance, Brunel University, Uxbridge UB8 3PH, UK
| | | |
Collapse
|
109
|
Nattrass GS, Quigley SP, Gardner GE, Bawden CS, McLaughlan CJ, Hegarty RS, Greenwood PL. Genotypic and nutritional regulation of gene expression in two sheep hindlimb muscles with distinct myofibre and metabolic characteristics. ACTA ACUST UNITED AC 2006. [DOI: 10.1071/ar05101] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study investigated whether the expression profile of GDF8 (myostatin), myogenic regulatory factors (MRFs: MYF5, MYOD1, MYOG (myogenin), and MYF6), and IGF-system (IGF1, IGF2, IGF1R) genes are correlated with anatomical muscle, nutrition level, and estimated breeding values (EBVs) for muscling, growth, and/or fatness. Real-time PCR was employed to quantitatively measure the mRNA levels of these genes in the semimembranosus (SM) and semitendinosus (ST) muscles of growing lambs. The lambs were sired by Poll Dorset rams with differing EBVs for growth, muscling, and fatness, and were fed either high or low quality and availability pasture from birth to ~8 months of age. With the exception of MYOD1, the mRNA levels of all genes examined in this study showed varying degrees of nutritional regulation. All the MRF mRNA levels were higher in the SM muscle than the ST muscle, whereas myostatin mRNA was higher in the ST muscle than the SM muscle. Interactions between muscle type and nutrition were detected for IGF2, MYF6, and myogenin, while positive correlations between IGF2 and IGF1R and between MYOD1 and myogenin mRNA levels were apparent in both muscles. At the genotypic level, subtle differences in mRNA levels suggested interactions between nutrition and sire EBV. The findings of this study confirm that the MRFs, IGFs, and myostatin genes are differentially affected by a variety of factors that include nutrition, muscle type, and sire EBVs. Together, these data suggest that this suite of genes has important roles during postnatal muscle growth, even at quite late stages of growth and development.
Collapse
|
110
|
Dhawan J, Rando TA. Stem cells in postnatal myogenesis: molecular mechanisms of satellite cell quiescence, activation and replenishment. Trends Cell Biol 2005; 15:666-73. [PMID: 16243526 DOI: 10.1016/j.tcb.2005.10.007] [Citation(s) in RCA: 333] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2005] [Revised: 08/12/2005] [Accepted: 10/07/2005] [Indexed: 12/13/2022]
Abstract
Satellite cells are the primary stem cells in adult skeletal muscle, and are responsible for postnatal muscle growth, hypertrophy and regeneration. In mature muscle, most satellite cells are in a quiescent state, but they activate and begin proliferating in response to extrinsic signals. Following activation, a subset of satellite cell progeny returns to the quiescent state during the process of self-renewal. Here, we review recent studies of satellite cell biology and focus on the key transitions from the quiescent state to the state of proliferative activation and myogenic lineage progression and back to the quiescent state. The molecular mechanisms of these transitions are considered in the context of the biology of the satellite cell niche, changes with age, and interactions with established pathways of myogenic commitment and differentiation.
Collapse
Affiliation(s)
- Jyotsna Dhawan
- Center for Cellular and Molecular Biology, Uppal Road, Hyderabad-500007, India
| | | |
Collapse
|
111
|
Brack AS, Bildsoe H, Hughes SM. Evidence that satellite cell decrement contributes to preferential decline in nuclear number from large fibres during murine age-related muscle atrophy. J Cell Sci 2005; 118:4813-21. [PMID: 16219688 DOI: 10.1242/jcs.02602] [Citation(s) in RCA: 213] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Skeletal muscle fibres are multinucleate syncitial cells that change size during adult life depending on functional demand. The relative contribution of change in nuclear number and/or cell growth to fibre size change is unclear. We report that nuclei/unit length decreases in larger fibres during skeletal muscle ageing. This leads to an increased size of nuclear domain (quantity of cytoplasm/number of nuclei within that cytoplasm). Initially, larger fibres have more satellite cells than small fibres, but this advantage is lost as satellite cells decline with age. These changes are accompanied by an overall decline in fibre size, returning domain size to the normal range. Exacerbated loss of fibre nuclei per unit length during ageing of myoD-null mice provides the first experimental support for the hypothesis that a satellite cell defect causes inadequate nuclear replacement. We propose a model in which a decline in satellite cell function and/or number during ageing leads to a loss of nuclei from large fibres and an associated domain size increase that triggers cytoplasmic atrophy through the normal cell-size-regulating machinery.
Collapse
Affiliation(s)
- Andrew S Brack
- MRC Centre for Developmental Neurobiology and Randall Division for Cell and Molecular Biophysics, New Hunt's House, King's College London, London, SE1 1UL, UK
| | | | | |
Collapse
|
112
|
Goding GS, Al-Sharif KI, McLoon LK. Myonuclear addition to uninjured laryngeal myofibers in adult rabbits. Ann Otol Rhinol Laryngol 2005; 114:552-7. [PMID: 16134353 DOI: 10.1177/000348940511400711] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVES In normal mature limb skeletal muscle, satellite cells are quiescent and myonuclei do not divide after formation of their associated myofibers in the absence of injury. The possibility of myonuclear addition in uninjured laryngeal myofibers of adult rabbits was investigated in an immunohistochemical pilot study. METHODS Bromodeoxyuridine (brdU), a marker for cell division, was administered by intraperitoneal injection over a 12-hour period in rabbits. The number of brdU-positive myonuclei per myofiber was determined on cross sections through the thyroarytenoid (TA) and posterior cricoarytenoid (PCA) muscles. RESULTS In the TA muscle, 0.13% +/- 0.03% (mean +/- SEM) of the myofibers counted had a brdU-positive nucleus. In the PCA muscle, 0.13% +/- 0.01% of the myofibers counted had a brdU-positive nucleus. Approximately 0.2% and 0.3% of the myofibers of the TA and PCA muscles, respectively, had brdU-positive satellite cells associated with them. Tibialis anterior and pectoralis major muscle controls were negative for brdU-positive myonuclei. CONCLUSIONS These data support the possibility of continuous remodeling in uninjured adult laryngeal myofibers and accentuate the distinct nature of laryngeal muscle relative to limb skeletal muscle in the rabbit model.
Collapse
Affiliation(s)
- George S Goding
- Department of Otolaryngology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | |
Collapse
|
113
|
Young HE, Duplaa C, Katz R, Thompson T, Hawkins KC, Boev AN, Henson NL, Heaton M, Sood R, Ashley D, Stout C, Morgan JH, Uchakin PN, Rimando M, Long GF, Thomas C, Yoon JI, Park JE, Hunt DJ, Walsh NM, Davis JC, Lightner JE, Hutchings AM, Murphy ML, Boswell E, McAbee JA, Gray BM, Piskurich J, Blake L, Collins JA, Moreau C, Hixson D, Bowyer FP, Black AC. Adult-derived stem cells and their potential for use in tissue repair and molecular medicine. J Cell Mol Med 2005; 9:753-69. [PMID: 16202227 PMCID: PMC6741352 DOI: 10.1111/j.1582-4934.2005.tb00510.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
This report reviews three categories of precursor cells present within adults. The first category of precursor cell, the epiblast-like stem cell, has the potential of forming cells from all three embryonic germ layer lineages, e.g., ectoderm, mesoderm, and endoderm. The second category of precursor cell, the germ layer lineage stem cell, consists of three separate cells. Each of the three cells is committed to form cells limited to a specific embryonic germ layer lineage. Thus the second category consists of germ layer lineage ectodermal stem cells, germ layer lineage mesodermal stem cells, and germ layer lineage endodermal stem cells. The third category of precursor cells, progenitor cells, contains a multitude of cells. These cells are committed to form specific cell and tissue types and are the immediate precursors to the differentiated cells and tissues of the adult. The three categories of precursor cells can be readily isolated from adult tissues. They can be distinguished from each other based on their size, growth in cell culture, expressed genes, cell surface markers, and potential for differentiation. This report also discusses new findings. These findings include the karyotypic analysis of germ layer lineage stem cells; the appearance of dopaminergic neurons after implantation of naive adult pluripotent stem cells into a 6-hydroxydopamine-lesioned Parkinson's model; and the use of adult stem cells as transport mechanisms for exogenous genetic material. We conclude by discussing the potential roles of adult-derived precursor cells as building blocks for tissue repair and as delivery vehicles for molecular medicine.
Collapse
Affiliation(s)
- Henry E Young
- Division of Basic Medical Sciences, Department of Pediatrics, Mercer University School of Medicine, Macon, GA 31207, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Aoi S, Shimotake T, Tsuda T, Deguchi E, Iwai N. Impaired expression of myogenic regulatory molecules in the pelvic floor muscles of murine embryos with anorectal malformations. J Pediatr Surg 2005; 40:805-9. [PMID: 15937819 DOI: 10.1016/j.jpedsurg.2005.01.047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
BACKGROUND/PURPOSE Recent biological studies have elucidated the molecular mechanism of muscle development, in which various regulatory factors (myogenic regulatory factors [MRFs]) play key roles during embryogenesis. To investigate the development of anorectal malformations (ARMs), we studied MRF expressions in myogenic cells in the pelvic floor using murine embryos affected with ARM. METHODS Anorectal malformation embryos were obtained from the 10.5th embryonal day (E10.5) to the 7.0th postnatal day (D7.0) in a natural mutant strain (Sd/+, RSV/Le). Serial frozen sections were prepared for immunohistochemistry using specific antibodies to M-cadherin, myoD, Myogenin, myosin heavy chain, and alfa-actin molecule. RESULTS In normal mice, embryonal caudal somites differentiated into myogenic stem cells and migrated to the pelvic floor between E11.0 and E14.0. In the ARM mice, however, caudal somites were irregularly arranged and MRF expressions in myogenic cells were markedly decreased in the dorsocaudal region at E11.5 to E13.0, leading to hypoplastic pelvic floor muscles. CONCLUSIONS The maldevelopment of pelvic floor muscles in ARM is derived from a deficient supply of myogenic stem cells, with impaired MRF expression. These results suggest that myogenic stem cells, available from bone marrow contents, may be used for postnatal muscle regeneration to reinforce the pelvic floor muscle function in children with ARM.
Collapse
Affiliation(s)
- Shigeyoshi Aoi
- Division of Surgery, Children's Research Hospital, Kyoto Prefectural University of Medicine, Kyoto 602-0841, Japan.
| | | | | | | | | |
Collapse
|
115
|
Liu C, McFarland DC, Velleman SG. Effect of genetic selection on MyoD and myogenin expression in turkeys with different growth rates. Poult Sci 2005; 84:376-84. [PMID: 15782905 DOI: 10.1093/ps/84.3.376] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Skeletal muscle development requires the ordered expression of specific myogenic regulatory factors, which include MyoD, Myf5, myogenin, and MRF4. The MyoD and Mrf5 factors are required for the determination of myoblasts, whereas myogenin and MRF4 play a pivotal role in terminal differentiation. In the current study, males and females of a turkey genetic line selected only for increased 16-wk BW (F line) and an unselected randombred control (RBC2 line) from which the F line was developed were used to investigate the developmental expression of MyoD and myogenin mRNA in embryonic pectoralis major muscle and myogenic satellite cells. Pectoralis major muscle was isolated at embryonic d (ED) 14, 16, 18, 20, 22, and 24. The mRNA levels of MyoD and myogenin were measured using a real-time quantitative polymerase chain reaction method. Both MyoD and myogenin expression declined during embryonic development. The decrease in MyoD expression started at ED 16 for the F line and at ED 18 for the RBC2 line for both sexes. Myogenin expression in both lines began to decline at ED 14. The F line males had lower myogenin expression at ED 14, 16, and 18 than the RBC2 line males, which was similar for the F line females compared with the RBC2 line females except there was no significant difference at ED 18. The RBC2 line males had greater expression than the females for myogenin at ED 16 and 18 for the RBC2 line. Proliferating myogenic satellite cells in both lines and sexes expressed low levels of MyoD and myogenin. After the initiation of differentiation in both lines and sexes, there was a sharp surge in MyoD expression at 24 h followed by a decrease at 48 h and then an increase in expression through 72 or 96 h of differentiation. There were line and sex differences in myogenin expression during the differentiation process. These data are suggestive of growth- and sex-related differences in the expression of myogenic regulatory factors key to muscle cell proliferation and differentiation, which will affect muscle growth rate.
Collapse
Affiliation(s)
- C Liu
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, Ohio 44691, USA
| | | | | |
Collapse
|
116
|
Wozniak AC, Kong J, Bock E, Pilipowicz O, Anderson JE. Signaling satellite-cell activation in skeletal muscle: markers, models, stretch, and potential alternate pathways. Muscle Nerve 2005; 31:283-300. [PMID: 15627266 DOI: 10.1002/mus.20263] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Activation of skeletal muscle satellite cells, defined as entry to the cell cycle from a quiescent state, is essential for normal growth and for regeneration of tissue damaged by injury or disease. This review focuses on early events of activation by signaling through nitric oxide and hepatocyte growth factor, and by mechanical stimuli. The impact of various model systems used to study activation and the regulation of satellite-cell quiescence are placed in the context of activation events in other tissues, concluding with a speculative model of alternate pathways signaling satellite-cell activation.
Collapse
Affiliation(s)
- Ashley C Wozniak
- Department of Human Anatomy and Cell Science, University of Manitoba, 730 William Avenue, Winnipeg, Manitoba R3E 0W2, Canada
| | | | | | | | | |
Collapse
|
117
|
Abstract
A population of myogenic progenitors termed satellite cells undertakes postnatal development and repair of skeletal muscle. Studies have indicated that atypical myogenic precursors can also participate in muscle regeneration. The source of this regenerative capacity has been attributed to "adult stem cells" that represent poorly understood multipotent cell lineages, believed to reside in all adult tissue populations. Here we review the origin and location of muscle satellite cells and stem cells, as well as the mechanisms by which they may be specified. We discuss how the experimental models utilized raise important questions regarding the validity of extrapolating these findings.
Collapse
Affiliation(s)
- Iain W McKinnell
- Ottawa Health Research Institute, Ottawa, Ontario, K1H 8L6 Canada
| | | | | |
Collapse
|
118
|
Araya R, Eckardt D, Maxeiner S, Krüger O, Theis M, Willecke K, Sáez JC. Expression of connexins during differentiation and regeneration of skeletal muscle: functional relevance of connexin43. J Cell Sci 2004; 118:27-37. [PMID: 15601660 DOI: 10.1242/jcs.01553] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The molecular mechanisms regulating skeletal muscle regeneration and differentiation are not well understood. We analyzed the expression of connexins (Cxs) 40, 43 and 45 in normal and regenerating tibialis anterior muscle and in primary cultures of differentiating myoblasts in adult and newborn mice, respectively. Cxs 45 and 43, but not 40, were strongly expressed in normal muscle and their expression was upregulated during regeneration. Furthermore, the functional role of Cx43 during differentiation and regeneration was examined after induced deletion of Cx43 in transgenic mice. In vivo, the inducible deletion of Cx43 delayed the formation of myofibers and prolonged the expression of myogenin during regeneration. In primary cultures of satellite cell-derived myoblasts, induced deletion of Cx43 led to decreased expression of myogenin and MyoD, dye coupling, creatine kinase activity and myoblast fusion. Thus, the expression of Cx45 and Cx43 is upregulated during skeletal muscle regeneration and Cx43 is required for normal myogenesis in vitro and adult muscle regeneration in vivo.
Collapse
Affiliation(s)
- Roberto Araya
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | | | | | | | | | | | | |
Collapse
|
119
|
Pizza FX, Peterson JM, Baas JH, Koh TJ. Neutrophils contribute to muscle injury and impair its resolution after lengthening contractions in mice. J Physiol 2004; 562:899-913. [PMID: 15550464 PMCID: PMC1665528 DOI: 10.1113/jphysiol.2004.073965] [Citation(s) in RCA: 167] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We tested the hypotheses that: (1) neutrophil accumulation after contraction-induced muscle injury is dependent on the beta(2) integrin CD18, (2) neutrophils contribute to muscle injury and oxidative damage after contraction-induced muscle injury, and (3) neutrophils aid the resolution of contraction-induced muscle injury. These hypotheses were tested by exposing extensor digitorum longus (EDL) muscles of mice deficient in CD18 (CD18(-/-); Itgb2(tm1Bay)) and of wild type mice (C57BL/6) to in situ lengthening contractions and by quantifying markers of muscle inflammation, injury, oxidative damage and regeneration/repair. Neutrophil concentrations were significantly elevated in wild type mice at 6 h and 3 days post-lengthening contractions; however, neutrophils remained at control levels at these time points in CD18-/- mice. These data indicate that CD18 is required for neutrophil accumulation after contraction-induced muscle injury. Histological and functional (isometric force deficit) signs of muscle injury and total carbonyl content, a marker of oxidative damage, were significantly higher in wild type relative to CD18-/- mice 3 days after lengthening contractions. These data show that neutrophils exacerbate contraction-induced muscle injury. After statistically controlling for differences in the force deficit at 3 days, wild type mice also demonstrated a higher force deficit at 7 days, a lower percentage of myofibres expressing embryonic myosin heavy chain at 3 and 7 days, and a smaller cross sectional area of central nucleated myofibres at 14 days relative to CD18-/- mice. These observations suggest that neutrophils impair the restoration of muscle structure and function after injury. In conclusion, neutrophil accumulation after contraction-induced muscle injury is dependent on CD18. Furthermore, neutrophils appear to contribute to muscle injury and impair some of the events associated with the resolution of contraction-induced muscle injury.
Collapse
Affiliation(s)
- Francis X Pizza
- Dept of Kinesiology, The University of Toledo, 2801 W. Bancroft Street, Toledo, OH 43606, USA.
| | | | | | | |
Collapse
|
120
|
te Pas MFW, Visscher AH, de Greef KH. Molecular genetic and physiologic background of the growth hormone-IGF-I axis in relation to breeding for growth rate and leanness in pigs. Domest Anim Endocrinol 2004; 27:287-301. [PMID: 15451075 DOI: 10.1016/j.domaniend.2004.06.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2004] [Accepted: 06/21/2004] [Indexed: 11/19/2022]
Abstract
The GH-IGF-I axis is of major importance for the regulation of body growth and composition, and cellular proliferation and differentiation processes. Selective breeding aiming to improve growth rate and/or body composition is accompanied by changes of the GH-IGF-I axis. Research aiming to elucidate the genetic and physiologic mechanism(s) underlying these changes may best use single-trait selection lines. Two such pig selection lines, one for growth rate and one for high lean content, were used in experiments to investigate the mechanisms of the GH-IGF-I axis change during selection. This contribution reviews the selection-related changes in the GH-IGF-I axis as the consequences of selection for whole body growth rate or body composition and effects on local tissue growth rate. A model explaining the observed effects and consequences for the pressure on the physiology is presented. In short, selection related demand for GH induces GH synthesis until a limit is reached. After that the pulsatile GH plasma profile changes, which may also affect expression profiles of genes regulating body composition.
Collapse
Affiliation(s)
- Marinus F W te Pas
- Wageningen University, Animal Sciences Group, Division of Animal Resources Development, PO Box 65, 8200 AB Lelystad, The Netherlands.
| | | | | |
Collapse
|
121
|
Abstract
Embryonic myogenesis involves the staged induction of myogenic regulatory factors and positional cues that dictate cell determination, proliferation, and differentiation into adult muscle. Muscle is able to regenerate after damage, and muscle regeneration is generally thought to recapitulate myogenesis during embryogenesis. There has been considerable progress in the delineation of myogenesis pathways during embryogenesis, but it is not known whether the same signaling pathways are relevant to muscle regeneration in adults. Here, we defined the subset of embryogenesis pathways induced in muscle regeneration using a 27 time-point in vivo muscle regeneration series. The embryonic Wnt (Wnt1, 3a, 7a, 11), Shh pathway, and the BMP (BMP2, 4, 7) pathway were not induced during muscle regeneration. Moreover, antagonists of Wnt signaling, sFRP1, sFRP2, and sFRP4 (secreted frizzled-related proteins) were significantly up-regulated, suggesting active inhibition of the Wnt pathway. The pro-differentiation FGFR4 pathway was transiently expressed at day 3, commensurate with expression of MyoD, Myogenin, Myf5, and Pax7. Protein verification studies showed fibroblast growth factor receptor 4 (FGFR4) protein to be strongly expressed in differentiating myoblasts and newly formed myotubes. We present evidence that FGF6 is likely the key ligand for FGFR4 during muscle regeneration, and further suggest that FGF6 is released from necrotic myofibers where it is then sequestered by basal laminae. We also confirmed activation of Notch1 in the regenerating muscle. Finally, known MyoD coactivators (MEF2A, p/CIP, TCF12) and repressors (Twist, Id2) were strongly induced at appropriate time points. Taken together, our results suggest that embryonic positional signals (Wnt, Shh, and BMP) are not induced in postnatal muscle regeneration, whereas cell-autonomous factors (Pax7, MRFs, FGFR4) involving muscle precursor proliferation and differentiation are recapitulated by muscle regeneration.
Collapse
Affiliation(s)
- Po Zhao
- Center for Genetic Medicine, Children's National Medical Center, Washington, DC, USA
| | | |
Collapse
|
122
|
Zammit PS, Carvajal JJ, Golding JP, Morgan JE, Summerbell D, Zolnerciks J, Partridge TA, Rigby PWJ, Beauchamp JR. Myf5 expression in satellite cells and spindles in adult muscle is controlled by separate genetic elements. Dev Biol 2004; 273:454-65. [PMID: 15328025 DOI: 10.1016/j.ydbio.2004.05.038] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2003] [Revised: 05/05/2004] [Accepted: 05/21/2004] [Indexed: 02/01/2023]
Abstract
The myogenic regulatory factor Myf5 is integral to the initiation and control of skeletal muscle formation. In adult muscle, Myf5 is expressed in satellite cells, stem cells of mature muscle, but not in the myonuclei that sustain the myofibre. Using the Myf5(nlacZ/+) mouse, we now show that Myf5 is also constitutively expressed in muscle spindles-stretch-sensitive mechanoreceptors, while muscle denervation induces extensive reactivation of the Myf5 gene in myonuclei. To identify the elements involved in the regulation of Myf5 in adult muscle, we analysed reporter gene expression in a transgenic bacterial artificial chromosome (BAC) deletion series of the Mrf4/Myf5 locus. A BAC carrying 140 kb upstream of the Myf5 transcription start site was sufficient to drive all aspects of Myf5 expression in adult muscle. In contrast, BACs carrying 88 and 59 kb upstream were unable to drive consistent expression in satellite cells, although expression in muscle spindles and reactivation of the locus in myonuclei were retained. Therefore, as during development, multiple enhancers are required to generate the full expression pattern of Myf5 in the adult. Together, these observations show that elements controlling adult Myf5 expression are genetically separable and possibly distinct from those that control Myf5 during development. These studies are a first step towards identifying cognate transcription factors involved in muscle stem cell regulation.
Collapse
Affiliation(s)
- Peter S Zammit
- Muscle Cell Biology Group, Medical Research Council Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, W12 0NN, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Abstract
Development of a multicellular organism is accomplished through a series of events that are preprogrammed in the genome. These events encompass cellular proliferation, lineage commitment, lineage progression, lineage expression, cellular inhibition, and regulated apoptosis. The sequential progression of cells through these events results in the formation of the differentiated cells, tissues, and organs that constitute an individual. Although most cells progress through this sequence during development, a few cells leave the developmental continuum to become reserve precursor cells. The reserve precursor cells are involved in the continual maintenance and repair of the tissues and organs throughout the life span of the individual. Until recently it was generally assumed that the precursor cells in postnatal individuals were limited to lineage-committed progenitor cells specific for various tissues. However, studies by Young, his colleagues, and others have demonstrated the presence of two categories of precursor cells that reside within the organs and tissues of postnatal animals. These two categories of precursor cells are lineage-committed (multipotent, tripotent, bipotent, and unipotent) progenitor cells and lineage-uncommitted pluripotent (epiblastic-like, ectodermal, mesodermal, and endodermal) stem cells. These reserve precursor cells provide for the continual maintenance and repair of the organism after birth.
Collapse
Affiliation(s)
- Henry E Young
- Division of Basic Medical Sciences, Mercer University School of Medicine, Macon, Georgia 31207, USA.
| | | |
Collapse
|
124
|
Dhawan J, Helfman DM. Modulation of acto-myosin contractility in skeletal muscle myoblasts uncouples growth arrest from differentiation. J Cell Sci 2004; 117:3735-48. [PMID: 15252113 DOI: 10.1242/jcs.01197] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Cell-substratum interactions trigger key signaling pathways that modulate growth control and tissue-specific gene expression. We have previously shown that abolishing adhesive interactions by suspension culture results in G0 arrest of myoblasts. We report that blocking intracellular transmission of adhesion-dependent signals in adherent cells mimics the absence of adhesive contacts. We investigated the effects of pharmacological inhibitors of acto-myosin contractility on growth and differentiation of C2C12 myogenic cells. ML7 (5-iodonaphthalene-1-sulfonyl homopiperazine) and BDM (2,3, butanedione monoxime) are specific inhibitors of myosin light chain kinase, and myosin heavy chain ATPase, respectively. ML7 and BDM affected cell shape by reducing focal adhesions and stress fibers. Both inhibitors rapidly blocked DNA synthesis in a dose-dependent, reversible fashion. Furthermore, both ML7 and BDM suppressed expression of MyoD and myogenin, induced p27kip1 but not p21cip1, and inhibited differentiation. Thus, as with suspension-arrest, inhibition of acto-myosin contractility in adherent cells led to arrest uncoupled from differentiation. Over-expression of inhibitors of the small GTPase RhoA (dominant negative RhoA and C3 transferase) mimicked the effects of myosin inhibitors. By contrast, wild-type RhoA induced arrest, maintained MyoD and activated myogenin and p21 expression. The Rho effector kinase ROCK did not appear to mediate Rho's effects on MyoD. Thus, ROCK and MLCK play different roles in the myogenic program. Signals regulated by MLCK are critical, since inhibition of MLCK suppressed MyoD expression but inhibition of ROCK did not. Inhibition of contractility suppressed MyoD but did not reduce actin polymer levels. However, actin depolymerization with latrunculin B inhibited MyoD expression. Taken together, our observations indicate that actin polymer status and contractility regulate MyoD expression. We suggest that in myoblasts, the Rho pathway and regulation of acto-myosin contractility may define a control point for conditional uncoupling of differentiation and the cell cycle.
Collapse
Affiliation(s)
- Jyotsna Dhawan
- Center for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500 007, India.
| | | |
Collapse
|
125
|
Zammit PS, Golding JP, Nagata Y, Hudon V, Partridge TA, Beauchamp JR. Muscle satellite cells adopt divergent fates: a mechanism for self-renewal? ACTA ACUST UNITED AC 2004; 166:347-57. [PMID: 15277541 PMCID: PMC2172269 DOI: 10.1083/jcb.200312007] [Citation(s) in RCA: 671] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Growth, repair, and regeneration of adult skeletal muscle depends on the persistence of satellite cells: muscle stem cells resident beneath the basal lamina that surrounds each myofiber. However, how the satellite cell compartment is maintained is unclear. Here, we use cultured myofibers to model muscle regeneration and show that satellite cells adopt divergent fates. Quiescent satellite cells are synchronously activated to coexpress the transcription factors Pax7 and MyoD. Most then proliferate, down-regulate Pax7, and differentiate. In contrast, other proliferating cells maintain Pax7 but lose MyoD and withdraw from immediate differentiation. These cells are typically located in clusters, together with Pax7−ve progeny destined for differentiation. Some of the Pax7+ve/MyoD−ve cells then leave the cell cycle, thus regaining the quiescent satellite cell phenotype. Significantly, noncycling cells contained within a cluster can be stimulated to proliferate again. These observations suggest that satellite cells either differentiate or switch from terminal myogenesis to maintain the satellite cell pool.
Collapse
Affiliation(s)
- Peter S Zammit
- Muscle Cell Biology Group, Medical Research Council Clinical Sciences Centre, Faculty of Medicine, Imperial College, Hammersmith Hospital Campus, Du Cane Rd., London W12 0NN, UK.
| | | | | | | | | | | |
Collapse
|
126
|
Anderson JE, Wozniak AC. Satellite cell activation on fibers: modeling events in vivo — an invited review. Can J Physiol Pharmacol 2004; 82:300-10. [PMID: 15213729 DOI: 10.1139/y04-020] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Knowledge of the events underlying satellite cell activation and the counterpart maintenance of quiescence is essential for planning therapies that will promote the growth and regeneration of skeletal muscle in healthy, disease and aging. By modeling those events of satellite cell activation in studies of single muscle fibers or muscles in culture, the roles of mechanical stretching and nitric oxide are becoming understood. Recent studies demonstrated that stretch-induced activation is very rapid and exhibits some features of satellite cell heterogeneity. As well, gene expression studies showed that expression of the c-met receptor gene rises rapidly after stretching muscles in culture compared to those without stretch. This change in gene expression during activation, and the maintenance of quiescence in both normal and dystrophic muscles are dependent on NO, as they are blocked by inhibition of nitric oxide synthase (NOS). Mechanical, contractile activity is the defining feature of muscle function. Therefore, ongoing studies of stretch effects in satellite cell activation and quiescence in quiescent fiber and muscle cultures provides appropriate models by which to explore the regulatory steps in muscle in vivo under many conditions related to disease, repair, rehabilitation, growth and the prevention or treatment of atrophy.Key words: regeneration, stretch, myofiber culture, muscular dystrophy, quiescence.
Collapse
Affiliation(s)
- Judy E Anderson
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Canada.
| | | |
Collapse
|
127
|
Storbeck CJ, Daniel K, Zhang YH, Lunde J, Scime A, Asakura A, Jasmin B, Korneluk RG, Sabourin LA. Ste20-like kinase SLK displays myofiber type specificity and is involved in C2C12 myoblast differentiation. Muscle Nerve 2004; 29:553-64. [PMID: 15052621 DOI: 10.1002/mus.20000] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Cell growth and terminal differentiation are controlled by complex signaling cascades that regulate the expression of specific subsets of genes implicated in cell fate and morphogenic processes. We have recently cloned and characterized a novel Ste20-like kinase termed SLK that is associated with adhesion structures during cell adhesion and spreading. However, the specific function of SLK is poorly understood. To gain further insight into the role of SLK, we have characterized its activity, expression, and distribution in skeletal muscle and during the in vitro differentiation of C2C12 myoblasts. Although SLK is expressed ubiquitously in adult tissues, our results show that it is predominantly expressed in muscle masses during development. Furthermore, SLK activity is upregulated during the differentiation of C2C12 myoblasts. In addition, we have found that SLK localizes presynaptically at neuromuscular junctions and that it is preferentially expressed in types I and IIA myofibers at major myofibrillar striations. Supporting a role in myoblast function and differentiation, SLK expression is induced in Myf5- and Pax7-positive activated satellite cells during regeneration and expression of dominant negative SLK in C2C12 cultures impairs myoblast fusion, suggesting a role for SLK in muscle cell differentiation.
Collapse
Affiliation(s)
- Christopher J Storbeck
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Abstract
Under normal circumstances, mammalian adult skeletal muscle is a stable tissue with very little turnover of nuclei. However, upon injury, skeletal muscle has the remarkable ability to initiate a rapid and extensive repair process preventing the loss of muscle mass. Skeletal muscle repair is a highly synchronized process involving the activation of various cellular responses. The initial phase of muscle repair is characterized by necrosis of the damaged tissue and activation of an inflammatory response. This phase is rapidly followed by activation of myogenic cells to proliferate, differentiate, and fuse leading to new myofiber formation and reconstitution of a functional contractile apparatus. Activation of adult muscle satellite cells is a key element in this process. Muscle satellite cell activation resembles embryonic myogenesis in several ways including the de novo induction of the myogenic regulatory factors. Signaling factors released during the regenerating process have been identified, but their functions remain to be fully defined. In addition, recent evidence supports the possible contribution of adult stem cells in the muscle regeneration process. In particular, bone marrow-derived and muscle-derived stem cells contribute to new myofiber formation and to the satellite cell pool after injury.
Collapse
|
129
|
Kadi F, Johansson F, Johansson R, Sjöström M, Henriksson J. Effects of one bout of endurance exercise on the expression of myogenin in human quadriceps muscle. Histochem Cell Biol 2004; 121:329-34. [PMID: 14997318 DOI: 10.1007/s00418-004-0630-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2004] [Indexed: 10/26/2022]
Abstract
The objective of this study was to investigate the cellular localisation of MyoD and myogenin in human skeletal muscle fibres as well as the possible alterations in the expression of MyoD and myogenin in response to a single bout of endurance exercise at 40% and 75% of maximum oxygen uptake (VO(2) max). Twenty-five biopsies (5 per subject) from the vastus lateralis muscle were obtained before exercise, from the exercising leg at 40% and 75% of VO(2) max and from the resting leg following these exercise bouts. The tyramide signal amplification-direct and the Vectastain ABC methods using specific monoclonal antibodies were used to determine the exact location of myogenin and MyoD, to identify muscle satellite cells and to determine myosin heavy chain (MyHC) composition. At rest, myonuclei did not express MyoD or myogenin. Following a single bout of exercise at 40% and 75% of VO(2) max, an accumulation of myogenin in myonuclei and not in satellite cells was observed in biopsies from the exercised leg but not in biopsies before exercise and from the resting leg. The number of myogenin-positive myonuclei varied among individuals indicating differences in the response to a single exercise bout. In conclusion, this immunohistochemical study showed that a rapid rearrangement of myogenin expression occurs in exercised human skeletal muscles in response to a single bout of exercise.
Collapse
Affiliation(s)
- Fawzi Kadi
- Department of Physical Education and Health, Orebro University, 70182, Orebro, Sweden.
| | | | | | | | | |
Collapse
|
130
|
Young HE. Existence of reserve quiescent stem cells in adults, from amphibians to humans. Curr Top Microbiol Immunol 2004; 280:71-109. [PMID: 14594208 DOI: 10.1007/978-3-642-18846-6_2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Several theories have been proposed to explain the phenomenon of tissue restoration in amphibians and higher order animals. These theories include dedifferentiation of damaged tissues, transdifferentiation of lineage-committed stem cells, and activation of quiescent stem cells. Young and colleagues demonstrated that connective tissues throughout the body contain multiple populations of quiescent lineage-committed progenitor stem cells and lineage-uncommitted pluripotent stem cells. Subsequent cloning and cell sorting studies identified quiescent lineage-uncommitted pluripotent mesenchymal stem cells, capable of forming any mesodermal cell type, and pluripotent epiblastic-like stem cells, capable of forming any somatic cell type. Based on their studies, they propose at least 11 categories of quiescent reserve stem cells resident within postnatal animals, including humans. These categories are pluripotent epiblastic-like stem cells, pluripotent ectodermal stem cells, pluripotent epidermal stem cells, pluripotent neuronal stem cells, pluripotent neural crest stem cells, pluripotent mesenchymal (mesodermal) stem cells, pluripotent endodermal stem cells, multipotent progenitor stem cells, tripotent progenitor stem cells, bipotent progenitor stem cells, and unipotent progenitor stem cells. Thus, activation of quiescent reserve stem cells, i.e., lineage-committed progenitor stem cells and lineage-uncommitted pluripotent stem cells, resident within the connective tissues could provide for the continual maintenance and repair of the postnatal organism after birth.
Collapse
Affiliation(s)
- H E Young
- Division of Basic Medical Sciences, Department of Pediatrics, Mercer University School of Medicine, 1550 College Street, Macon, GA 31207, USA.
| |
Collapse
|
131
|
Abstract
The review explains why the myotoxic phospholipases A2 and cardiotoxins are such important tools in the study of the regeneration and maturation of mammalian skeletal muscle. The role of satellite cells as precursors of cell-based regeneration is discussed and recent controversies on the origin of myogenic cells involved in the regeneration of mature skeletal muscle are addressed. This is followed by discussions of sarcomere reconstruction, myosin and sarcoplasmic reticulum ATPase expression, the electrophysiological properties of regenerating muscle, and the reconstruction of the neuromuscular junction. The emphasis throughout is on the plastic changes of major structural and functional proteins that occur during regeneration, and on other influences that determine the final outcome of regenerative activity such as innervation, thyroid status, mechanical work and the functional integrity of the microcirculation. The review closes with a discussion of some of the factors--such as active regeneration--that influence the success of gene-based therapies applied to inherited muscle disease.
Collapse
Affiliation(s)
- J B Harris
- School of Neurology, Neurobiology and Psychiatry, Faculty of Medical Sciences, University of Newcastle upon Tyne, Newcastle upon Tyne NE2 4HH, UK.
| |
Collapse
|
132
|
Tsivitse SK, McLoughlin TJ, Peterson JM, Mylona E, McGregor SJ, Pizza FX. Downhill running in rats: influence on neutrophils, macrophages, and MyoD+ cells in skeletal muscle. Eur J Appl Physiol 2003; 90:633-8. [PMID: 12955516 DOI: 10.1007/s00421-003-0909-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2003] [Indexed: 10/26/2022]
Abstract
The accumulation of neutrophils and macrophages, as well as the activation of satellite cells, are early events following skeletal muscle injury. We examined the temporal relationship between changes in neutrophils, macrophages, and MyoD protein, a marker of satellite cell activation, after injurious exercise. Male rats ( n=47) performed an intermittent downhill (-16% grade) running (17 m/min) protocol and the solei were obtained at 0, 2, 6, 24, 48, or 72 h post-exercise. Neutrophils, macrophages (ED1 and ED2), and MyoD+ cells were determined in muscle cross sections using immunohistochemistry. Downhill running increased ( P<or=0.05) the percentage of injured fibers and elevated blood creatine kinase activity. Neutrophils were elevated 18-fold relative to controls at 24 h post-exercise. ED1 macrophages were elevated four- and twofold at 24 and 48 h post-exercise, respectively. Neither ED2 macrophages nor MyoD+ cells were elevated post-exercise. These observations may indicate that elevations in neutrophils and ED1 macrophages after injurious exercise are not temporally associated with an increase in satellite cell activation.
Collapse
Affiliation(s)
- Susan K Tsivitse
- Department of Kinesiology, The University of Toledo, 2801 W Bancroft St, Toledo, OH 43606, USA
| | | | | | | | | | | |
Collapse
|
133
|
Wozniak AC, Pilipowicz O, Yablonka-Reuveni Z, Greenway S, Craven S, Scott E, Anderson JE. C-Met expression and mechanical activation of satellite cells on cultured muscle fibers. J Histochem Cytochem 2003; 51:1437-1445. [PMID: 14566016 PMCID: PMC3957553 DOI: 10.1177/002215540305101104] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2003] [Accepted: 06/18/2003] [Indexed: 11/15/2022] Open
Abstract
Single-fiber cultures can be used to model satellite cell activation in vivo. Although technical deficiencies previously prevented study of stretch-induced events, here we describe a method developed to study satellite cell gene expression by in situ hybridization (ISH) using protocol modifications for fiber adhesion and fixation. The hypothesis that mechanical stretching activates satellite cells was tested. Fiber cultures were established from normal flexor digitorum brevis muscles and plated on FlexCell dishes with a layer of Vitrogen. After 2 hr of stretch in the presence of BrdU, satellite cells on fibers attached to Vitrogen were activated above control levels. In the absence of activating treatments or mechanical stretch, ISH studies showed 0-6 c-Met+ satellite cells per fiber. Time course experiments demonstrated stable quiescence in the absence of stretch and significant peaks in activation after 30 min and 2 hr of stretch. Frequency distributions for unstretched fiber cultures showed a significantly greater number of quiescent c-Met+ satellite cells than were activated by stretching, suggesting that typical activation stimuli did not trigger cycling in the entire c-Met+ population of satellite cells. These methods have a strong potential to further dissect the nature of stretch-induced activation and gene expression among characterized populations of individual quiescent and activated satellite cells.
Collapse
Affiliation(s)
- Ashley C Wozniak
- Department of Human Anatomy and Cell Science, Faculty of Medicine, University of Manitoba, 730 William Avenue, Winnipeg, Manitoba R3E 0W3, Canada
| | | | | | | | | | | | | |
Collapse
|
134
|
Armand AS, Launay T, Gaspera BD, Charbonnier F, Gallien CL, Chanoine C. Effects of eccentric treadmill running on mouse soleus: degeneration/regeneration studied with Myf-5 and MyoD probes. ACTA PHYSIOLOGICA SCANDINAVICA 2003; 179:75-84. [PMID: 12940941 DOI: 10.1046/j.1365-201x.2003.01187.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
AIM The aim of this report is to show that eccentric exercise under well-controlled conditions is an alternative model, to chemical and mechanical analyses, and analyse the process of degeneration/regeneration in mouse soleus. METHODS For this, mice were submitted to a single bout of eccentric exercise on a treadmill down a 14 degrees decline for 150 min and the soleus muscle was analysed at different times following exercise by histology and in situ hybridization in comparison with cardiotoxin-injured muscles. RESULTS We analyse the regenerative process by detection of the accumulation of transcripts coding for the two myogenic regulatory factors, Myf-5 and MyoD, which are good markers of the activated satellite cells. From 24 h post-exercise (P-E), clusters of mononucleated Myf-5/MyoD-positive cells were detected. Their number increased up to 96 h P-E when young MyoD-positive myotubes with central nuclei began to appear. From 96 to 168 h P-E the number of myotubes increased, about 10-fold, the new myotubes representing 58% of the muscle cells (168 h P-E). CONCLUSION These results show that this protocol of eccentric exercise is able to induce a drastic degeneration/regeneration process in the soleus muscle. This offers the opportunity to perform biochemical and molecular analyses of a process of regeneration without muscle environment defects. The advantages of this model are discussed in the context of fundamental and therapeutical perspectives.
Collapse
Affiliation(s)
- A-S Armand
- Biologie du Développement et de la Différenciation Neuromusculaire, LNRS UMR 7060 CNRS, Université René Descartes, Paris Cedex, France
| | | | | | | | | | | |
Collapse
|
135
|
Anderson JE, Vargas C. Correlated NOS-Imu and myf5 expression by satellite cells in mdx mouse muscle regeneration during NOS manipulation and deflazacort treatment. Neuromuscul Disord 2003; 13:388-96. [PMID: 12798794 DOI: 10.1016/s0960-8966(03)00029-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Satellite cells, muscle precursor cells in skeletal muscle, are normally quiescent and become activated by disease or injury. A lack of dystrophin and changes in the expression or activity of neuronal nitric oxide synthase (NOS-I) affect the timing of activation in vivo. Nitric oxide synthase inhibition delays muscle repair in normal mice, and worsens muscular dystrophy in the mdx mouse, a genetic homologue of Duchenne muscular dystrophy. However, the potential role of activation and repair events mediated by nitric oxide in determining the outcome of steroid or other treatments for muscular dystrophy is not clear. We tested the hypothesis that the extent of repair in dystrophic muscles of mdx mice is partly dependent on NOS-Imu expression and activity. Myotube formation in regenerating muscle was promoted by deflazacort treatment of mdx dystrophic mice (P<0.05), and improved by combination with the nitric oxide synthase substrate, L-arginine, especially in the diaphragm. NOS-Imu mRNA expression and activity were present in satellite cells and very new myotubes of regenerating and dystrophic muscle. Deflazacort treatment resulted in increased NOS-Imu expression in regenerating muscles in a strong and specific correlation with myf5 expression (r=0.95, P<0.01), a marker for muscle repair. Nitric oxide synthase inhibition prevented the deflazacort-induced rise in NOS-Imu and myf5 expression in the diaphragm without affecting the diameter of non-regenerating fibres. These in vivo studies suggest that gains in NOS-Imu expression and nitric oxide synthase activity in satellite cells can increase the extent and speed of repair, even in the absence of dystrophin in muscle fibres. NOS-Imu may be a useful therapeutic target to augment the effects of steroidal or other treatments of muscular dystrophy.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents/pharmacology
- Anti-Inflammatory Agents/therapeutic use
- Arginine/pharmacology
- Arginine/therapeutic use
- Blotting, Northern
- DNA-Binding Proteins
- Diaphragm/metabolism
- Dystrophin/deficiency
- Immunosuppressive Agents/pharmacology
- Immunosuppressive Agents/therapeutic use
- In Situ Hybridization
- Mice
- Mice, Inbred mdx
- Muscle Fibers, Skeletal/metabolism
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/enzymology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Dystrophies/metabolism
- Myogenic Regulatory Factor 5
- Nitric Oxide Synthase/antagonists & inhibitors
- Nitric Oxide Synthase/metabolism
- Pregnenediones/pharmacology
- Pregnenediones/therapeutic use
- Protein Isoforms
- Proto-Oncogene Proteins c-fos/metabolism
- Proto-Oncogene Proteins c-met/metabolism
- RNA, Messenger/metabolism
- Regeneration/drug effects
- Satellite Cells, Skeletal Muscle/drug effects
- Satellite Cells, Skeletal Muscle/enzymology
- Satellite Cells, Skeletal Muscle/metabolism
- Trans-Activators/metabolism
Collapse
Affiliation(s)
- Judy E Anderson
- Department of Human Anatomy and Cell Science, Faculty of Medicine, University of Manitoba, 730 William Avenue, Winnipeg, MB, Canada R3E 0W3.
| | | |
Collapse
|
136
|
Armand AS, Della Gaspera B, Launay T, Charbonnier F, Gallien CL, Chanoine C. Expression and neural control of follistatin versus myostatin genes during regeneration of mouse soleus. Dev Dyn 2003; 227:256-65. [PMID: 12761853 DOI: 10.1002/dvdy.10306] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Follistatin and myostatin are two secreted proteins involved in the control of muscle mass during development. These two proteins have opposite effects on muscle growth, as documented by genetic models. The aims of this work were to analyze in mouse, by using in situ hybridization, the spatial and temporal expression patterns of follistatin and myostatin mRNAs during soleus regeneration after cardiotoxin injury, and to investigate the influence of innervation on the accumulation of these two transcripts. Follistatin transcripts could be detected in activated satellite cells as early as the first stages of regeneration and were transiently expressed in forming myotubes. In contrast, myostatin mRNAs accumulated persistently throughout the regeneration process as well as in adult control soleus. Denervation significantly affected both follistatin and myostatin transcript accumulation, but in opposite ways. Muscle denervation persistently reduced the levels of myostatin transcripts as early as the young myotube stage, whereas the levels of follistatin mRNA were strongly increased in the small myotubes in the late stages of regeneration. These results are discussed with regard to the potential functions of both follistatin, as a positive regulator of muscle differentiation, and myostatin, as a negative regulator of skeletal muscle growth. We suggest that the belated up-regulation of the follistatin mRNA level in the small myotubes of the regenerating soleus as well as the down-regulation of the myostatin transcript level after denervation contribute to the differentiation process in denervated regenerating muscle.
Collapse
Affiliation(s)
- Anne-Sophie Armand
- Biologie du Développement et de la Différenciation Neuromusculaire, LNRS ESA 7060 CNRS, Université René Descartes, Paris, France
| | | | | | | | | | | |
Collapse
|
137
|
McLoon LK, Wirtschafter J. Activated satellite cells in extraocular muscles of normal adult monkeys and humans. Invest Ophthalmol Vis Sci 2003; 44:1927-32. [PMID: 12714625 PMCID: PMC1796845 DOI: 10.1167/iovs.02-0673] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
PURPOSE Mammalian extraocular muscles (EOMs) are both physiologically and biochemically unique when compared with nonocular skeletal muscles. Recent studies have demonstrated a process of continuous myonuclear addition in normal uninjured myofibers in adult EOMs of rabbits and mice. The current study was conducted to determine whether this process of myonuclear addition is a universal phenomenon in mammalian EOMs. METHODS The EOMs from adult uninjured monkeys and humans were examined immunohistochemically for the expression of specific markers of activated satellite cells: hepatocyte growth factor (HGF); the myogenic regulatory factors MyoD, myogenin, and Pax7; and a marker for nuclei in all proliferative phases of the cell cycle, Ki-67. The satellite cell identity of the cells positive for Ki-67, HGF, and Pax7 was determined by colabeling sets of serial sections with either laminin or dystrophin. RESULTS In cross sections of monkey and human EOMs, approximately 7% to 8% of the myofiber profiles were associated with Pax7-positive satellite cells and between 2% and 4% were associated with MyoD-positive satellite cells or HGF-positive satellite cells. Similar percentages of satellite cells were positive for myogenin in the orbital layer, but the global layer had few satellite cells that were myogenin positive. An average of 0.72% of the myofibers had Ki-67-positive cells associated with them in the satellite cell position. CONCLUSIONS Activated satellite cells were present on myofibers in normal uninjured adult monkey and human EOMs, as visualized with these five distinct markers. The data support the hypothesis that the process of continuous myonuclear addition is most likely active in primate and human EOMs. The presence of continuous myofiber remodeling in EOM suggests new mechanisms that may be responsible for EOM sparing or involvement in skeletal muscle diseases.
Collapse
Affiliation(s)
- Linda K McLoon
- Department of Ophthalmology, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| | | |
Collapse
|
138
|
Dedkov EI, Kostrominova TY, Borisov AB, Carlson BM. MyoD and myogenin protein expression in skeletal muscles of senile rats. Cell Tissue Res 2003; 311:401-16. [PMID: 12658448 DOI: 10.1007/s00441-002-0686-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2002] [Accepted: 11/26/2002] [Indexed: 01/26/2023]
Abstract
We analyzed the level of protein expression of two myogenic regulatory factors (MRFs), MyoD and myogenin, in senile skeletal muscles and determined the cellular source of their production in young adult (4 months old), old (24, 26, and 28 months old), and senile (32 months old) male rats. Immunoblotting demonstrated levels of myogenin approximately 3.2, approximately 4.0, and approximately 5.5 times higher in gastrocnemius muscles of 24-, 26-, and 32-month-old animals, respectively, than in those of young adult rats. Anti-MyoD antibody recognized two major areas of immunoreactivity in Western blots: a single MyoD-specific band (approximately 43-45 kDa) and a double (or triple) MyoD-like band (approximately 55-65 kDa). Whereas the level of MyoD-specific protein in the 43- to 45-kDa band remained relatively unchanged during aging compared with that of young adult rats, the total level of MyoD-like immunoreactivity within the 55- to 65-kDa bands was approximately 3.4, approximately 4.7, approximately 9.1, and approximately 11.7 times higher in muscles of 24-, 26-, 28-, and 32-month-old rats, respectively. The pattern of MRF protein expression in intact senile muscles was similar to that recorded in young adult denervated muscles. Ultrastructural analysis of extensor digitorum longus muscle from senile rats showed that, occasionally, the area of the nerve-muscle junction was partially or completely devoid of axons, and satellite cells with the features of activated cells were found on the surface of living fibers. Immunohistochemistry detected accumulated MyoD and myogenin proteins in the nuclei of both fibers and satellite cells in 32-month-old muscles. We suggest that the up-regulated production of MyoD and myogenin proteins in the nuclei of both fibers and satellite cells could account for the high level of MRF expression in muscles of senile rats.
Collapse
MESH Headings
- Aging/metabolism
- Animals
- Immunohistochemistry
- Male
- Microscopy, Electron
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle Fibers, Skeletal/ultrastructure
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscle, Skeletal/ultrastructure
- MyoD Protein/biosynthesis
- MyoD Protein/metabolism
- Myogenin/biosynthesis
- Myogenin/metabolism
- Neuromuscular Junction/metabolism
- Neuromuscular Junction/pathology
- Neuromuscular Junction/ultrastructure
- Rats
- Rats, Wistar
- Regeneration/physiology
- Satellite Cells, Skeletal Muscle/metabolism
- Satellite Cells, Skeletal Muscle/pathology
- Satellite Cells, Skeletal Muscle/ultrastructure
- Up-Regulation/physiology
Collapse
Affiliation(s)
- Eduard I Dedkov
- Department of Cell and Developmental Biology, 4643 Medical Sciences II Building, University of Michigan, Ann Arbor, Michigan 48109, USA.
| | | | | | | |
Collapse
|
139
|
Jiang X, Edstrom E, Altun M, Ulfhake B. Differential regulation of Shc adaptor proteins in skeletal muscle, spinal cord and forebrain of aged rats with sensorimotor impairment. Aging Cell 2003; 2:47-57. [PMID: 12882334 DOI: 10.1046/j.1474-9728.2003.00030.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The Shc family of proteins participates in mitogenic and survival signalling through binding to receptor tyrosine kinases. We report here on the expression of Shc in forebrain, spinal cord and hind limb muscles from 30-month-old rats with different degrees of sensorimotor impairment. ShcA (mRNA and protein) is up-regulated in skeletal muscles and spinal cord of aged rats, and this change relates to biological age, i.e. degree of behavioural incapacitation, rather than to chronological age. Western blot and RT-PCR revealed that the increase in ShcA selectively affected the p46 isoform in the spinal cord, whereas in muscle tissue a robust increase of p66(ShcA) was also evident. Furthermore, in parallel with the up-regulation of ShcA, an increase of p75(NTR) mRNA in the aged animals was observed. ShcB mRNA showed a tendency for down-regulation in both spinal cord and skeletal muscles, whereas the expression of ShcC was unaltered. Our data show that the regulation of Shc mRNAs in senescence is region as well as isoform specific. The regulatory changes may reflect changes in mitogenic/survival signalling induced by age-related cell and tissue damage. The coup-regulation of p66(ShcA) and p75(NTR) is interesting since both molecules have been associated with apoptosis.
Collapse
Affiliation(s)
- Xiaogang Jiang
- Experimental Neurogerontology, Department of Neuroscience, Retzius Laboratory, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | | | | |
Collapse
|
140
|
Patel K, Christ B, Stockdale FE. Control of muscle size during embryonic, fetal, and adult life. Results Probl Cell Differ 2003; 38:163-86. [PMID: 12132394 DOI: 10.1007/978-3-540-45686-5_8] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Affiliation(s)
- Ketan Patel
- Department of Veterinary Basic Sciences, Royal Veterinary College, Royal College Street, London NW1 OTU, UK
| | | | | |
Collapse
|
141
|
Kami K, Senba E. In vivo activation of STAT3 signaling in satellite cells and myofibers in regenerating rat skeletal muscles. J Histochem Cytochem 2002; 50:1579-89. [PMID: 12486080 DOI: 10.1177/002215540205001202] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Although growth factors and cytokines play critical roles in skeletal muscle regeneration, intracellular signaling molecules that are activated by these factors in regenerating muscles have been not elucidated. Several lines of evidence suggest that leukemia inhibitory factor (LIF) is an important cytokine for the proliferation and survival of myoblasts in vitro and acceleration of skeletal muscle regeneration. To elucidate the role of LIF signaling in regenerative responses of skeletal muscles, we examined the spatial and temporal activation patterns of an LIF-associated signaling molecule, the signal transducer and activator transcription 3 (STAT3) proteins in regenerating rat skeletal muscles induced by crush injury. At the early stage of regeneration, activated STAT3 proteins were first detected in the nuclei of activated satellite cells and then continued to be activated in proliferating myoblasts expressing both PCNA and MyoD proteins. When muscle regeneration progressed, STAT3 signaling was no longer activated in differentiated myoblasts and myotubes. In addition, activation of STAT3 was also detected in myonuclei within intact sarcolemmas of surviving myofibers that did not show signs of necrosis. These findings suggest that activation of STAT3 signaling is an important molecular event that induces the successful regeneration of injured skeletal muscles.
Collapse
Affiliation(s)
- Katsuya Kami
- Department of Health Science, Osaka University of Health and Sport Sciences, Wakayama, Japan.
| | | |
Collapse
|
142
|
Zammit PS, Heslop L, Hudon V, Rosenblatt JD, Tajbakhsh S, Buckingham ME, Beauchamp JR, Partridge TA. Kinetics of myoblast proliferation show that resident satellite cells are competent to fully regenerate skeletal muscle fibers. Exp Cell Res 2002; 281:39-49. [PMID: 12441128 DOI: 10.1006/excr.2002.5653] [Citation(s) in RCA: 203] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The satellite cell compartment provides skeletal muscle with a remarkable capacity for regeneration. Here, we have used isolated myofibers to investigate the activation and proliferative potential of satellite cells. We have previously shown that satellite cells are heterogeneous: the majority express Myf5 and M-cadherin protein, presumably reflecting commitment to myogenesis, while a minority is negative for both. Although MyoD is rarely detected in quiescent satellite cells, over 98% of satellite cells contain MyoD within 24 h of stimulation. Significantly, MyoD is only observed in cells that are already expressing Myf5. In contrast, a minority population does not activate by the criteria of Myf5 or MyoD expression. Following the synchronous activation of the myogenic regulatory factor+ve satellite cells, their daughter myoblasts proliferate with a doubling time of approximately 17 h, irrespective of the fiber type (type I, IIa, or IIb) from which they originate. Although fast myofibers have fewer associated satellite cells than slow, and accordingly produce fewer myoblasts, each myofiber phenotype is associated with a complement of satellite cells that has sufficient proliferative potential to fully regenerate the parent myofiber within 4 days. This time course is similar to that observed in vivo following acute injury and indicates that cells other than satellite cells are not required for complete myofiber regeneration.
Collapse
Affiliation(s)
- Peter S Zammit
- Muscle Cell Biology Group, Medical Research Council Clinical Sciences Centre, Faculty of Medicine, Imperial College, Hammersmith Hospital Campus, Du Cane Road, London, United Kingdom.
| | | | | | | | | | | | | | | |
Collapse
|
143
|
Asakura A, Seale P, Girgis-Gabardo A, Rudnicki MA. Myogenic specification of side population cells in skeletal muscle. J Cell Biol 2002; 159:123-34. [PMID: 12379804 PMCID: PMC2173497 DOI: 10.1083/jcb.200202092] [Citation(s) in RCA: 483] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Skeletal muscle contains myogenic progenitors called satellite cells and muscle-derived stem cells that have been suggested to be pluripotent. We further investigated the differentiation potential of muscle-derived stem cells and satellite cells to elucidate relationships between these two populations of cells. FACS(R) analysis of muscle side population (SP) cells, a fraction of muscle-derived stem cells, revealed expression of hematopoietic stem cell marker Sca-1 but did not reveal expression of any satellite cell markers. Muscle SP cells were greatly enriched for cells competent to form hematopoietic colonies. Moreover, muscle SP cells with hematopoietic potential were CD45 positive. However, muscle SP cells did not differentiate into myocytes in vitro. By contrast, satellite cells gave rise to myocytes but did not express Sca-1 or CD45 and never formed hematopoietic colonies. Importantly, muscle SP cells exhibited the potential to give rise to both myocytes and satellite cells after intramuscular transplantation. In addition, muscle SP cells underwent myogenic specification after co-culture with myoblasts. Co-culture with myoblasts or forced expression of MyoD also induced muscle differentiation of muscle SP cells prepared from mice lacking Pax7 gene, an essential gene for satellite cell development. Therefore, these data document that satellite cells and muscle-derived stem cells represent distinct populations and demonstrate that muscle-derived stem cells have the potential to give rise to myogenic cells via a myocyte-mediated inductive interaction.
Collapse
MESH Headings
- Animals
- Antigens, Ly/metabolism
- Cell Differentiation/physiology
- Cell Separation
- Cell Transplantation
- Cells, Cultured
- Coculture Techniques
- DNA-Binding Proteins
- Flow Cytometry
- Genes, Reporter
- Hematopoiesis
- Hematopoietic Stem Cells/cytology
- Hematopoietic Stem Cells/physiology
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Leukocyte Common Antigens/metabolism
- Membrane Proteins/metabolism
- Mice
- Mice, SCID
- Mice, Transgenic
- Muscle Development
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Muscle, Skeletal/cytology
- Muscle, Skeletal/physiology
- MyoD Protein
- Myoblasts, Skeletal/cytology
- Myoblasts, Skeletal/physiology
- Myogenic Regulatory Factor 5
- PAX7 Transcription Factor
- Recombinant Fusion Proteins/metabolism
- Satellite Cells, Skeletal Muscle/cytology
- Satellite Cells, Skeletal Muscle/physiology
- Trans-Activators
- Transcription Factors/genetics
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- Atsushi Asakura
- Molecular Medicine Program, Ottawa Health Research Institute, Ottawa, Ontario, K1H 8L6 Canada
| | | | | | | |
Collapse
|
144
|
Abstract
The myogenic transcription factors of the MyoD family are not expressed in normal adult skeletal muscle. They are upregulated at the transcript and protein levels in a precisely coordinated manner during regeneration. While the cellular distribution of MyoD, myf-5, and myogenin expression in regenerating muscle is well documented, little is known about the exact localization of MRF4. It was the aim of this study to monitor the cellular distribution of MRF4 protein during regeneration. The soleus muscle of 6-week-old male Wistar rats was devascularized and allowed to regenerate for 2, 5, 10 or 14 days. Immunostaining revealed the presence of MRF4 throughout the time periods studied. Expression was detected in the nuclei of myofibers which had survived the devascularization procedure 2 days after necrosis was induced. In nuclei of newly formed myotubes and young myofibers, MRF4 was co-expressed with MyoD and myogenin. MRF4 protein was absent from satellite cells (SC), with anti-M-cadherin being used as a SC marker. Taken together, our results demonstrate that MRF4 protein expression in regenerated fibers is restricted to the time around and after fusion. The absence of MRF4 protein in SC suggests that the role of MRF4 during regeneration is distinct from myf-5, MyoD, and myogenin.
Collapse
Affiliation(s)
- Z Zhou
- Institute of Brain Research, University of Tübingen, Germany
| | | |
Collapse
|
145
|
Sachidanandan C, Sambasivan R, Dhawan J. Tristetraprolin and LPS-inducible CXC chemokine are rapidly induced in presumptive satellite cells in response to skeletal muscle injury. J Cell Sci 2002; 115:2701-12. [PMID: 12077361 DOI: 10.1242/jcs.115.13.2701] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Myogenic precursor cells known as satellite cells persist in adult skeletal muscle and are responsible for its ability to regenerate after injury. Quiescent satellite cells are activated by signals emanating from damaged muscle. Here we describe the rapid activation of two genes in response to muscle injury; these transcripts encode LPS-inducible CXC chemokine (LIX), a neutrophil chemoattractant, and Tristetraprolin (TTP), an RNA-binding protein implicated in the regulation of cytokine expression. Using a synchronized cell culture model we show that C2C12 myoblasts arrested in G0 exhibit some molecular attributes of satellite cells in vivo: suppression of MyoD and Myf5 expression during G0 and their reactivation in G1. Synchronization also revealed cell cycle dependent expression of CD34, M-cadherin, HGF and PEA3, genes implicated in satellite cell biology. To identify other genes induced in synchronized C2C12 myoblasts we used differential display PCR and isolated LIX and TTP cDNAs. Both LIX and TTP mRNAs are short-lived, encode molecules implicated in inflammation and are transiently induced during growth activation in vitro. Further, LIX and TTP are rapidly induced in response to muscle damage in vivo. TTP expression precedes that of MyoD and is detected 30 minutes after injury. The spatial distribution of LIX and TTP transcripts in injured muscle suggests expression by satellite cells. Our studies suggest that in addition to generating new cells for repair, activated satellite cells may be a source of signaling molecules involved in tissue remodeling during regeneration.
Collapse
MESH Headings
- Animals
- Cadherins/genetics
- Cadherins/metabolism
- Cell Differentiation/genetics
- Cells, Cultured
- Chemokine CXCL5
- Chemokines, CXC/genetics
- Chemokines, CXC/metabolism
- Creatine Kinase/genetics
- Creatine Kinase/metabolism
- DNA-Binding Proteins
- Down-Regulation/genetics
- Fetus
- Gene Expression Regulation, Developmental/genetics
- Hepatocyte Growth Factor/genetics
- Hepatocyte Growth Factor/metabolism
- Immediate-Early Proteins/genetics
- Immediate-Early Proteins/metabolism
- Mice
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Muscle, Skeletal/growth & development
- Muscle, Skeletal/injuries
- Muscle, Skeletal/metabolism
- MyoD Protein/genetics
- MyoD Protein/metabolism
- Myoblasts, Skeletal/metabolism
- Myogenic Regulatory Factor 5
- Proto-Oncogene Proteins c-met/genetics
- Proto-Oncogene Proteins c-met/metabolism
- RNA, Messenger/metabolism
- Reaction Time/genetics
- Regeneration/genetics
- Resting Phase, Cell Cycle/genetics
- Satellite Cells, Skeletal Muscle/cytology
- Satellite Cells, Skeletal Muscle/metabolism
- Trans-Activators
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Tristetraprolin
Collapse
|
146
|
McLoon LK, Wirtschafter JD. Continuous myonuclear addition to single extraocular myofibers in uninjured adult rabbits. Muscle Nerve 2002; 25:348-58. [PMID: 11870711 DOI: 10.1002/mus.10056] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Extraocular muscles (EOM) are unique among mammalian skeletal muscles in that they normally express molecules associated with muscle development and regeneration. In this study we show that satellite cells of EOM, unlike those of other skeletal muscles, continually divide in the normal, uninjured adult. Adult EOM contained activated satellite cells positive for the myogenic regulatory factor MyoD. EOM satellite cells did not require a prolonged activation period prior to onset of cell division and differentiation in vitro. EOM satellite cells incorporated bromodeoxyuridine (brdU), a marker for cell division, and with longer postlabeling survival, brdU-labeled nuclei populated EOM myofibers. This was not seen with leg muscle. These findings suggest the possibility that continual division of satellite cells and fusion of their daughter myocytes with existing adult EOM myofibers contribute to the unique sparing or susceptibility of EOM to certain muscle diseases.
Collapse
Affiliation(s)
- Linda K McLoon
- Department of Ophthalmology, University of Minnesota, Lions Research Building, Room 374, 2001 Sixth Street SE, Minneapolis, Minnesota 55455, USA.
| | | |
Collapse
|
147
|
Mizuno H, Zuk PA, Zhu M, Lorenz HP, Benhaim P, Hedrick MH. Myogenic differentiation by human processed lipoaspirate cells. Plast Reconstr Surg 2002; 109:199-209; discussion 210-1. [PMID: 11786812 DOI: 10.1097/00006534-200201000-00030] [Citation(s) in RCA: 306] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The use of undifferentiated cells for cell-based tissue engineering and regeneration strategies represents a promising approach for skeletal muscle repair. For such strategies to succeed, a readily available source of myogenic precursor cells must be identified. We have previously shown that cells isolated from raw human lipoaspirates, called processed lipoaspirate cells, display multilineage mesodermal potential in vitro. Because human liposuctioned fat is available in large quantities and can be harvested with low morbidity, it may be an ideal source of stem cells for tissue-engineering applications. In this study, processed lipoaspirate cells were isolated from raw lipoaspirates harvested from eight patients who underwent cosmetic surgery. Processed lipoaspirate cells were placed in promyogenic conditions for up to 6 weeks, and the expression of the myogenic markers MyoD1 and myosin heavy chain was confirmed by using structure, histology, and reverse transcriptase-polymerase chain reaction. Histologic results were quantitated as an indicator or myogenic differentiation levels. We found that induced human processed lipoaspirate cells form multinucleated cells after 3 weeks of induction, indicative of the formation of myotubes. In addition, MyoD1 and skeletal muscle myosin heavy chain are expressed at distinct time points during differentiation with MyoD1 expression preceding expression of myosin. Finally, approximately 15 percent of human processed lipoaspirate cells can be induced toward myogenic differentiation 6 weeks after induction. In summary, our findings suggest that human processed lipoaspirate cells differentiate into myogenic cells. Furthermore, these cells may be a useful source for skeletal muscle engineering and repair.
Collapse
Affiliation(s)
- Hiroshi Mizuno
- Laboratory for Regenerative Bioengineering and Repair, Department of Surgery, University of California-Los Angeles School of Medicine, 90095-1665, USA
| | | | | | | | | | | |
Collapse
|
148
|
Kicic A, Shen W, Rakoczy PE. The potential of marrow stromal cells in stem cell therapy. Eye (Lond) 2001; 15:695-707. [PMID: 11826986 DOI: 10.1038/eye.2001.233] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- A Kicic
- Stem Cell Unit, Lions Eye Institute, Nedlands, Western Australia, Australia
| | | | | |
Collapse
|
149
|
Asakura A, Komaki M, Rudnicki M. Muscle satellite cells are multipotential stem cells that exhibit myogenic, osteogenic, and adipogenic differentiation. Differentiation 2001; 68:245-53. [PMID: 11776477 DOI: 10.1046/j.1432-0436.2001.680412.x] [Citation(s) in RCA: 567] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Muscle satellite cells are believed to represent a committed stem cell population that is responsible for the postnatal growth and regeneration of skeletal muscle. However, the observation that cultured myoblasts differentiate into osteocytes or adipocytes following treatment with bone morphogenetic proteins (BMPs) or adipogenic inducers, respectively, suggests some degree of plasticity within the mesenchymal lineage. To further investigate this phenomenon, we explore the osteogenic and adipogenic potential of satellite cells isolated from adult mice. Our experiments clearly demonstrate that satellite cell-derived primary myoblasts, expressing myogenic markers such as MyoD, Myf5, Pax7 and desmin, differentiated only into osteocytes or adipocytes following treatment with BMPs or adipogenic inducers, respectively However, satellite cells on isolated muscle fibers cultured in Matrigel readily differentiated into myocytes as well as osteogenic and adipogenic lineages, whereas primary myoblasts did not. Satellite cell-derived primary myoblasts isolated from mice lacking the myogenic transcription factor MyoD (MyoD-/-) differentiate into myocytes poorly in vivo and in vitro (Megeney et al., Genes Dev. 1996; Sabourin et. al, J. Cell Biol., 1999). Therefore, we tested whether MyoD-/- primary myoblasts display increased plasticity relative to wild type cells. Unexpectedly, the osteogenic or adipogenic differentiation potential of MyoD-/- primary myoblasts did not increase compared to wild-type cells. Taken together, these results strongly suggest that muscle satellite cells possess multipotential mesenchymal stem cell activity and are capable of forming osteocytes and adipocytes as well as myocytes.
Collapse
Affiliation(s)
- A Asakura
- Ottawa Health Research Institute, Ontario, Canada
| | | | | |
Collapse
|
150
|
Abstract
Adult skeletal muscle has a remarkable ability to regenerate following myotrauma. Because adult myofibers are terminally differentiated, the regeneration of skeletal muscle is largely dependent on a small population of resident cells termed satellite cells. Although this population of cells was identified 40 years ago, little is known regarding the molecular phenotype or regulation of the satellite cell. The use of cell culture techniques and transgenic animal models has improved our understanding of this unique cell population; however, the capacity and potential of these cells remain ill-defined. This review will highlight the origin and unique markers of the satellite cell population, the regulation by growth factors, and the response to physiological and pathological stimuli. We conclude by highlighting the potential therapeutic uses of satellite cells and identifying future research goals for the study of satellite cell biology.
Collapse
Affiliation(s)
- T J Hawke
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | |
Collapse
|