301
|
Willis-Owen SAG, Thompson A, Kemp PR, Polkey MI, Cookson WOCM, Moffatt MF, Natanek SA. COPD is accompanied by co-ordinated transcriptional perturbation in the quadriceps affecting the mitochondria and extracellular matrix. Sci Rep 2018; 8:12165. [PMID: 30111857 PMCID: PMC6093887 DOI: 10.1038/s41598-018-29789-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 07/16/2018] [Indexed: 12/04/2022] Open
Abstract
Skeletal muscle dysfunction is a frequent extra-pulmonary manifestation of Chronic Obstructive Pulmonary Disease (COPD) with implications for both quality of life and survival. The underlying biology nevertheless remains poorly understood. We measured global gene transcription in the quadriceps using Affymetrix HuGene1.1ST arrays in an unselected cohort of 79 stable COPD patients in secondary care and 16 healthy age- and gender-matched controls. We detected 1,826 transcripts showing COPD-related variation. Eighteen exhibited ≥2fold changes (SLC22A3, FAM184B, CDKN1A, FST, LINC01405, MUSK, PANX1, ANKRD1, C12orf75, MYH1, POSTN, FRZB, TNC, ACTC1, LINC00310, MYH3, MYBPH and AREG). Thirty-one transcripts possessed previous reported evidence of involvement in COPD through genome-wide association, including FAM13A. Network analysis revealed a substructure comprising 6 modules of co-expressed genes. We identified modules with mitochondrial and extracellular matrix features, of which IDH2, a central component of the mitochondrial antioxidant pathway, and ABI3BP, a proposed switch between proliferation and differentiation, represent hubs respectively. COPD is accompanied by coordinated patterns of transcription in the quadriceps involving the mitochondria and extracellular matrix and including genes previously implicated in primary disease processes.
Collapse
Affiliation(s)
- Saffron A G Willis-Owen
- Centre for Genomic Medicine, National Heart and Lung Institute, Imperial College London, SW3 6LY, London, United Kingdom.
| | - Anna Thompson
- Centre for Genomic Medicine, National Heart and Lung Institute, Imperial College London, SW3 6LY, London, United Kingdom
| | - Paul R Kemp
- Respiratory Sciences, National Heart and Lung Institute, Imperial College London, SW3 6NP, London, United Kingdom
| | - Michael I Polkey
- Respiratory Sciences, National Heart and Lung Institute, Imperial College London, SW3 6NP, London, United Kingdom
| | - William O C M Cookson
- Centre for Genomic Medicine, National Heart and Lung Institute, Imperial College London, SW3 6LY, London, United Kingdom
| | - Miriam F Moffatt
- Centre for Genomic Medicine, National Heart and Lung Institute, Imperial College London, SW3 6LY, London, United Kingdom
| | - Samantha A Natanek
- Respiratory Sciences, National Heart and Lung Institute, Imperial College London, SW3 6NP, London, United Kingdom.
| |
Collapse
|
302
|
Liu S, Chen H, Ronquist S, Seaman L, Ceglia N, Meixner W, Chen PY, Higgins G, Baldi P, Smale S, Hero A, Muir LA, Rajapakse I. Genome Architecture Mediates Transcriptional Control of Human Myogenic Reprogramming. iScience 2018; 6:232-246. [PMID: 30240614 PMCID: PMC6137960 DOI: 10.1016/j.isci.2018.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 06/23/2018] [Accepted: 07/31/2018] [Indexed: 12/21/2022] Open
Abstract
Genome architecture has emerged as a critical element of transcriptional regulation, although its role in the control of cell identity is not well understood. Here we use transcription factor (TF)-mediated reprogramming to examine the interplay between genome architecture and transcriptional programs that transition cells into the myogenic identity. We recently developed new methods for evaluating the topological features of genome architecture based on network centrality. Through integrated analysis of these features of genome architecture and transcriptome dynamics during myogenic reprogramming of human fibroblasts we find that significant architectural reorganization precedes activation of a myogenic transcriptional program. This interplay sets the stage for a critical transition observed at several genomic scales reflecting definitive adoption of the myogenic phenotype. Subsequently, TFs within the myogenic transcriptional program participate in entrainment of biological rhythms. These findings reveal a role for topological features of genome architecture in the initiation of transcriptional programs during TF-mediated human cellular reprogramming. 4D Nucleome analysis of direct human fibroblast to muscle reprogramming A space-time bifurcation marks transit to a new cell identity Chromatin reorganization precedes significant transcriptional changes Myogenic master regulators have a role in entraining biological rhythms
Collapse
Affiliation(s)
- Sijia Liu
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI 48109, USA
| | - Haiming Chen
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Scott Ronquist
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Laura Seaman
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nicholas Ceglia
- Department of Computer Science, University of California-Irvine, Irvine, CA 92697, USA
| | - Walter Meixner
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Pin-Yu Chen
- AI Foundations, IBM T. J. Watson Research Center, Yorktown Heights, NY 10598, USA
| | - Gerald Higgins
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Pierre Baldi
- Department of Computer Science, University of California-Irvine, Irvine, CA 92697, USA
| | - Steve Smale
- Department of Mathematics, City University of Hong Kong, Hong Kong 999077, China; Department of Mathematics, University of California, Berkeley, CA 94720, USA
| | - Alfred Hero
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lindsey A Muir
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Indika Rajapakse
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Mathematics, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
303
|
Lin IH, Chang JL, Hua K, Huang WC, Hsu MT, Chen YF. Skeletal muscle in aged mice reveals extensive transformation of muscle gene expression. BMC Genet 2018; 19:55. [PMID: 30089464 PMCID: PMC6083496 DOI: 10.1186/s12863-018-0660-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/26/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Aging leads to decreased skeletal muscle function in mammals and is associated with a progressive loss of muscle mass, quality and strength. Age-related muscle loss (sarcopenia) is an important health problem associated with the aged population. RESULTS We investigated the alteration of genome-wide transcription in mouse skeletal muscle tissue (rectus femoris muscle) during aging using a high-throughput sequencing technique. Analysis revealed significant transcriptional changes between skeletal muscles of mice at 3 (young group) and 24 (old group) months of age. Specifically, genes associated with energy metabolism, cell proliferation, muscle myosin isoforms, as well as immune functions were found to be altered. We observed several interesting gene expression changes in the elderly, many of which have not been reported before. CONCLUSIONS Those data expand our understanding of the various compensatory mechanisms that can occur with age, and further will assist in the development of methods to prevent and attenuate adverse outcomes of aging.
Collapse
Affiliation(s)
- I-Hsuan Lin
- VYM Genome Research Center, National Yang-Ming University, Taipei, 112, Taiwan
| | - Junn-Liang Chang
- Department of Pathology & Laboratory Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, 325, Taiwan
| | - Kate Hua
- VYM Genome Research Center, National Yang-Ming University, Taipei, 112, Taiwan
| | - Wan-Chen Huang
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, No.250, Wu-Hsing Street, Taipei, 11031, Taiwan
| | - Ming-Ta Hsu
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, 112, Taiwan
| | - Yi-Fan Chen
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, No.250, Wu-Hsing Street, Taipei, 11031, Taiwan.
| |
Collapse
|
304
|
Cheng X, Huang H, Luo X, Shi B, Li J. Wnt7a induces satellite cell expansion, myofiber hyperplasia and hypertrophy in rat craniofacial muscle. Sci Rep 2018; 8:10613. [PMID: 30006540 PMCID: PMC6045621 DOI: 10.1038/s41598-018-28917-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/25/2018] [Indexed: 02/05/2023] Open
Abstract
Craniofacial muscles drive critical functions in the head, including speech, feeding and expression. Compared with their counterparts in trunk and limbs, craniofacial muscles are of distinct embryonic origins, which might consequently lead to different growth patterns and regenerative potential. In this study, rat levator veli palatini muscle and masseter muscle were compared with tibialis anterior muscle in their response to exogenous Wnt7a stimulus, which has been proved effective in promoting muscle regeneration in the limbs. Histological, cellular and molecular analyses were performed both under basal condition and after a single dose injection of recombinant human Wnt7a. Under basal condition, levator veli palatini muscle demonstrated considerably more satellite cells than the others. After Wnt7a administration, regeneration-related activities, including satellite cell expansion, myofiber hyperplasia and hypertrophy were generally observed in all three muscles, but with obvious differences in the extent. The composition of fast/slow myofibers underwent substantial alterations, and the pattern varied among the three muscles. Location-specific alterations in the expression level of core components in planar cell polarity pathway, Akt/mTOR pathway and myostatin pathway were also observed. In conclusion, both craniofacial and limb muscles could be effectively expanded by exogenous Wnt7a stimulus, but muscle-to-muscle variations in response patterns existed.
Collapse
Affiliation(s)
- Xu Cheng
- State Key Laboratory of Oral Diseases & National Clinical Research Centre for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, 14 Ren Min Nan Road, Chengdu, 610041, P. R. China
| | - Hanyao Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Centre for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, 14 Ren Min Nan Road, Chengdu, 610041, P. R. China
| | - Xiangyou Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Centre for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, 14 Ren Min Nan Road, Chengdu, 610041, P. R. China
| | - Bing Shi
- State Key Laboratory of Oral Diseases & National Clinical Research Centre for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, 14 Ren Min Nan Road, Chengdu, 610041, P. R. China
| | - Jingtao Li
- State Key Laboratory of Oral Diseases & National Clinical Research Centre for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, 14 Ren Min Nan Road, Chengdu, 610041, P. R. China.
| |
Collapse
|
305
|
A new approach of gene co-expression network inference reveals significant biological processes involved in porcine muscle development in late gestation. Sci Rep 2018; 8:10150. [PMID: 29977047 PMCID: PMC6033925 DOI: 10.1038/s41598-018-28173-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 06/14/2018] [Indexed: 12/28/2022] Open
Abstract
The integration of genetic information in the cellular and nuclear environments is crucial for deciphering the way in which the genome functions under different physiological conditions. Experimental techniques of 3D nuclear mapping, a high-flow approach such as transcriptomic data analyses, and statistical methods for the development of co-expressed gene networks, can be combined to develop an integrated approach for depicting the regulation of gene expression. Our work focused more specifically on the mechanisms involved in the transcriptional regulation of genes expressed in muscle during late foetal development in pig. The data generated by a transcriptomic analysis carried out on muscle of foetuses from two extreme genetic lines for birth mortality are used to construct networks of differentially expressed and co-regulated genes. We developed an innovative co-expression networking approach coupling, by means of an iterative process, a new statistical method for graph inference with data of gene spatial co-localization (3D DNA FISH) to construct a robust network grouping co-expressed genes. This enabled us to highlight relevant biological processes related to foetal muscle maturity and to discover unexpected gene associations between IGF2, MYH3 and DLK1/MEG3 in the nuclear space, genes that are up-regulated at this stage of muscle development.
Collapse
|
306
|
Ahrens HE, Huettemeister J, Schmidt M, Kaether C, von Maltzahn J. Klotho expression is a prerequisite for proper muscle stem cell function and regeneration of skeletal muscle. Skelet Muscle 2018; 8:20. [PMID: 29973273 PMCID: PMC6030782 DOI: 10.1186/s13395-018-0166-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 06/21/2018] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Klotho is a well-known anti-aging hormone, which serves as a suppressor of aging through a variety of mechanisms. Aging of skeletal muscle is concomitant with a decrease in muscle stem cell function resulting in impaired regeneration. METHODS Here we investigate the functional role of the anti-aging hormone Klotho for muscle stem cell function after cardiotoxin-induced injury of skeletal muscle using a klotho hypomorphic mouse line, which is characterized by a premature aging phenotype. Furthermore, we perform floating single myofiber cultures with their adjacent muscle stem cells to investigate the interplay between canonical Wnt signaling and Klotho function. RESULTS We demonstrate that muscle stem cell numbers are significantly decreased in klotho hypomorphic mice. Furthermore, we show that muscle stem cell function is also severely impaired upon loss of klotho expression, in culture and during regeneration in vivo. Moreover, we demonstrate that addition of recombinant Klotho protein inhibits aberrant excessive Wnt signaling in aged muscle stem cells thereby restoring their functionality. CONCLUSIONS The anti-aging hormone Klotho counteracts aberrant canonical Wnt signaling in muscle stem cells and might be one of the naturally occurring inhibitors of canonical Wnt signaling in skeletal muscle.
Collapse
Affiliation(s)
- Hellen E Ahrens
- Leibniz-Institute on Aging - Fritz-Lipmann-Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Judith Huettemeister
- Leibniz-Institute on Aging - Fritz-Lipmann-Institute, Beutenbergstrasse 11, 07745, Jena, Germany.,Present address: Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Manuel Schmidt
- Leibniz-Institute on Aging - Fritz-Lipmann-Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Christoph Kaether
- Leibniz-Institute on Aging - Fritz-Lipmann-Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Julia von Maltzahn
- Leibniz-Institute on Aging - Fritz-Lipmann-Institute, Beutenbergstrasse 11, 07745, Jena, Germany.
| |
Collapse
|
307
|
Cameron-Christie SR, Wells CF, Simon M, Wessels M, Tang CZN, Wei W, Takei R, Aarts-Tesselaar C, Sandaradura S, Sillence DO, Cordier MP, Veenstra-Knol HE, Cassina M, Ludwig K, Trevisson E, Bahlo M, Markie DM, Jenkins ZA, Robertson SP. Recessive Spondylocarpotarsal Synostosis Syndrome Due to Compound Heterozygosity for Variants in MYH3. Am J Hum Genet 2018; 102:1115-1125. [PMID: 29805041 PMCID: PMC5992117 DOI: 10.1016/j.ajhg.2018.04.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 04/13/2018] [Indexed: 11/23/2022] Open
Abstract
Spondylocarpotarsal synostosis syndrome (SCTS) is characterized by intervertebral fusions and fusion of the carpal and tarsal bones. Biallelic mutations in FLNB cause this condition in some families, whereas monoallelic variants in MYH3, encoding embryonic heavy chain myosin 3, have been implicated in dominantly inherited forms of the disorder. Here, five individuals without FLNB mutations from three families were hypothesized to be affected by recessive SCTS on account of sibling recurrence of the phenotype. Initial whole-exome sequencing (WES) showed that all five were heterozygous for one of two independent splice-site variants in MYH3. Despite evidence indicating that three of the five individuals shared two allelic haplotypes encompassing MYH3, no second variant could be located in the WES datasets. Subsequent genome sequencing of these three individuals demonstrated a variant altering a 5' UTR splice donor site (rs557849165 in MYH3) not represented by exome-capture platforms. When the cohort was expanded to 16 SCTS-affected individuals without FLNB mutations, nine had truncating mutations transmitted by unaffected parents, and six inherited the rs557849165 variant in trans, an observation at odds with the population allele frequency for this variant. The rs557849165 variant disrupts splicing in the 5' UTR but is still permissive of MYH3 translational initiation, albeit with reduced efficiency. Although some MYH3 variants cause dominant SCTS, these data indicate that others (notably truncating variants) do not, except in the context of compound heterozygosity for a second hypomorphic allele. These observations make genetic diagnosis challenging in the context of simplex presentations of the disorder.
Collapse
Affiliation(s)
- Sophia R Cameron-Christie
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand
| | - Constance F Wells
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand; Paris Diderot University, Sorbonne Paris Cité, Faculty of Medicine, Paris 75007, France; Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Centre Hospitalier Universitaire de Montpellier, Université de Montpellier 34295, Montpellier Cedex 5, France
| | - Marleen Simon
- Department of Genetics, University Medical Center Utrecht, 3508 GA Utrecht, the Netherlands; Department of Clinical Genetics, Erasmus Medical Center, Rotterdam 3015 CE, the Netherlands
| | - Marja Wessels
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam 3015 CE, the Netherlands
| | - Candy Z N Tang
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand
| | - Wenhua Wei
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand
| | - Riku Takei
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand
| | | | - Sarah Sandaradura
- Department of Clinical Genetics Children's Hospital at Westmead, Sydney, NSW 2145, Australia; Discipline of Child and Adolescent Health, University of Sydney, Sydney, NSW 2006, Australia
| | - David O Sillence
- Department of Clinical Genetics Children's Hospital at Westmead, Sydney, NSW 2145, Australia; Discipline of Genetic Medicine, University of Sydney, Sydney, NSW 2006, Australia
| | - Marie-Pierre Cordier
- Clinical Genetics, Hôpital Femme Mère Enfant, Hôpitaux de Lyon, Lyon 69677, France
| | - Hermine E Veenstra-Knol
- Department of Medical Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, the Netherlands
| | - Matteo Cassina
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Padova 35128, Italy
| | - Kathrin Ludwig
- Cardiovascular Pathology Unit, University Hospital of Padova, Padova 35128, Italy
| | - Eva Trevisson
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Padova 35128, Italy
| | - Melanie Bahlo
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, University of Melbourne, Melbourne, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - David M Markie
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand
| | - Zandra A Jenkins
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand
| | - Stephen P Robertson
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand.
| |
Collapse
|
308
|
Spletter ML, Barz C, Yeroslaviz A, Zhang X, Lemke SB, Bonnard A, Brunner E, Cardone G, Basler K, Habermann BH, Schnorrer F. A transcriptomics resource reveals a transcriptional transition during ordered sarcomere morphogenesis in flight muscle. eLife 2018; 7:34058. [PMID: 29846170 PMCID: PMC6005683 DOI: 10.7554/elife.34058] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 05/26/2018] [Indexed: 01/07/2023] Open
Abstract
Muscles organise pseudo-crystalline arrays of actin, myosin and titin filaments to build force-producing sarcomeres. To study sarcomerogenesis, we have generated a transcriptomics resource of developing Drosophila flight muscles and identified 40 distinct expression profile clusters. Strikingly, most sarcomeric components group in two clusters, which are strongly induced after all myofibrils have been assembled, indicating a transcriptional transition during myofibrillogenesis. Following myofibril assembly, many short sarcomeres are added to each myofibril. Subsequently, all sarcomeres mature, reaching 1.5 µm diameter and 3.2 µm length and acquiring stretch-sensitivity. The efficient induction of the transcriptional transition during myofibrillogenesis, including the transcriptional boost of sarcomeric components, requires in part the transcriptional regulator Spalt major. As a consequence of Spalt knock-down, sarcomere maturation is defective and fibers fail to gain stretch-sensitivity. Together, this defines an ordered sarcomere morphogenesis process under precise transcriptional control - a concept that may also apply to vertebrate muscle or heart development.
Collapse
Affiliation(s)
- Maria L Spletter
- Muscle Dynamics GroupMax Planck Institute of BiochemistryMartinsriedGermany
- Biomedical Center, Physiological ChemistryLudwig-Maximilians-Universität MünchenMartinsriedGermany
| | - Christiane Barz
- Muscle Dynamics GroupMax Planck Institute of BiochemistryMartinsriedGermany
| | - Assa Yeroslaviz
- Computational Biology GroupMax Planck Institute of BiochemistryMartinsriedGermany
| | - Xu Zhang
- Muscle Dynamics GroupMax Planck Institute of BiochemistryMartinsriedGermany
- Aix Marseille Univ, CNRS, IBDMMarseilleFrance
- School of Life Science and EngineeringFoshan UniversityGuangdongChina
| | - Sandra B Lemke
- Muscle Dynamics GroupMax Planck Institute of BiochemistryMartinsriedGermany
| | - Adrien Bonnard
- Aix Marseille Univ, CNRS, IBDMMarseilleFrance
- Aix Marseille Univ, INSERM, TAGCMarseilleFrance
| | - Erich Brunner
- Institute of Molecular Life SciencesUniversity of ZurichZurichSwitzerland
| | - Giovanni Cardone
- Imaging FacilityMax Planck Institute of BiochemistryMartinsriedGermany
| | - Konrad Basler
- Institute of Molecular Life SciencesUniversity of ZurichZurichSwitzerland
| | - Bianca H Habermann
- Computational Biology GroupMax Planck Institute of BiochemistryMartinsriedGermany
- Aix Marseille Univ, CNRS, IBDMMarseilleFrance
- Aix Marseille Univ, INSERM, TAGCMarseilleFrance
| | - Frank Schnorrer
- Muscle Dynamics GroupMax Planck Institute of BiochemistryMartinsriedGermany
- Aix Marseille Univ, CNRS, IBDMMarseilleFrance
| |
Collapse
|
309
|
André LM, Ausems CRM, Wansink DG, Wieringa B. Abnormalities in Skeletal Muscle Myogenesis, Growth, and Regeneration in Myotonic Dystrophy. Front Neurol 2018; 9:368. [PMID: 29892259 PMCID: PMC5985300 DOI: 10.3389/fneur.2018.00368] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 05/07/2018] [Indexed: 12/16/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) and 2 (DM2) are autosomal dominant degenerative neuromuscular disorders characterized by progressive skeletal muscle weakness, atrophy, and myotonia with progeroid features. Although both DM1 and DM2 are characterized by skeletal muscle dysfunction and also share other clinical features, the diseases differ in the muscle groups that are affected. In DM1, distal muscles are mainly affected, whereas in DM2 problems are mostly found in proximal muscles. In addition, manifestation in DM1 is generally more severe, with possible congenital or childhood-onset of disease and prominent CNS involvement. DM1 and DM2 are caused by expansion of (CTG•CAG)n and (CCTG•CAGG)n repeats in the 3' non-coding region of DMPK and in intron 1 of CNBP, respectively, and in overlapping antisense genes. This critical review will focus on the pleiotropic problems that occur during development, growth, regeneration, and aging of skeletal muscle in patients who inherited these expansions. The current best-accepted idea is that most muscle symptoms can be explained by pathomechanistic effects of repeat expansion on RNA-mediated pathways. However, aberrations in DNA replication and transcription of the DM loci or in protein translation and proteome homeostasis could also affect the control of proliferation and differentiation of muscle progenitor cells or the maintenance and physiological integrity of muscle fibers during a patient's lifetime. Here, we will discuss these molecular and cellular processes and summarize current knowledge about the role of embryonic and adult muscle-resident stem cells in growth, homeostasis, regeneration, and premature aging of healthy and diseased muscle tissue. Of particular interest is that also progenitor cells from extramuscular sources, such as pericytes and mesoangioblasts, can participate in myogenic differentiation. We will examine the potential of all these types of cells in the application of regenerative medicine for muscular dystrophies and evaluate new possibilities for their use in future therapy of DM.
Collapse
Affiliation(s)
- Laurène M André
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - C Rosanne M Ausems
- Department of Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Derick G Wansink
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Bé Wieringa
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
310
|
Schiaffino S. Muscle fiber type diversity revealed by anti-myosin heavy chain antibodies. FEBS J 2018; 285:3688-3694. [PMID: 29761627 DOI: 10.1111/febs.14502] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 04/24/2018] [Accepted: 05/08/2018] [Indexed: 01/02/2023]
Abstract
Different forms of myosin heavy chains (MyHCs), coded by a large family of sarcomeric MYH genes, are expressed in striated muscles. The generation of specific anti-MyHC antibodies has provided a powerful tool to define the fiber types present in skeletal muscles, their functional properties, their response to conditions that affect muscle plasticity and their changes in muscle disorders. Cardiomyocyte heterogeneity has been revealed by the serendipitous observation that different MyHCs are present in atrial and ventricular myocardium and in heart conduction tissue. Developmental MyHCs present in embryonic and fetal/neonatal skeletal muscle are re-expressed during muscle regeneration and can be used to identify regenerating fibers in muscle diseases. MyHC isoforms provide cell type-specific markers to identify the signaling pathways that control muscle cell identity and are an essential reference to interpret the results of single-cell transcriptomics and proteomics.
Collapse
|
311
|
Abstract
PURPOSE OF REVIEW The review is focused on the unexpected role of myogenic regulatory factor 4 (MRF4) in controlling muscle mass by repressing myocyte enhancer binding factor 2 (MEF2) activity in adult skeletal muscle, and on the emerging role of MEF2 in skeletal muscle growth. RECENT FINDINGS The MRF4s of the MyoD family (MyoD, MYF5, MRF4, myogenin) and the MEF2 factors are known to play a major role in embryonic myogenesis. However, their function in adult muscle tissue is not known. A recent study shows that MRF4 loss in adult skeletal muscle causes muscle hypertrophy and prevents denervation atrophy. This effect is mediated by MEF2 factors that promote muscle growth, with MRF4 acting as a repressor of MEF2 activity. The role of MEF2 in skeletal muscle growth is supported by the finding that muscle regeneration is impaired by muscle-specific triple knockout of Mef2a, c, and d genes. SUMMARY The finding that the MRF4-MEF2 axis controls muscle growth opens a new perspective for preventing muscle wasting. A unique feature of this pathway is that MRF4 is exclusively expressed in skeletal muscle, thus reducing the risk that interventions aimed at down-regulating MRF4 or interfering with the interaction between MRF4 and MEF2 may have off-target effects in other tissues.
Collapse
Affiliation(s)
| | - Kenneth A Dyar
- Molecular Endocrinology, Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HMGU) and German Center for Diabetes Research (DZD), Munich, Germany
| | - Elisa Calabria
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
312
|
Lund J, S Tangen D, Wiig H, Stadheim HK, Helle SA, B Birk J, Ingemann-Hansen T, Rustan AC, Thoresen GH, Wojtaszewski JFP, T Kase E, Jensen J. Glucose metabolism and metabolic flexibility in cultured skeletal muscle cells is related to exercise status in young male subjects. Arch Physiol Biochem 2018; 124:119-130. [PMID: 28862046 DOI: 10.1080/13813455.2017.1369547] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
We hypothesised that skeletal muscles of healthy young people have a large variation in oxidative capacity and fibre-type composition, and aimed therefore to investigate glucose metabolism in biopsies and myotubes isolated from musculus vastus lateralis from healthy males with varying degrees of maximal oxygen uptake. Trained and intermediary trained subjects showed higher carbohydrate oxidation in vivo. Fibre-type distribution in biopsies and myotubes did not differ between groups. There was no correlation between fibre-type I expression in biopsies and myotubes. Myotubes from trained had higher deoxyglucose accumulation and fractional glucose oxidation (glucose oxidation relative to glucose uptake), and were also more sensitive to the suppressive action of acutely added oleic acid to the cells. Despite lack of correlation of fibre types between skeletal muscle biopsies and cultured cells, myotubes from trained subjects retained some of their phenotypes in vitro with respect to enhanced glucose metabolism and metabolic flexibility.
Collapse
Affiliation(s)
- Jenny Lund
- a Department of Pharmaceutical Biosciences , School of Pharmacy, University of Oslo , Oslo , Norway
| | - Daniel S Tangen
- b Department of Physical Performance , Norwegian School of Sport Sciences , Oslo , Norway
| | - Håvard Wiig
- b Department of Physical Performance , Norwegian School of Sport Sciences , Oslo , Norway
| | - Hans K Stadheim
- b Department of Physical Performance , Norwegian School of Sport Sciences , Oslo , Norway
| | - Siw A Helle
- a Department of Pharmaceutical Biosciences , School of Pharmacy, University of Oslo , Oslo , Norway
| | - Jesper B Birk
- c Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science , University of Copenhagen , Copenhagen , Denmark
| | | | - Arild C Rustan
- a Department of Pharmaceutical Biosciences , School of Pharmacy, University of Oslo , Oslo , Norway
| | - G Hege Thoresen
- a Department of Pharmaceutical Biosciences , School of Pharmacy, University of Oslo , Oslo , Norway
- e Department of Pharmacology , Institute of Clinical Medicine, University of Oslo , Oslo , Norway
| | - Jørgen F P Wojtaszewski
- c Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science , University of Copenhagen , Copenhagen , Denmark
| | - Eili T Kase
- a Department of Pharmaceutical Biosciences , School of Pharmacy, University of Oslo , Oslo , Norway
| | - Jørgen Jensen
- b Department of Physical Performance , Norwegian School of Sport Sciences , Oslo , Norway
| |
Collapse
|
313
|
Schiaffino S. Knockout of human muscle genes revealed by large scale whole-exome studies. Mol Genet Metab 2018; 123:411-415. [PMID: 29452748 DOI: 10.1016/j.ymgme.2018.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 02/06/2018] [Accepted: 02/06/2018] [Indexed: 12/22/2022]
Abstract
Large scale whole-exome sequence studies have revealed that a number of individuals from different populations have predicted loss-of-function of different genes due to nonsense, frameshift, or canonical splice-site mutations. Surprisingly, many of these mutations do not apparently show the deleterious phenotypic consequences expected from gene knockout. These homozygous null mutations, when confirmed, can provide insight into human gene function and suggest novel approaches to correct gene dysfunction, as the lack of the expected disease phenotype may reflect the existence of modifier genes that reveal potential therapeutic targets. Human knockouts complement the information derived from mouse knockouts, which are not always good models of human disease. We have examined human knockout datasets searching for genes expressed exclusively or predominantly in striated muscle. A number of well-known muscle genes was found in one or more datasets, including genes coding for sarcomeric myosins, components of the sarcomeric cytoskeleton, sarcoplasmic reticulum and plasma membrane, and enzymes involved in muscle metabolism. The surprising absence of phenotype in some of these human knockouts is critically discussed, focusing on the comparison with the corresponding mouse knockouts.
Collapse
|
314
|
Bortolini MAT, Feitosa SM, Bilhar APM, Salerno GGR, Zanoteli E, Simões MJ, Castro RA. Molecular and histomorphological evaluation of female rats’ urethral tissues after an innovative trauma model of prolonged vaginal distention: immediate, short-term and long-term effects. Int Urogynecol J 2018; 30:465-476. [DOI: 10.1007/s00192-018-3634-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 03/08/2018] [Indexed: 02/06/2023]
|
315
|
Expression profiling of disease progression in canine model of Duchenne muscular dystrophy. PLoS One 2018; 13:e0194485. [PMID: 29554127 PMCID: PMC5858769 DOI: 10.1371/journal.pone.0194485] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 03/05/2018] [Indexed: 12/17/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) causes progressive disability in 1 of every 5,000 boys due to the lack of functional dystrophin protein. Despite much advancement in knowledge about DMD disease presentation and progression—attributable in part to studies using mouse and canine models of the disease–current DMD treatments are not equally effective in all patients. There remains, therefore, a need for translational animal models in which novel treatment targets can be identified and evaluated. Golden Retriever muscular dystrophy (GRMD) is a phenotypically and genetically homologous animal model of DMD. As with DMD, speed of disease progression in GRMD varies substantially. However, unlike DMD, all GRMD dogs possess the same causal mutation; therefore genetic modifiers of phenotypic variation are relatively easier to identify. Furthermore, the GRMD dogs used in this study reside within the same colony, reducing the confounding effects of environment on phenotypic variation. To detect modifiers of disease progression, we developed gene expression profiles using RNA sequencing for 9 dogs: 6 GRMD dogs (3 with faster-progressing and 3 with slower-progressing disease, based on quantitative, objective biomarkers) and 3 control dogs from the same colony. All dogs were evaluated at 2 time points: early disease onset (3 months of age) and the point at which GRMD stabilizes (6 months of age) using quantitative, objective biomarkers identified as robust against the effects of relatedness/inbreeding. Across all comparisons, the most differentially expressed genes fell into 3 categories: myogenesis/muscle regeneration, metabolism, and inflammation. Our findings are largely in concordance with DMD and mouse model studies, reinforcing the utility of GRMD as a translational model. Novel findings include the strong up-regulation of chitinase 3-like 1 (CHI3L1) in faster-progressing GRMD dogs, suggesting previously unexplored mechanisms underlie progression speed in GRMD and DMD. In summary, our findings support the utility of RNA sequencing for evaluating potential biomarkers of GRMD progression speed, and are valuable for identifying new avenues of exploration in DMD research.
Collapse
|
316
|
Wang L, Geist J, Grogan A, Hu LYR, Kontrogianni-Konstantopoulos A. Thick Filament Protein Network, Functions, and Disease Association. Compr Physiol 2018; 8:631-709. [PMID: 29687901 PMCID: PMC6404781 DOI: 10.1002/cphy.c170023] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sarcomeres consist of highly ordered arrays of thick myosin and thin actin filaments along with accessory proteins. Thick filaments occupy the center of sarcomeres where they partially overlap with thin filaments. The sliding of thick filaments past thin filaments is a highly regulated process that occurs in an ATP-dependent manner driving muscle contraction. In addition to myosin that makes up the backbone of the thick filament, four other proteins which are intimately bound to the thick filament, myosin binding protein-C, titin, myomesin, and obscurin play important structural and regulatory roles. Consistent with this, mutations in the respective genes have been associated with idiopathic and congenital forms of skeletal and cardiac myopathies. In this review, we aim to summarize our current knowledge on the molecular structure, subcellular localization, interacting partners, function, modulation via posttranslational modifications, and disease involvement of these five major proteins that comprise the thick filament of striated muscle cells. © 2018 American Physiological Society. Compr Physiol 8:631-709, 2018.
Collapse
Affiliation(s)
- Li Wang
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Janelle Geist
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Alyssa Grogan
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Li-Yen R. Hu
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | | |
Collapse
|
317
|
D'Souza RF, Zeng N, Markworth JF, Figueiredo VC, Roberts LA, Raastad T, Coombes JS, Peake JM, Cameron-Smith D, Mitchell CJ. Divergent effects of cold water immersion versus active recovery on skeletal muscle fiber type and angiogenesis in young men. Am J Physiol Regul Integr Comp Physiol 2018; 314:R824-R833. [PMID: 29466686 DOI: 10.1152/ajpregu.00421.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Resistance training (RT) increases muscle fiber size and induces angiogenesis to maintain capillary density. Cold water immersion (CWI), a common postexercise recovery modality, may improve acute recovery, but it attenuates muscle hypertrophy compared with active recovery (ACT). It is unknown if CWI following RT alters muscle fiber type expression or angiogenesis. Twenty-one men strength trained for 12 wk, with either 10 min of CWI ( n = 11) or ACT ( n = 10) performed following each session. Vastus lateralis biopsies were collected at rest before and after training. Type IIx myofiber percent decreased ( P = 0.013) and type IIa myofiber percent increased with training ( P = 0.012), with no difference between groups. The number of capillaries per fiber increased from pretraining in the CWI group ( P = 0.004) but not the ACT group ( P = 0.955). Expression of myosin heavy chain genes ( MYH1 and MYH2), encoding type IIx and IIa fibers, respectively, decreased in the ACT group, whereas MYH7 (encoding type I fibers) increased in the ACT group versus CWI ( P = 0.004). Myosin heavy chain IIa protein increased with training ( P = 0.012) with no difference between groups. The proangiogenic vascular endothelial growth factor protein decreased posttraining in the ACT group versus CWI ( P < 0.001), whereas antiangiogenic Sprouty-related, EVH1 domain-containing protein 1 protein increased with training in both groups ( P = 0.015). Expression of microRNAs that regulate muscle fiber type (miR-208b and -499a) and angiogenesis (miR-15a, -16, and -126) increased only in the ACT group ( P < 0.05). CWI recovery after each training session altered the angiogenic and fiber type-specific response to RT through regulation at the levels of microRNA, gene, and protein expression.
Collapse
Affiliation(s)
- Randall F D'Souza
- Liggins Institute, The University of Auckland , Auckland , New Zealand
| | - Nina Zeng
- Liggins Institute, The University of Auckland , Auckland , New Zealand
| | - James F Markworth
- Liggins Institute, The University of Auckland , Auckland , New Zealand
| | - Vandre C Figueiredo
- Liggins Institute, The University of Auckland , Auckland , New Zealand.,Center for Muscle Biology, University of Kentucky , Lexington, Kentucky
| | - Llion A Roberts
- School of Human Movement and Nutrition Sciences, University of Queensland , Brisbane , Australia.,Sports Performance Innovation and Knowledge Excellence, Queensland Academy of Sport , Brisbane , Australia.,School of Allied Health Sciences & Menzies Health Institute Queensland, Griffith University , Gold Coast, Queensland , Australia
| | - Truls Raastad
- Department of Physical Performance, Norwegian School of Sport Sciences , Oslo , Norway
| | - Jeff S Coombes
- School of Human Movement and Nutrition Sciences, University of Queensland , Brisbane , Australia
| | - Jonathan M Peake
- Sports Performance Innovation and Knowledge Excellence, Queensland Academy of Sport , Brisbane , Australia.,School of Biomedical Sciences and Institute of Health and Biomedical Innovation, Queensland University of Technology , Brisbane Australia
| | - David Cameron-Smith
- Liggins Institute, The University of Auckland , Auckland , New Zealand.,Food & Bio-Based Products Group, AgResearch, Palmerston North , New Zealand.,Riddet Institute , Palmerston North , New Zealand
| | | |
Collapse
|
318
|
Rao L, Qian Y, Khodabukus A, Ribar T, Bursac N. Engineering human pluripotent stem cells into a functional skeletal muscle tissue. Nat Commun 2018; 9:126. [PMID: 29317646 PMCID: PMC5760720 DOI: 10.1038/s41467-017-02636-4] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 12/14/2017] [Indexed: 12/24/2022] Open
Abstract
The generation of functional skeletal muscle tissues from human pluripotent stem cells (hPSCs) has not been reported. Here, we derive induced myogenic progenitor cells (iMPCs) via transient overexpression of Pax7 in paraxial mesoderm cells differentiated from hPSCs. In 2D culture, iMPCs readily differentiate into spontaneously contracting multinucleated myotubes and a pool of satellite-like cells endogenously expressing Pax7. Under optimized 3D culture conditions, iMPCs derived from multiple hPSC lines reproducibly form functional skeletal muscle tissues (iSKM bundles) containing aligned multi-nucleated myotubes that exhibit positive force-frequency relationship and robust calcium transients in response to electrical or acetylcholine stimulation. During 1-month culture, the iSKM bundles undergo increased structural and molecular maturation, hypertrophy, and force generation. When implanted into dorsal window chamber or hindlimb muscle in immunocompromised mice, the iSKM bundles survive, progressively vascularize, and maintain functionality. iSKM bundles hold promise as a microphysiological platform for human muscle disease modeling and drug development.
Collapse
Affiliation(s)
- Lingjun Rao
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Ying Qian
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Alastair Khodabukus
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Thomas Ribar
- Duke iPSC Shared Resource Facility, Duke University, Durham, NC, 27708, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA.
| |
Collapse
|
319
|
Raz V, Riaz M, Tatum Z, Kielbasa SM, 't Hoen PAC. The distinct transcriptomes of slow and fast adult muscles are delineated by noncoding RNAs. FASEB J 2018; 32:1579-1590. [PMID: 29141996 DOI: 10.1096/fj.201700861r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Adult muscles have a vast adaptation capacity, enabling function switches in response to altered conditions. During intensive physical activity, disease, or aging, adult skeletal muscles change and adjust their functions. The competence to adjust varies among muscles. Muscle-specific molecular mechanisms in healthy and normal conditions could designate changes in physiologic and pathologic conditions. We generated deep mRNA-sequencing data in adult fast and slow mouse muscles, and applying paired analysis, we identified that the muscle-specific signatures are composed of half of the muscle transcriptome. The fast muscles showed a more compact gene network that is concordant with homogenous myofiber typing, compared with the pattern in the slow muscle. The muscle-specific mRNA landscape did not correlate with alternative spicing, alternative polyadenylation, or the expression of muscle transcription factor gene networks. However, we found significant correlation between the differentially expressed noncoding RNAs, microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) and their target genes. More than 25% of the genes expressed in a muscle-specific fashion were found to be targets of muscle-specific miRNAs and lncRNAs. We suggest that muscle-specific miRNAs and lncRNAs contribute to the establishment of muscle-specific transcriptomes in adult muscles.-Raz, V., Riaz, M., Tatum, Z., Kielbasa, S. M., 't Hoen, P. A. C. The distinct transcriptomes of slow and fast adult muscles are delineated by noncoding RNAs.
Collapse
Affiliation(s)
- Vered Raz
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Muhammad Riaz
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Zuotian Tatum
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Szymon M Kielbasa
- Department of Medical Statistics and Bioinformatics, Bioinformatics Center of Expertise, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter A C 't Hoen
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
320
|
Hicks MR, Hiserodt J, Paras K, Fujiwara W, Eskin A, Jan M, Xi H, Young CS, Evseenko D, Nelson SF, Spencer MJ, Handel BV, Pyle AD. ERBB3 and NGFR mark a distinct skeletal muscle progenitor cell in human development and hPSCs. Nat Cell Biol 2018; 20:46-57. [PMID: 29255171 PMCID: PMC5962356 DOI: 10.1038/s41556-017-0010-2] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 11/16/2017] [Indexed: 12/24/2022]
Abstract
Human pluripotent stem cells (hPSCs) can be directed to differentiate into skeletal muscle progenitor cells (SMPCs). However, the myogenicity of hPSC-SMPCs relative to human fetal or adult satellite cells remains unclear. We observed that hPSC-SMPCs derived by directed differentiation are less functional in vitro and in vivo compared to human satellite cells. Using RNA sequencing, we found that the cell surface receptors ERBB3 and NGFR demarcate myogenic populations, including PAX7 progenitors in human fetal development and hPSC-SMPCs. We demonstrated that hPSC skeletal muscle is immature, but inhibition of transforming growth factor-β signalling during differentiation improved fusion efficiency, ultrastructural organization and the expression of adult myosins. This enrichment and maturation strategy restored dystrophin in hundreds of dystrophin-deficient myofibres after engraftment of CRISPR-Cas9-corrected Duchenne muscular dystrophy human induced pluripotent stem cell-SMPCs. The work provides an in-depth characterization of human myogenesis, and identifies candidates that improve the in vivo myogenic potential of hPSC-SMPCs to levels that are equal to directly isolated human fetal muscle cells.
Collapse
MESH Headings
- Adult
- Aged
- CRISPR-Cas Systems
- Cell Differentiation
- Dystrophin/genetics
- Dystrophin/metabolism
- Female
- Gene Editing
- Gene Expression Regulation, Developmental
- Humans
- Induced Pluripotent Stem Cells/cytology
- Induced Pluripotent Stem Cells/metabolism
- Male
- Middle Aged
- Muscle Development/genetics
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/therapy
- Myoblasts/cytology
- Myoblasts/metabolism
- Myosins/genetics
- Myosins/metabolism
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- PAX7 Transcription Factor/genetics
- PAX7 Transcription Factor/metabolism
- Receptor, ErbB-3/genetics
- Receptor, ErbB-3/metabolism
- Receptors, Nerve Growth Factor/genetics
- Receptors, Nerve Growth Factor/metabolism
- Signal Transduction
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
Collapse
Affiliation(s)
- Michael R Hicks
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Julia Hiserodt
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Katrina Paras
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Wakana Fujiwara
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Ascia Eskin
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
- Department of Human Genetics, University of California, Los Angeles, CA, USA
| | - Majib Jan
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Haibin Xi
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Courtney S Young
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Denis Evseenko
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Stanley F Nelson
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Melissa J Spencer
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA, USA
- Department of Neurology, University of California, Los Angeles, CA, USA
| | - Ben Van Handel
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - April D Pyle
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA.
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA.
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA.
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA, USA.
| |
Collapse
|
321
|
Bloise FF, Cordeiro A, Ortiga-Carvalho TM. Role of thyroid hormone in skeletal muscle physiology. J Endocrinol 2018; 236:R57-R68. [PMID: 29051191 DOI: 10.1530/joe-16-0611] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 10/19/2017] [Indexed: 12/31/2022]
Abstract
Thyroid hormones (TH) are crucial for development, growth, differentiation, metabolism and thermogenesis. Skeletal muscle (SM) contractile function, myogenesis and bioenergetic metabolism are influenced by TH. These effects depend on the presence of the TH transporters MCT8 and MCT10 in the plasma membrane, the expression of TH receptors (THRA or THRB) and hormone availability, which is determined either by the activation of thyroxine (T4) into triiodothyronine (T3) by type 2 iodothyronine deiodinases (D2) or by the inactivation of T4 into reverse T3 by deiodinases type 3 (D3). SM relaxation and contraction rates depend on T3 regulation of myosin expression and energy supplied by substrate oxidation in the mitochondria. The balance between D2 and D3 expression determines TH intracellular levels and thus influences the proliferation and differentiation of satellite cells, indicating an important role of TH in muscle repair and myogenesis. During critical illness, changes in TH levels and in THR and deiodinase expression negatively affect SM function and repair. This review will discuss the influence of TH action on SM contraction, bioenergetics metabolism, myogenesis and repair in health and illness conditions.
Collapse
Affiliation(s)
- Flavia F Bloise
- Institute of Biophysics Carlos Chagas FilhoLaboratory of Translational Endocrinology, Rio de Janeiro, Brazil
| | - Aline Cordeiro
- Institute of Biophysics Carlos Chagas FilhoLaboratory of Translational Endocrinology, Rio de Janeiro, Brazil
| | - Tania Maria Ortiga-Carvalho
- Institute of Biophysics Carlos Chagas FilhoLaboratory of Translational Endocrinology, Rio de Janeiro, Brazil
| |
Collapse
|
322
|
Rebalka IA, Cao AW, Raleigh MJ, Henriksbo BD, Coleman SK, Schertzer JD, Hawke TJ. Statin Therapy Negatively Impacts Skeletal Muscle Regeneration and Cutaneous Wound Repair in Type 1 Diabetic Mice. Front Physiol 2017; 8:1088. [PMID: 29311999 PMCID: PMC5742241 DOI: 10.3389/fphys.2017.01088] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 12/11/2017] [Indexed: 12/26/2022] Open
Abstract
Those with diabetes invariably develop complications including cardiovascular disease (CVD). To reduce their CVD risk, diabetics are generally prescribed cholesterol-lowering 3-hydroxy-methylglutaryl coenzyme A reductase inhibitors (i.e., statins). Statins inhibit cholesterol biosynthesis, but also reduce the synthesis of a number of mevalonate pathway intermediates, leading to several cholesterol-independent effects. One of the pleiotropic effects of statins is the reduction of the anti-fibrinolytic hormone plasminogen activator inhibitor-1 (PAI-1). We have previously demonstrated that a PAI-1 specific inhibitor alleviated diabetes-induced delays in skin and muscle repair. Here we tested if statin administration, through its pleiotropic effects on PAI-1, could improve skin and muscle repair in a diabetic rodent model. Six weeks after diabetes onset, adult male streptozotocin-induced diabetic (STZ), and WT mice were assigned to receive control chow or a diet enriched with 600 mg/kg Fluvastatin. Tibialis anterior muscles were injured via Cardiotoxin injection to induce skeletal muscle injury. Punch biopsies were administered on the dorsal scapular region to induce injury of skin. Twenty-four days after the onset of statin therapy (10 days post-injury), tissues were harvested and analyzed. PAI-1 levels were attenuated in statin-treated diabetic tissue when compared to control-treated tissue, however no differences were observed in non-diabetic tissue as a result of treatment. Muscle and skin repair were significantly attenuated in Fluvastatin-treated STZ-diabetic mice as demonstrated by larger wound areas, less mature granulation tissue, and an increased presence of smaller regenerating muscle fibers. Despite attenuating PAI-1 levels in diabetic tissue, Fluvastatin treatment impaired cutaneous healing and skeletal muscle repair in STZ-diabetic mice.
Collapse
Affiliation(s)
- Irena A Rebalka
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Andrew W Cao
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Matthew J Raleigh
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Brandyn D Henriksbo
- Department of Biochemistry and Biomedical Sciences and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Samantha K Coleman
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Thomas J Hawke
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
323
|
Downregulation of myostatin pathway in neuromuscular diseases may explain challenges of anti-myostatin therapeutic approaches. Nat Commun 2017; 8:1859. [PMID: 29192144 PMCID: PMC5709430 DOI: 10.1038/s41467-017-01486-4] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 09/21/2017] [Indexed: 02/07/2023] Open
Abstract
Muscular dystrophies are characterized by weakness and wasting of skeletal muscle tissues. Several drugs targeting the myostatin pathway have been used in clinical trials to increase muscle mass and function but most showed limited efficacy. Here we show that the expression of components of the myostatin signaling pathway is downregulated in muscle wasting or atrophying diseases, with a decrease of myostatin and activin receptor, and an increase of the myostatin antagonist, follistatin. We also provide in vivo evidence in the congenital myotubular myopathy mouse model (knock-out for the myotubularin coding gene Mtm1) that a down-regulated myostatin pathway can be reactivated by correcting the underlying gene defect. Our data may explain the poor clinical efficacy of anti-myostatin approaches in several of the clinical studies and the apparent contradictory results in mice regarding the efficacy of anti-myostatin approaches and may inform patient selection and stratification for future trials.
Collapse
|
324
|
Crawford Parks TE, Ravel-Chapuis A, Bondy-Chorney E, Renaud JM, Côté J, Jasmin BJ. Muscle-specific expression of the RNA-binding protein Staufen1 induces progressive skeletal muscle atrophy via regulation of phosphatase tensin homolog. Hum Mol Genet 2017; 26:1821-1838. [PMID: 28369467 DOI: 10.1093/hmg/ddx085] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 03/02/2017] [Indexed: 12/14/2022] Open
Abstract
Converging lines of evidence have now highlighted the key role for post-transcriptional regulation in the neuromuscular system. In particular, several RNA-binding proteins are known to be misregulated in neuromuscular disorders including myotonic dystrophy type 1, spinal muscular atrophy and amyotrophic lateral sclerosis. In this study, we focused on the RNA-binding protein Staufen1, which assumes multiple functions in both skeletal muscle and neurons. Given our previous work that showed a marked increase in Staufen1 expression in various physiological and pathological conditions including denervated muscle, in embryonic and undifferentiated skeletal muscle, in rhabdomyosarcomas as well as in myotonic dystrophy type 1 muscle samples from both mouse models and humans, we investigated the impact of sustained Staufen1 expression in postnatal skeletal muscle. To this end, we generated a skeletal muscle-specific transgenic mouse model using the muscle creatine kinase promoter to drive tissue-specific expression of Staufen1. We report that sustained Staufen1 expression in postnatal skeletal muscle causes a myopathy characterized by significant morphological and functional deficits. These deficits are accompanied by a marked increase in the expression of several atrophy-associated genes and by the negative regulation of PI3K/AKT signaling. We also uncovered that Staufen1 mediates PTEN expression through indirect transcriptional and direct post-transcriptional events thereby providing the first evidence for Staufen1-regulated PTEN expression. Collectively, our data demonstrate that Staufen1 is a novel atrophy-associated gene, and highlight its potential as a biomarker and therapeutic target for neuromuscular disorders and conditions.
Collapse
Affiliation(s)
- Tara E Crawford Parks
- Department of Cellular and Molecular Medicine, Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Aymeric Ravel-Chapuis
- Department of Cellular and Molecular Medicine, Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Emma Bondy-Chorney
- Department of Cellular and Molecular Medicine, Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jean-Marc Renaud
- Department of Cellular and Molecular Medicine, Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jocelyn Côté
- Department of Cellular and Molecular Medicine, Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Bernard J Jasmin
- Department of Cellular and Molecular Medicine, Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
325
|
Comprehensive comparison of neonate and adult human platelet transcriptomes. PLoS One 2017; 12:e0183042. [PMID: 28813466 PMCID: PMC5559076 DOI: 10.1371/journal.pone.0183042] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 07/30/2017] [Indexed: 12/21/2022] Open
Abstract
Understanding the underlying mechanisms of the well-substantiated platelet hyporeactivity in neonates is of interest given their implications for the clinical management of newborns, a population at higher bleeding risk than adults (especially sick and preterm infants), as well as for gaining insight into the regulatory mechanisms of platelet biology. Transcriptome analysis is useful in identifying mRNA signatures affecting platelet function. However, human fetal/neonatal platelet transcriptome analysis has never before been reported. We have used mRNA expression array for the first time to compare platelet transcriptome changes during development. Microarray analysis was performed in pure platelet RNA obtained from adult and cord blood, using the same platform in two independent laboratories. A high correlation was obtained between array results for both adult and neonate platelet samples. There was also good agreement between results in our adult samples and outcomes previously reported in three different studies. Gene enrichment analysis showed that immunity- and platelet function-related genes are highly expressed at both developmental stages. Remarkably, 201 genes were found to be differentially expressed throughout development. In particular, neonatal platelets contain higher levels of mRNA that are associated with protein synthesis and processing, while carrying significantly lower levels of genes involved in calcium transport/metabolism and cell signaling (including GNAZ). Overall, our results point to variations in platelet transcriptome as possibly underlining the hypo-functional phenotype of neonatal platelets and provide further support for the role of platelets in cellular immune response. Better characterization of the platelet transcriptome throughout development can contribute to elucidate how transcriptome changes impact different pathological conditions.
Collapse
|
326
|
Beauclercq S, Hennequet-Antier C, Praud C, Godet E, Collin A, Tesseraud S, Métayer-Coustard S, Bourin M, Moroldo M, Martins F, Lagarrigue S, Bihan-Duval EL, Berri C. Muscle transcriptome analysis reveals molecular pathways and biomarkers involved in extreme ultimate pH and meat defect occurrence in chicken. Sci Rep 2017; 7:6447. [PMID: 28743971 PMCID: PMC5526995 DOI: 10.1038/s41598-017-06511-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 06/13/2017] [Indexed: 02/06/2023] Open
Abstract
The processing ability and sensory quality of chicken breast meat are highly related to its ultimate pH (pHu), which is mainly determined by the amount of glycogen in the muscle at death. To unravel the molecular mechanisms underlying glycogen and meat pHu variations and to identify predictive biomarkers of these traits, a transcriptome profiling analysis was performed using an Agilent custom chicken 8 × 60 K microarray. The breast muscle gene expression patterns were studied in two chicken lines experimentally selected for high (pHu+) and low (pHu-) pHu values of the breast meat. Across the 1,436 differentially expressed (DE) genes found between the two lines, many were involved in biological processes related to muscle development and remodelling and carbohydrate and energy metabolism. The functional analysis showed an intensive use of carbohydrate metabolism to produce energy in the pHu- line, while alternative catabolic pathways were solicited in the muscle of the pHu+ broilers, compromising their muscle development and integrity. After a validation step on a population of 278 broilers using microfluidic RT-qPCR, 20 genes were identified by partial least squares regression as good predictors of the pHu, opening new perspectives of screening broilers likely to present meat quality defects.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Marie Bourin
- ITAVI-Institut Technique de l'Aviculture, F-37380, Nouzilly, France
| | - Marco Moroldo
- GABI, AgroParisTech, INRA, Université Paris-Saclay, F-78350, Jouy-en-Josas, France
| | - Frédéric Martins
- Plateforme Génome et Transcriptome, Génopole de Toulouse, France.,INSERM, UMR1048, F-31432, Toulouse, France
| | | | | | | |
Collapse
|
327
|
Tascher G, Brioche T, Maes P, Chopard A, O'Gorman D, Gauquelin-Koch G, Blanc S, Bertile F. Proteome-wide Adaptations of Mouse Skeletal Muscles during a Full Month in Space. J Proteome Res 2017; 16:2623-2638. [PMID: 28590761 DOI: 10.1021/acs.jproteome.7b00201] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The safety of space flight is challenged by a severe loss of skeletal muscle mass, strength, and endurance that may compromise the health and performance of astronauts. The molecular mechanisms underpinning muscle atrophy and decreased performance have been studied mostly after short duration flights and are still not fully elucidated. By deciphering the muscle proteome changes elicited in mice after a full month aboard the BION-M1 biosatellite, we observed that the antigravity soleus incurred the greatest changes compared with locomotor muscles. Proteomics data notably suggested mitochondrial dysfunction, metabolic and fiber type switching toward glycolytic type II fibers, structural alterations, and calcium signaling-related defects to be the main causes for decreased muscle performance in flown mice. Alterations of the protein balance, mTOR pathway, myogenesis, and apoptosis were expected to contribute to muscle atrophy. Moreover, several signs reflecting alteration of telomere maintenance, oxidative stress, and insulin resistance were found as possible additional deleterious effects. Finally, 8 days of recovery post flight were not sufficient to restore completely flight-induced changes. Thus in-depth proteomics analysis unraveled the complex and multifactorial remodeling of skeletal muscle structure and function during long-term space flight, which should help define combined sets of countermeasures before, during, and after the flight.
Collapse
Affiliation(s)
- Georg Tascher
- Université de Strasbourg, CNRS, IPHC UMR 7178, F-670000 Strasbourg, France.,Centre National d'Etudes Spatiales, CNES , 75039 Paris, France
| | - Thomas Brioche
- Université de Montpellier, INRA, UMR 866 Dynamique Musculaire et Métabolisme, Montpellier F-34060, France
| | - Pauline Maes
- Université de Strasbourg, CNRS, IPHC UMR 7178, F-670000 Strasbourg, France
| | - Angèle Chopard
- Université de Montpellier, INRA, UMR 866 Dynamique Musculaire et Métabolisme, Montpellier F-34060, France
| | - Donal O'Gorman
- National Institute for Cellular Biotechnology and the School of Health and Human Performance, Dublin City University , Dublin 9, Ireland
| | | | - Stéphane Blanc
- Université de Strasbourg, CNRS, IPHC UMR 7178, F-670000 Strasbourg, France
| | - Fabrice Bertile
- Université de Strasbourg, CNRS, IPHC UMR 7178, F-670000 Strasbourg, France
| |
Collapse
|
328
|
Lang F, Aravamudhan S, Nolte H, Türk C, Hölper S, Müller S, Günther S, Blaauw B, Braun T, Krüger M. Dynamic changes in the mouse skeletal muscle proteome during denervation-induced atrophy. Dis Model Mech 2017; 10:881-896. [PMID: 28546288 PMCID: PMC5536905 DOI: 10.1242/dmm.028910] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 05/16/2017] [Indexed: 01/07/2023] Open
Abstract
Loss of neuronal stimulation enhances protein breakdown and reduces protein synthesis, causing rapid loss of muscle mass. To elucidate the pathophysiological adaptations that occur in atrophying muscles, we used stable isotope labelling and mass spectrometry to quantify protein expression changes accurately during denervation-induced atrophy after sciatic nerve section in the mouse gastrocnemius muscle. Additionally, mice were fed a stable isotope labelling of amino acids in cell culture (SILAC) diet containing 13C6-lysine for 4, 7 or 11 days to calculate relative levels of protein synthesis in denervated and control muscles. Ubiquitin remnant peptides (K-ε-GG) were profiled by immunoaffinity enrichment to identify potential substrates of the ubiquitin-proteasomal pathway. Of the 4279 skeletal muscle proteins quantified, 850 were differentially expressed significantly within 2 weeks after denervation compared with control muscles. Moreover, pulse labelling identified Lys6 incorporation in 4786 proteins, of which 43 had differential Lys6 incorporation between control and denervated muscle. Enrichment of diglycine remnants identified 2100 endogenous ubiquitination sites and revealed a metabolic and myofibrillar protein diglycine signature, including myosin heavy chains, myomesins and titin, during denervation. Comparative analysis of these proteomic data sets with known atrogenes using a random forest approach identified 92 proteins subject to atrogene-like regulation that have not previously been associated directly with denervation-induced atrophy. Comparison of protein synthesis and proteomic data indicated that upregulation of specific proteins in response to denervation is mainly achieved by protein stabilization. This study provides the first integrated analysis of protein expression, synthesis and ubiquitin signatures during muscular atrophy in a living animal. Summary: Comprehensive proteomic profiling of protein expression, synthesis and ubiquitination during skeletal muscle atrophy reveals that complex regulatory networks are activated during muscle wasting.
Collapse
Affiliation(s)
- Franziska Lang
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany
| | - Sriram Aravamudhan
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Hendrik Nolte
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany
| | - Clara Türk
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany
| | - Soraya Hölper
- Institute of Biochemistry II, Goethe University Medical School, 60590 Frankfurt, Germany
| | - Stefan Müller
- Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Stefan Günther
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Bert Blaauw
- Venetian Institute of Molecular Medicine (VIMM), Department of Biomedical Sciences Padova, University of Padova, 35137 Padova, Italy
| | - Thomas Braun
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Marcus Krüger
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany .,Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
329
|
Amirouche A, Jahnke VE, Lunde JA, Koulmann N, Freyssenet DG, Jasmin BJ. Muscle-specific microRNA-206 targets multiple components in dystrophic skeletal muscle representing beneficial adaptations. Am J Physiol Cell Physiol 2017; 312:C209-C221. [DOI: 10.1152/ajpcell.00185.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 12/06/2016] [Accepted: 12/21/2016] [Indexed: 01/31/2023]
Abstract
Over the last several years, converging lines of evidence have indicated that miR-206 plays a pivotal role in promoting muscle differentiation and regeneration, thereby potentially impacting positively on the progression of neuromuscular disorders, including Duchenne muscular dystrophy (DMD). Despite several studies showing the regulatory function of miR-206 on target mRNAs in skeletal muscle cells, the effects of overexpression of miR-206 in dystrophic muscles remain to be established. Here, we found that miR-206 overexpression in mdx mouse muscles simultaneously targets multiple mRNAs and proteins implicated in satellite cell differentiation, muscle regeneration, and at the neuromuscular junction. Overexpression of miR-206 also increased the levels of several muscle-specific mRNAs/proteins, while enhancing utrophin A expression at the sarcolemma. Finally, we also observed that the increased expression of miR-206 in dystrophin-deficient mouse muscle decreased the production of proinflammatory cytokines and infiltration of macrophages. Taken together, our results show that miR-206 acts as a pleiotropic regulator that targets multiple key mRNAs and proteins expected to provide beneficial adaptations in dystrophic muscle, thus highlighting its therapeutic potential for DMD.
Collapse
Affiliation(s)
- Adel Amirouche
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université de Saint Etienne, Université de Lyon, Lyon, France
- Department of Cellular and Molecular Medicine and Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada; and
| | - Vanessa E. Jahnke
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université de Saint Etienne, Université de Lyon, Lyon, France
| | - John A. Lunde
- Department of Cellular and Molecular Medicine and Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada; and
| | - Nathalie Koulmann
- Institut de Recherche Biomédicale des Armées, Département Environnements Opérationnels, Bretigny-sur-Orge, France
| | - Damien G. Freyssenet
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université de Saint Etienne, Université de Lyon, Lyon, France
| | - Bernard J. Jasmin
- Department of Cellular and Molecular Medicine and Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada; and
| |
Collapse
|
330
|
Group I Paks Promote Skeletal Myoblast Differentiation In Vivo and In Vitro. Mol Cell Biol 2017; 37:MCB.00222-16. [PMID: 27920252 DOI: 10.1128/mcb.00222-16] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 11/26/2016] [Indexed: 12/15/2022] Open
Abstract
Skeletal myogenesis is regulated by signal transduction, but the factors and mechanisms involved are not well understood. The group I Paks Pak1 and Pak2 are related protein kinases and direct effectors of Cdc42 and Rac1. Group I Paks are ubiquitously expressed and specifically required for myoblast fusion in Drosophila We report that both Pak1 and Pak2 are activated during mammalian myoblast differentiation. One pathway of activation is initiated by N-cadherin ligation and involves the cadherin coreceptor Cdo with its downstream effector, Cdc42. Individual genetic deletion of Pak1 and Pak2 in mice has no overt effect on skeletal muscle development or regeneration. However, combined muscle-specific deletion of Pak1 and Pak2 results in reduced muscle mass and a higher proportion of myofibers with a smaller cross-sectional area. This phenotype is exacerbated after repair to acute injury. Furthermore, primary myoblasts lacking Pak1 and Pak2 display delayed expression of myogenic differentiation markers and myotube formation. These results identify Pak1 and Pak2 as redundant regulators of myoblast differentiation in vitro and in vivo and as components of the promyogenic Ncad/Cdo/Cdc42 signaling pathway.
Collapse
|
331
|
Begue G, Raue U, Jemiolo B, Trappe S. DNA methylation assessment from human slow- and fast-twitch skeletal muscle fibers. J Appl Physiol (1985) 2017; 122:952-967. [PMID: 28057818 DOI: 10.1152/japplphysiol.00867.2016] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/07/2016] [Accepted: 12/30/2016] [Indexed: 11/22/2022] Open
Abstract
A new application of the reduced representation bisulfite sequencing method was developed using low-DNA input to investigate the epigenetic profile of human slow- and fast-twitch skeletal muscle fibers. Successful library construction was completed with as little as 15 ng of DNA, and high-quality sequencing data were obtained with 32 ng of DNA. Analysis identified 143,160 differentially methylated CpG sites across 14,046 genes. In both fiber types, selected genes predominantly expressed in slow or fast fibers were hypomethylated, which was supported by the RNA-sequencing analysis. These are the first fiber type-specific methylation data from human skeletal muscle and provide a unique platform for future research.NEW & NOTEWORTHY This study validates a low-DNA input reduced representation bisulfite sequencing method for human muscle biopsy samples to investigate the methylation patterns at a fiber type-specific level. These are the first fiber type-specific methylation data reported from human skeletal muscle and thus provide initial insight into basal state differences in myosin heavy chain I and IIa muscle fibers among young, healthy men.
Collapse
Affiliation(s)
- Gwénaëlle Begue
- Human Performance Laboratory, Ball State University, Muncie, Indiana
| | - Ulrika Raue
- Human Performance Laboratory, Ball State University, Muncie, Indiana
| | - Bozena Jemiolo
- Human Performance Laboratory, Ball State University, Muncie, Indiana
| | - Scott Trappe
- Human Performance Laboratory, Ball State University, Muncie, Indiana
| |
Collapse
|
332
|
Long non-coding RNAs (lncRNAs) in skeletal and cardiac muscle: potential therapeutic and diagnostic targets? Clin Sci (Lond) 2016; 130:2245-2256. [DOI: 10.1042/cs20160244] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 09/22/2016] [Indexed: 12/20/2022]
Abstract
The recent discovery that thousands of RNAs are transcribed by the cell but are never translated into protein, highlights a significant void in our current understanding of how transcriptional networks regulate cellular function. This is particularly astounding when we consider that over 75% of the human genome is transcribed into RNA, but only approximately 2% of RNA is translated into known proteins. This raises the question as to what function the other so-called ‘non-coding RNAs’ (ncRNAs) are performing in the cell. Over the last decade, an enormous amount of research has identified several classes of ncRNAs, predominantly short ncRNAs (<200 nt) that have been confirmed to have functional significance. Recent advances in sequencing technology and bioinformatics have also allowed for the identification of a novel class of ncRNAs, termed long ncRNA (lncRNA) (>200 nt). Several studies have recently shown that long non-coding RNAs (lncRNAs) are associated with tissue development and disease, particularly in cell types that undergo differentiation such as stem cells, cancer cells and striated muscle (skeletal/cardiac). Therefore, understanding the function of these lncRNAs and designing strategies to detect and manipulate them, may present novel therapeutic and diagnostic opportunities. This review will explore the current literature on lncRNAs in skeletal and cardiac muscle and discuss their recent implication in development and disease. Lastly, we will also explore the possibility of using lncRNAs as therapeutic and diagnostic tools and discuss the opportunities and potential shortcomings to these applications.
Collapse
|
333
|
Mizbani A, Luca E, Rushing EJ, Krützfeldt J. MicroRNA deep sequencing in two adult stem cell populations identifies miR-501 as a novel regulator of myosin heavy chain during muscle regeneration. Development 2016; 143:4137-4148. [PMID: 27707793 PMCID: PMC5117213 DOI: 10.1242/dev.136051] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 09/22/2016] [Indexed: 12/15/2022]
Abstract
MicroRNAs (miRNAs) are important regulators of skeletal muscle regeneration, but the underlying mechanisms are still incompletely understood. Here, comparative miRNA sequencing analysis of myogenic progenitor cells (MPs) and non-myogenic fibroblast-adipocyte progenitors (FAPs) during cardiotoxin (CTX)-induced muscle injury uncovered miR-501 as a novel muscle-specific miRNA. miR-501 is an intronic miRNA and its expression levels in MPs correlated with its host gene, chloride channel, voltage-sensitive 5 (Clcn5). Pharmacological inhibition of miR-501 dramatically blunted the induction of embryonic myosin heavy chain (MYH3) and, to a lesser extent, adult myosin isoforms during muscle regeneration, and promoted small-diameter neofibers. An unbiased target identification approach in primary myoblasts validated gigaxonin as a target of miR-501 that mimicked the effect of miR-501 inhibition on MYH3 expression. In the mdx mouse model, which models a pathological disease state, not only was miR-501 induced in regenerating skeletal muscle, but also its serum levels were increased, which correlated with the disease state of the animals. Our results suggest that miR-501 plays a key role in adult muscle regeneration and might serve as a novel serum biomarker for the activation of adult muscle stem cells. Summary: MicroRNA 501 is a novel muscle-specific microRNA that is induced during muscle regeneration and regulates the transition of myosin heavy chains during early myogenesis.
Collapse
Affiliation(s)
- Amir Mizbani
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, Zurich 8091, Switzerland.,Competence Center Personalized Medicine UZH/ETH, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Edlira Luca
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, Zurich 8091, Switzerland
| | - Elisabeth J Rushing
- Institute of Neuropathology, University Zurich and University Hospital Zurich, Zurich 8091, Switzerland
| | - Jan Krützfeldt
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, Zurich 8091, Switzerland .,Competence Center Personalized Medicine UZH/ETH, ETH Zurich and University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
334
|
Coenen-Stass AML, Betts CA, Lee YF, Mäger I, Turunen MP, El Andaloussi S, Morgan JE, Wood MJA, Roberts TC. Selective release of muscle-specific, extracellular microRNAs during myogenic differentiation. Hum Mol Genet 2016; 25:3960-3974. [PMID: 27466195 PMCID: PMC5291232 DOI: 10.1093/hmg/ddw237] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 05/11/2016] [Accepted: 07/11/2016] [Indexed: 01/06/2023] Open
Abstract
MyomiRs are muscle-specific microRNAs (miRNAs) that regulate myoblast proliferation and differentiation. Extracellular myomiRs (ex-myomiRs) are highly enriched in the serum of Duchenne Muscular Dystrophy (DMD) patients and dystrophic mouse models and consequently have potential as disease biomarkers. The biological significance of miRNAs present in the extracellular space is not currently well understood. Here we demonstrate that ex-myomiR levels are elevated in perinatal muscle development, during the regenerative phase that follows exercise-induced myoinjury, and concomitant with myoblast differentiation in culture. Whereas ex-myomiRs are progressively and specifically released by differentiating human primary myoblasts and C2C12 cultures, chemical induction of apoptosis in C2C12 cells results in indiscriminate miRNA release. The selective release of myomiRs as a consequence of cellular differentiation argues against the idea that they are solely waste products of muscle breakdown, and suggests they may serve a biological function in specific physiological contexts. Ex-myomiRs in culture supernatant and serum are predominantly non-vesicular, and their release is independent of ceramide-mediated vesicle secretion. Furthermore, ex-myomiRs levels are reduced in aged dystrophic mice, likely as a consequence of chronic muscle wasting. In conclusion, we show that myomiR release accompanies periods of myogenic differentiation in cell culture and in vivo. Serum myomiR abundance is therefore a function of the regenerative/degenerative status of the muscle, overall muscle mass, and tissue expression levels. These findings have implications for the use of ex-myomiRs as biomarkers for DMD disease progression and monitoring response to therapy.
Collapse
Affiliation(s)
- Anna M L Coenen-Stass
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Corinne A Betts
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Yi F Lee
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm SE-141 57, Sweden
| | - Imre Mäger
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia
| | - Mikko P Turunen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute, University of Eastern Finland, 70150 Kuopio, Finland
| | - Samir El Andaloussi
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm SE-141 57, Sweden
| | - Jennifer E Morgan
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Thomas C Roberts
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Sanford Burnham Prebys Medical Discovery Institute, Development, Aging and Regeneration Program, La Jolla, CA 92037, USA
| |
Collapse
|
335
|
Naldaiz-Gastesi N, Goicoechea M, Alonso-Martín S, Aiastui A, López-Mayorga M, García-Belda P, Lacalle J, San José C, Araúzo-Bravo MJ, Trouilh L, Anton-Leberre V, Herrero D, Matheu A, Bernad A, García-Verdugo JM, Carvajal JJ, Relaix F, Lopez de Munain A, García-Parra P, Izeta A. Identification and Characterization of the Dermal Panniculus Carnosus Muscle Stem Cells. Stem Cell Reports 2016; 7:411-424. [PMID: 27594590 PMCID: PMC5032673 DOI: 10.1016/j.stemcr.2016.08.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 08/01/2016] [Accepted: 08/01/2016] [Indexed: 01/05/2023] Open
Abstract
The dermal Panniculus carnosus (PC) muscle is important for wound contraction in lower mammals and represents an interesting model of muscle regeneration due to its high cell turnover. The resident satellite cells (the bona fide muscle stem cells) remain poorly characterized. Here we analyzed PC satellite cells with regard to developmental origin and purported function. Lineage tracing shows that they originate in Myf5+, Pax3/Pax7+ cell populations. Skin and muscle wounding increased PC myofiber turnover, with the satellite cell progeny being involved in muscle regeneration but with no detectable contribution to the wound-bed myofibroblasts. Since hematopoietic stem cells fuse to PC myofibers in the absence of injury, we also studied the contribution of bone marrow-derived cells to the PC satellite cell compartment, demonstrating that cells of donor origin are capable of repopulating the PC muscle stem cell niche after irradiation and bone marrow transplantation but may not fully acquire the relevant myogenic commitment. PC satellite cells originate from Myf5+, Pax3/Pax7+ cell lineages Skin and muscle wounding increase PC myofiber turnover Donor bone marrow cells repopulate the PC satellite niche after BMT Dermis-derived myogenesis originates from the PC satellite cell population
Collapse
Affiliation(s)
- Neia Naldaiz-Gastesi
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastián 20014, Spain; Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain
| | - María Goicoechea
- Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Sonia Alonso-Martín
- INSERM U955-E10, Université Paris Est, Faculté de Médicine, IMRB U955-E10, Creteil 94000, France
| | - Ana Aiastui
- Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Macarena López-Mayorga
- Molecular Embryology Team, Centro Andaluz de Biología del Desarrollo, Sevilla 41013, Spain
| | - Paula García-Belda
- CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain; Laboratorio de Neurobiología Comparada, Instituto Cavanilles, Universidad de Valencia, Valencia 46980, Spain
| | - Jaione Lacalle
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastián 20014, Spain; Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; Faculty of Medicine and Nursing, UPV-EHU, San Sebastián 20014, Spain
| | - Carlos San José
- Animal Facility and Experimental Surgery, Instituto Biodonostia, San Sebastián 20014, Spain
| | - Marcos J Araúzo-Bravo
- Computational Biology and Systems Biomedicine, Instituto Biodonostia, San Sebastián 20014, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao 48013, Spain
| | - Lidwine Trouilh
- INSA, UPS, INP, LISBP, Université de Toulouse, 31077 Toulouse, France; INRA, UMR792, Ingénierie des Systèmes Biologiques et des Procédés, 31400 Toulouse, France; CNRS, UMR5504, 31400 Toulouse, France
| | - Véronique Anton-Leberre
- INSA, UPS, INP, LISBP, Université de Toulouse, 31077 Toulouse, France; INRA, UMR792, Ingénierie des Systèmes Biologiques et des Procédés, 31400 Toulouse, France; CNRS, UMR5504, 31400 Toulouse, France
| | - Diego Herrero
- Immunology and Oncology Department, Spanish National Center for Biotechnology (CNB-CSIC), Madrid 28049, Spain
| | - Ander Matheu
- IKERBASQUE, Basque Foundation for Science, Bilbao 48013, Spain; Cellular Oncology Group, Oncology Area, Instituto Biodonostia, San Sebastián 20014, Spain
| | - Antonio Bernad
- Immunology and Oncology Department, Spanish National Center for Biotechnology (CNB-CSIC), Madrid 28049, Spain
| | - José Manuel García-Verdugo
- CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain; Laboratorio de Neurobiología Comparada, Instituto Cavanilles, Universidad de Valencia, Valencia 46980, Spain
| | - Jaime J Carvajal
- Molecular Embryology Team, Centro Andaluz de Biología del Desarrollo, Sevilla 41013, Spain
| | - Frédéric Relaix
- INSERM U955-E10, Université Paris Est, Faculté de Médicine, IMRB U955-E10, Creteil 94000, France
| | - Adolfo Lopez de Munain
- Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain; Faculty of Medicine and Nursing, Department of Neurosciences, UPV-EHU, San Sebastián 20014, Spain; Department of Neurology, Hospital Universitario Donostia, San Sebastián 20014, Spain
| | - Patricia García-Parra
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastián 20014, Spain; Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain.
| | - Ander Izeta
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastián 20014, Spain; Department of Biomedical Engineering, School of Engineering, Tecnun-University of Navarra, San Sebastián 20009, Spain.
| |
Collapse
|
336
|
Schiaffino S, Pereira MG, Ciciliot S, Rovere-Querini P. Regulatory T cells and skeletal muscle regeneration. FEBS J 2016; 284:517-524. [DOI: 10.1111/febs.13827] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 07/29/2016] [Indexed: 12/20/2022]
Affiliation(s)
| | - Marcelo G. Pereira
- Venetian Institute of Molecular Medicine (VIMM); Padova Italy
- Department of Biomedical Sciences; University of Padova; Italy
| | - Stefano Ciciliot
- Venetian Institute of Molecular Medicine (VIMM); Padova Italy
- Department of Medicine (DIMED); University of Padova; Italy
| | - Patrizia Rovere-Querini
- Division of Immunology, Transplantation and Infectious Disease; San Raffaele Scientific Institute; Vita-Salute San Raffaele University; Milano Italy
| |
Collapse
|
337
|
Rui Y, Pan F, Mi J. Composition of Muscle Fiber Types in Rat Rotator Cuff Muscles. Anat Rec (Hoboken) 2016; 299:1397-401. [PMID: 27314819 DOI: 10.1002/ar.23384] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 04/20/2016] [Accepted: 04/21/2016] [Indexed: 12/26/2022]
Abstract
The rat is a suitable model to study human rotator cuff pathology owing to the similarities in morphological anatomy structure. However, few studies have reported the composition muscle fiber types of rotator cuff muscles in the rat. In this study, the myosin heavy chain (MyHC) isoforms were stained by immunofluorescence to show the muscle fiber types composition and distribution in rotator cuff muscles of the rat. It was found that rotator cuff muscles in the rat were of mixed fiber type composition. The majority of rotator cuff fibers labeled positively for MyHCII. Moreover, the rat rotator cuff muscles contained hybrid fibers. So, compared with human rotator cuff muscles composed partly of slow-twitch fibers, the majority of fast-twitch fibers in rat rotator cuff muscles should be considered when the rat model study focus on the pathological process of rotator cuff muscles after injury. Gaining greater insight into muscle fiber types in rotator cuff muscles of the rat may contribute to elucidate the mechanism of pathological change in rotator cuff muscles-related diseases. Anat Rec, 299:1397-1401, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yongjun Rui
- Department of Hand Surgery, Wuxi Number 9 People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu Province 214062, the People's Republic of China.
| | | | - Jingyi Mi
- Department of Hand Surgery, Wuxi Number 9 People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu Province 214062, the People's Republic of China
| |
Collapse
|
338
|
Gurevich DB, Nguyen PD, Siegel AL, Ehrlich OV, Sonntag C, Phan JMN, Berger S, Ratnayake D, Hersey L, Berger J, Verkade H, Hall TE, Currie PD. Asymmetric division of clonal muscle stem cells coordinates muscle regeneration in vivo. Science 2016; 353:aad9969. [PMID: 27198673 DOI: 10.1126/science.aad9969] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 05/10/2016] [Indexed: 12/11/2022]
Abstract
Skeletal muscle is an example of a tissue that deploys a self-renewing stem cell, the satellite cell, to effect regeneration. Recent in vitro studies have highlighted a role for asymmetric divisions in renewing rare "immortal" stem cells and generating a clonal population of differentiation-competent myoblasts. However, this model currently lacks in vivo validation. We define a zebrafish muscle stem cell population analogous to the mammalian satellite cell and image the entire process of muscle regeneration from injury to fiber replacement in vivo. This analysis reveals complex interactions between satellite cells and both injured and uninjured fibers and provides in vivo evidence for the asymmetric division of satellite cells driving both self-renewal and regeneration via a clonally restricted progenitor pool.
Collapse
Affiliation(s)
- David B Gurevich
- Australian Regenerative Medicine Institute, Level 1, 15 Innovation Walk, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| | - Phong Dang Nguyen
- Australian Regenerative Medicine Institute, Level 1, 15 Innovation Walk, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| | - Ashley L Siegel
- Australian Regenerative Medicine Institute, Level 1, 15 Innovation Walk, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| | - Ophelia V Ehrlich
- Australian Regenerative Medicine Institute, Level 1, 15 Innovation Walk, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| | - Carmen Sonntag
- Australian Regenerative Medicine Institute, Level 1, 15 Innovation Walk, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| | - Jennifer M N Phan
- Australian Regenerative Medicine Institute, Level 1, 15 Innovation Walk, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| | - Silke Berger
- Australian Regenerative Medicine Institute, Level 1, 15 Innovation Walk, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| | - Dhanushika Ratnayake
- Australian Regenerative Medicine Institute, Level 1, 15 Innovation Walk, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| | - Lucy Hersey
- Australian Regenerative Medicine Institute, Level 1, 15 Innovation Walk, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| | - Joachim Berger
- Australian Regenerative Medicine Institute, Level 1, 15 Innovation Walk, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| | - Heather Verkade
- School of Biological Sciences, Building 18, Monash University, Clayton, Victoria 3800, Australia
| | - Thomas E Hall
- Australian Regenerative Medicine Institute, Level 1, 15 Innovation Walk, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Level 1, 15 Innovation Walk, Monash University, Wellington Road, Clayton, Victoria 3800, Australia. European Molecular Biology Laboratory Australia Melbourne Node, Level 1, Building 75, Monash University, Wellington Road, Clayton, Victoria 3800, Australia.
| |
Collapse
|
339
|
Cong X, Doering J, Mazala DAG, Chin ER, Grange RW, Jiang H. The SH3 and cysteine-rich domain 3 (Stac3) gene is important to growth, fiber composition, and calcium release from the sarcoplasmic reticulum in postnatal skeletal muscle. Skelet Muscle 2016; 6:17. [PMID: 27073615 PMCID: PMC4828897 DOI: 10.1186/s13395-016-0088-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 03/30/2016] [Indexed: 12/25/2022] Open
Abstract
Background The SH3 and cysteine-rich domain 3 (Stac3) gene is specifically expressed in the skeletal muscle. Stac3 knockout mice die perinatally. In this study, we determined the potential role of Stac3 in postnatal skeletal muscle growth, fiber composition, and contraction by generating conditional Stac3 knockout mice. Methods We disrupted the Stac3 gene in 4-week-old male mice using the Flp-FRT and tamoxifen-inducible Cre-loxP systems. Results RT-qPCR and western blotting analyses of the limb muscles of target mice indicated that nearly all Stac3 mRNA and more than 70 % of STAC3 protein were deleted 4 weeks after tamoxifen injection. Postnatal Stac3 deletion inhibited body and limb muscle mass gains. Histological staining and gene expression analyses revealed that postnatal Stac3 deletion decreased the size of myofibers and increased the percentage of myofibers containing centralized nuclei, with no effect on the total myofiber number. Grip strength and grip time tests indicated that postnatal Stac3 deletion decreased limb muscle strength in mice. Muscle contractile tests revealed that postnatal Stac3 deletion reduced electrostimulation-induced but not the ryanodine receptor agonist caffeine-induced maximal force output in the limb muscles. Calcium imaging analysis of single flexor digitorum brevis myofibers indicated that postnatal Stac3 deletion reduced electrostimulation- but not caffeine-induced calcium release from the sarcoplasmic reticulum. Conclusions This study demonstrates that STAC3 is important to myofiber hypertrophy, myofiber-type composition, contraction, and excitation-induced calcium release from the sarcoplasmic reticulum in the postnatal skeletal muscle. Electronic supplementary material The online version of this article (doi:10.1186/s13395-016-0088-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaofei Cong
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA USA
| | - Jonathan Doering
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA USA
| | - Davi A G Mazala
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, MD USA
| | - Eva R Chin
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, MD USA
| | - Robert W Grange
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA USA
| | - Honglin Jiang
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA USA
| |
Collapse
|
340
|
Janghra N, Morgan JE, Sewry CA, Wilson FX, Davies KE, Muntoni F, Tinsley J. Correlation of Utrophin Levels with the Dystrophin Protein Complex and Muscle Fibre Regeneration in Duchenne and Becker Muscular Dystrophy Muscle Biopsies. PLoS One 2016; 11:e0150818. [PMID: 26974331 PMCID: PMC4790853 DOI: 10.1371/journal.pone.0150818] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 02/19/2016] [Indexed: 01/16/2023] Open
Abstract
Duchenne muscular dystrophy is a severe and currently incurable progressive neuromuscular condition, caused by mutations in the DMD gene that result in the inability to produce dystrophin. Lack of dystrophin leads to loss of muscle fibres and a reduction in muscle mass and function. There is evidence from dystrophin-deficient mouse models that increasing levels of utrophin at the muscle fibre sarcolemma by genetic or pharmacological means significantly reduces the muscular dystrophy pathology. In order to determine the efficacy of utrophin modulators in clinical trials, it is necessary to accurately measure utrophin levels and other biomarkers on a fibre by fibre basis within a biopsy section. Our aim was to develop robust and reproducible staining and imaging protocols to quantify sarcolemmal utrophin levels, sarcolemmal dystrophin complex members and numbers of regenerating fibres within a biopsy section. We quantified sarcolemmal utrophin in mature and regenerating fibres and the percentage of regenerating muscle fibres, in muscle biopsies from Duchenne, the milder Becker muscular dystrophy and controls. Fluorescent immunostaining followed by image analysis was performed to quantify utrophin intensity and β-dystrogylcan and ɣ –sarcoglycan intensity at the sarcolemma. Antibodies to fetal and developmental myosins were used to identify regenerating muscle fibres allowing the accurate calculation of percentage regeneration fibres in the biopsy. Our results indicate that muscle biopsies from Becker muscular dystrophy patients have fewer numbers of regenerating fibres and reduced utrophin intensity compared to muscle biopsies from Duchenne muscular dystrophy patients. Of particular interest, we show for the first time that the percentage of regenerating muscle fibres within the muscle biopsy correlate with the clinical severity of Becker and Duchenne muscular dystrophy patients from whom the biopsy was taken. The ongoing development of these tools to quantify sarcolemmal utrophin and muscle regeneration in muscle biopsies will be invaluable for assessing utrophin modulator activity in future clinical trials.
Collapse
Affiliation(s)
- Narinder Janghra
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Institute of Child Health, 30 Guilford Street, London, WC1N1EH, United Kingdom
| | - Jennifer E. Morgan
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Institute of Child Health, 30 Guilford Street, London, WC1N1EH, United Kingdom
- * E-mail:
| | - Caroline A. Sewry
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Institute of Child Health, 30 Guilford Street, London, WC1N1EH, United Kingdom
| | - Francis X. Wilson
- Summit Therapeutics plc, 85b Park Drive, Milton Park, Abingdon, Oxfordshire, OX14 4RY, United Kingdom
| | - Kay E. Davies
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, United Kingdom
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Institute of Child Health, 30 Guilford Street, London, WC1N1EH, United Kingdom
| | - Jonathon Tinsley
- Summit Therapeutics plc, 85b Park Drive, Milton Park, Abingdon, Oxfordshire, OX14 4RY, United Kingdom
| |
Collapse
|
341
|
Walklate J, Vera C, Bloemink MJ, Geeves MA, Leinwand L. The Most Prevalent Freeman-Sheldon Syndrome Mutations in the Embryonic Myosin Motor Share Functional Defects. J Biol Chem 2016; 291:10318-31. [PMID: 26945064 PMCID: PMC4858979 DOI: 10.1074/jbc.m115.707489] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Indexed: 11/12/2022] Open
Abstract
The embryonic myosin isoform is expressed during fetal development and rapidly down-regulated after birth. Freeman-Sheldon syndrome (FSS) is a disease associated with missense mutations in the motor domain of this myosin. It is the most severe form of distal arthrogryposis, leading to overcontraction of the hands, feet, and orofacial muscles and other joints of the body. Availability of human embryonic muscle tissue has been a limiting factor in investigating the properties of this isoform and its mutations. Using a recombinant expression system, we have studied homogeneous samples of human motors for the WT and three of the most common FSS mutants: R672H, R672C, and T178I. Our data suggest that the WT embryonic myosin motor is similar in contractile speed to the slow type I/β cardiac based on the rate constant for ADP release and ADP affinity for actin-myosin. All three FSS mutations show dramatic changes in kinetic properties, most notably the slowing of the apparent ATP hydrolysis step (reduced 5–9-fold), leading to a longer lived detached state and a slowed Vmax of the ATPase (2–35-fold), indicating a slower cycling time. These mutations therefore seriously disrupt myosin function.
Collapse
Affiliation(s)
- Jonathan Walklate
- From the School of Biosciences, University of Kent, Canterbury CT2 7NJ, United Kingdom and
| | - Carlos Vera
- the Department of Molecular and Developmental Biology, University of Colorado, Boulder, Colorado 80309
| | - Marieke J Bloemink
- From the School of Biosciences, University of Kent, Canterbury CT2 7NJ, United Kingdom and
| | - Michael A Geeves
- From the School of Biosciences, University of Kent, Canterbury CT2 7NJ, United Kingdom and
| | - Leslie Leinwand
- the Department of Molecular and Developmental Biology, University of Colorado, Boulder, Colorado 80309
| |
Collapse
|
342
|
Lohmeier-Vogel EM, Heeley DH. Biochemical Comparison of Tpm1.1 (α) and Tpm2.2 (β) Tropomyosins from Rabbit Skeletal Muscle. Biochemistry 2016; 55:1418-27. [DOI: 10.1021/acs.biochem.5b01140] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Elke M. Lohmeier-Vogel
- Department
of Biological Sciences, University of Calgary, Calgary, Alberta T2N1N4, Canada
| | - David H. Heeley
- Department
of Biochemistry, Memorial University of Newfoundland, St. John’s, Newfoundland A1B 3X9, Canada
| |
Collapse
|
343
|
Beedle AM. Distribution of myosin heavy chain isoforms in muscular dystrophy: insights into disease pathology. MUSCULOSKELETAL REGENERATION 2016; 2:e1365. [PMID: 27430020 PMCID: PMC4943764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Myosin heavy chain isoforms are an important component defining fiber type specific properties in skeletal muscle, such as oxidative versus glycolytic metabolism, rate of contraction, and fatigability. While the molecular mechanisms that underlie specification of the different fiber types are becoming clearer, how this programming becomes disrupted in muscular dystrophy and the functional consequences of fiber type changes in disease are not fully resolved. Fiber type changes in disease, with specific focus on muscular dystrophies caused by defects in the dystrophin glycoprotein complex, are discussed.
Collapse
Affiliation(s)
- Aaron M Beedle
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia College of Pharmacy, Athens, GA 30602 USA
| |
Collapse
|