1
|
Zhang RX, Zhai YY, Ding RR, Huang JH, Shi XC, Liu H, Liu XP, Zhang JF, Lu JF, Zhang Z, Leng XK, Li DF, Xiao JY, Xia B, Wu JW. FNDC1 is a myokine that promotes myogenesis and muscle regeneration. EMBO J 2025; 44:30-53. [PMID: 39567831 PMCID: PMC11695938 DOI: 10.1038/s44318-024-00285-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 10/04/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024] Open
Abstract
Myogenesis is essential for skeletal muscle formation and regeneration after injury, yet its regulators are largely unknown. Here we identified fibronectin type III domain containing 1 (FNDC1) as a previously uncharacterized myokine. In vitro studies showed that knockdown of Fndc1 in myoblasts reduces myotube formation, while overexpression of Fndc1 promotes myogenic differentiation. We further generated recombinant truncated mouse FNDC1 (mFNDC1), which retains reliable activity in promoting myoblast differentiation in vitro. Gain- and loss-of-function studies collectively showed that FNDC1 promotes cardiotoxin (CTX)-induced muscle regeneration in adult mice. Furthermore, recombinant FNDC1 treatment ameliorated pathological muscle phenotypes in the mdx mouse model of Duchenne muscular dystrophy. Mechanistically, FNDC1 bound to the integrin α5β1 and activated the downstream FAK/PI3K/AKT/mTOR pathway to promote myogenic differentiation. Pharmacological inhibition of integrin α5β1 or of the downstream FAK/PI3K/AKT/mTOR pathway abolished the pro-myogenic effect of FNDC1. Collectively, these results suggested that myokine FNDC1 might be used as a therapeutic agent to regulate myogenic differentiation and muscle regeneration for the treatment of acute and chronic muscle disease.
Collapse
Affiliation(s)
- Rui Xin Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yuan Yuan Zhai
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Rong Rong Ding
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jia He Huang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xiao Chen Shi
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Huan Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xiao Peng Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jian Feng Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jun Feng Lu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Zhe Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xiang Kai Leng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - De Fu Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jun Ying Xiao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Bo Xia
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jiang Wei Wu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
2
|
Zapater I Morales C, Carman PJ, Soffar DB, Windner SE, Dominguez R, Baylies MK. Drosophila Tropomodulin is required for multiple actin-dependent processes within developing myofibers. Development 2023; 150:dev201194. [PMID: 36806912 PMCID: PMC10112908 DOI: 10.1242/dev.201194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 02/09/2023] [Indexed: 02/22/2023]
Abstract
Proper muscle contraction requires the assembly and maintenance of sarcomeres and myofibrils. Although the protein components of myofibrils are generally known, less is known about the mechanisms by which they individually function and together synergize for myofibril assembly and maintenance. For example, it is unclear how the disruption of actin filament (F-actin) regulatory proteins leads to the muscle weakness observed in myopathies. Here, we show that knockdown of Drosophila Tropomodulin (Tmod), results in several myopathy-related phenotypes, including reduction of muscle cell (myofiber) size, increased sarcomere length, disorganization and misorientation of myofibrils, ectopic F-actin accumulation, loss of tension-mediating proteins at the myotendinous junction, and misshaped and internalized nuclei. Our findings support and extend the tension-driven self-organizing myofibrillogenesis model. We show that, like its mammalian counterpart, Drosophila Tmod caps F-actin pointed-ends, and we propose that this activity is crucial for cellular processes in different locations within the myofiber that directly and indirectly contribute to the maintenance of muscle function. Our findings provide significant insights to the role of Tmod in muscle development, maintenance and disease.
Collapse
Affiliation(s)
- Carolina Zapater I Morales
- Biochemistry, Cell & Developmental Biology, and Molecular Biology (BCMB) program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering, Cancer Center, New York, NY 10065, USA
| | - Peter J Carman
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David B Soffar
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering, Cancer Center, New York, NY 10065, USA
| | - Stefanie E Windner
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering, Cancer Center, New York, NY 10065, USA
| | - Roberto Dominguez
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mary K Baylies
- Biochemistry, Cell & Developmental Biology, and Molecular Biology (BCMB) program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering, Cancer Center, New York, NY 10065, USA
| |
Collapse
|
3
|
Shi H, Wang C, Gao BZ, Henderson JH, Ma Z. Cooperation between myofibril growth and costamere maturation in human cardiomyocytes. Front Bioeng Biotechnol 2022; 10:1049523. [PMID: 36394013 PMCID: PMC9663467 DOI: 10.3389/fbioe.2022.1049523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 10/19/2022] [Indexed: 12/14/2022] Open
Abstract
Costameres, as striated muscle-specific cell adhesions, anchor both M-lines and Z-lines of the sarcomeres to the extracellular matrix. Previous studies have demonstrated that costameres intimately participate in the initial assembly of myofibrils. However, how costamere maturation cooperates with myofibril growth is still underexplored. In this work, we analyzed zyxin (costameres), α-actinin (Z-lines) and myomesin (M-lines) to track the behaviors of costameres and myofibrils within the cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs). We quantified the assembly and maturation of costameres associated with the process of myofibril growth within the hiPSC-CMs in a time-dependent manner. We found that asynchrony existed not only between the maturation of myofibrils and costameres, but also between the formation of Z-costameres and M-costameres that associated with different structural components of the sarcomeres. This study helps us gain more understanding of how costameres assemble and incorporate into the cardiomyocyte sarcomeres, which sheds a light on cardiomyocyte mechanobiology.
Collapse
Affiliation(s)
- Huaiyu Shi
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, United States,BioInspired Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, United States
| | - Chenyan Wang
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, United States,BioInspired Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, United States
| | - Bruce Z. Gao
- Department of Bioengineering, Clemson University, Clemson, SC, United States
| | - James H. Henderson
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, United States,BioInspired Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, United States
| | - Zhen Ma
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, United States,BioInspired Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, United States,*Correspondence: Zhen Ma,
| |
Collapse
|
4
|
Abstract
An ensemble of in vitro cardiac tissue models has been developed over the past several decades to aid our understanding of complex cardiovascular disorders using a reductionist approach. These approaches often rely on recapitulating single or multiple clinically relevant end points in a dish indicative of the cardiac pathophysiology. The possibility to generate disease-relevant and patient-specific human induced pluripotent stem cells has further leveraged the utility of the cardiac models as screening tools at a large scale. To elucidate biological mechanisms in the cardiac models, it is critical to integrate physiological cues in form of biochemical, biophysical, and electromechanical stimuli to achieve desired tissue-like maturity for a robust phenotyping. Here, we review the latest advances in the directed stem cell differentiation approaches to derive a wide gamut of cardiovascular cell types, to allow customization in cardiac model systems, and to study diseased states in multiple cell types. We also highlight the recent progress in the development of several cardiovascular models, such as cardiac organoids, microtissues, engineered heart tissues, and microphysiological systems. We further expand our discussion on defining the context of use for the selection of currently available cardiac tissue models. Last, we discuss the limitations and challenges with the current state-of-the-art cardiac models and highlight future directions.
Collapse
Affiliation(s)
- Dilip Thomas
- Stanford Cardiovascular Institute
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Suji Choi
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston MA 02134
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115
| | - Christina Alamana
- Stanford Cardiovascular Institute
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Kevin K. Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston MA 02134
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
| | - Joseph C. Wu
- Stanford Cardiovascular Institute
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
5
|
Shi H, Wu X, Sun S, Wang C, Vangelatos Z, Ash-Shakoor A, Grigoropoulos CP, Mather PT, Henderson JH, Ma Z. Profiling the responsiveness of focal adhesions of human cardiomyocytes to extracellular dynamic nano-topography. Bioact Mater 2022; 10:367-377. [PMID: 34901553 PMCID: PMC8636819 DOI: 10.1016/j.bioactmat.2021.08.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/17/2021] [Accepted: 08/25/2021] [Indexed: 01/02/2023] Open
Abstract
Focal adhesion complexes function as the mediators of cell-extracellular matrix interactions to sense and transmit the extracellular signals. Previous studies have demonstrated that cardiomyocyte focal adhesions can be modulated by surface topographic features. However, the response of focal adhesions to dynamic surface topographic changes remains underexplored. To study this dynamic responsiveness of focal adhesions, we utilized a shape memory polymer-based substrate that can produce a flat-to-wrinkle surface transition triggered by an increase of temperature. Using this dynamic culture system, we analyzed three proteins (paxillin, vinculin and zyxin) from different layers of the focal adhesion complex in response to dynamic extracellular topographic change. Hence, we quantified the dynamic profile of cardiomyocyte focal adhesion in a time-dependent manner, which provides new understanding of dynamic cardiac mechanobiology.
Collapse
Affiliation(s)
- Huaiyu Shi
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Syracuse Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| | - Xiangjun Wu
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Syracuse Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| | - Shiyang Sun
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Syracuse Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| | - Chenyan Wang
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Syracuse Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| | - Zacharias Vangelatos
- Department of Mechanical Engineering, University of California, Berkeley, PA, 94720, USA
| | - Ariel Ash-Shakoor
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
| | | | - Patrick T. Mather
- Department of Chemical Engineering, Bucknell University, Lewisburg, PA, 17837, USA
| | - James H. Henderson
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Syracuse Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| | - Zhen Ma
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Syracuse Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| |
Collapse
|
6
|
Koike TE, Fuziwara CS, Brum PC, Kimura ET, Rando TA, Miyabara EH. Muscle Stem Cell Function Is Impaired in β2-Adrenoceptor Knockout Mice. Stem Cell Rev Rep 2022; 18:2431-2443. [PMID: 35244862 DOI: 10.1007/s12015-022-10334-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2022] [Indexed: 11/30/2022]
Abstract
Knockout (ko) mice for the β2 adrenoceptor (Adrβ2) have impaired skeletal muscle regeneration, suggesting that this receptor is important for muscle stem cell (satellite cell) function. Here, we investigated the role of Adrβ2 in the function of satellite cells from β2ko mice in the context of muscle regeneration, through in vivo and in vitro experiments. Immunohistochemical analysis showed a significant reduction in the number of self-renewed Pax7+ satellite cells, proliferating Pax7+/MyoD+ myogenic precursor cells, and regenerating eMHC+ myofibers in regenerating muscle of β2ko mice at 30, 3, and 10 days post-injury, respectively. Quiescent satellite cells were isolated by fluorescence-activated cell sorting, and cell cycle entry was assessed by EdU incorporation. The results demonstrated a lower number of proliferating Pax7+/EdU+ satellite cells from β2ko mice. There was an increase in the gene expression of the cell cycle inhibitor Cdkn1a and Notch pathway components and the activation of Notch signaling in proliferating myoblasts from β2ko mice. There was a decrease in the number of myogenin-positive nuclei in myofibers maintained in differentiation media, and a lower fusion index in differentiating myoblasts from β2ko mice. Furthermore, the gene expression of Wnt/β-catenin signaling components, the expression of nuclear β-catenin and the activation of Wnt/β-catenin signaling decreased in differentiating myoblasts from β2ko mice. These results indicate that Adrβ2 plays a crucial role in satellite cell self-renewal, as well as in myoblast proliferation and differentiation by regulating Notch and Wnt/β-catenin signaling, respectively.
Collapse
Affiliation(s)
- Tatiana E Koike
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 2415. CEP, São Paulo, SP, 05508-000, Brazil
| | - Cesar S Fuziwara
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Patricia C Brum
- School of Physical Education and Sport, University of São Paulo, São Paulo, SP, Brazil
| | - Edna T Kimura
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Thomas A Rando
- Veterans Affairs Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA, USA.,Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Broad Stem Cell Research Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Elen H Miyabara
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 2415. CEP, São Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
7
|
Roman W, Pinheiro H, Pimentel MR, Segalés J, Oliveira LM, García-Domínguez E, Gómez-Cabrera MC, Serrano AL, Gomes ER, Muñoz-Cánoves P. Muscle repair after physiological damage relies on nuclear migration for cellular reconstruction. Science 2021; 374:355-359. [PMID: 34648328 DOI: 10.1126/science.abe5620] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- William Roman
- Department of Experimental & Health Sciences, University Pompeu Fabra, CIBERNED, 08003 Barcelona, Spain.,Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Helena Pinheiro
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Mafalda R Pimentel
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Jessica Segalés
- Department of Experimental & Health Sciences, University Pompeu Fabra, CIBERNED, 08003 Barcelona, Spain
| | - Luis M Oliveira
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Esther García-Domínguez
- FreshAge Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, Valencia, Spain
| | - Mari Carmen Gómez-Cabrera
- FreshAge Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, Valencia, Spain
| | - Antonio L Serrano
- Department of Experimental & Health Sciences, University Pompeu Fabra, CIBERNED, 08003 Barcelona, Spain
| | - Edgar R Gomes
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Pura Muñoz-Cánoves
- Department of Experimental & Health Sciences, University Pompeu Fabra, CIBERNED, 08003 Barcelona, Spain.,Centro Nacional de Investigaciones Cardiovasculares, 28019 Madrid, Spain.,ICREA, 08010 Barcelona, Spain
| |
Collapse
|
8
|
Hu Z, Cao J, Zhang J, Ge L, Zhang H, Liu X. Skeletal Muscle Transcriptome Analysis of Hanzhong Ma Duck at Different Growth Stages Using RNA-Seq. Biomolecules 2021; 11:315. [PMID: 33669581 PMCID: PMC7927120 DOI: 10.3390/biom11020315] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/12/2021] [Accepted: 02/14/2021] [Indexed: 01/02/2023] Open
Abstract
As one of the most important poultry worldwide, ducks (Anas platyrhynchos) are raised mainly for meat and egg products, and muscle development in ducks is important for meat production. Therefore, an investigation of gene expression in duck skeletal muscle would significantly contribute to our understanding of muscle development. In this study, twenty-four cDNA libraries were constructed from breast and leg muscles of Hanzhong Ma ducks at day 17, 21, 27 of the embryo and postnatal at 6-month-old. High-throughput sequencing and bioinformatics were used to determine the abundances and characteristics of transcripts. A total of 632,172,628 (average 52,681,052) and 637,213,938 (average 53,101,162) reads were obtained from the sequencing data of breast and leg muscles, respectively. Over 71.63% and 77.36% of the reads could be mapped to the Anas platyrhynchos genome. In the skeletal muscle of Hanzhong duck, intron variant (INTRON), synonymous variant (SYNONYMOUS_CODING), and prime 3' UTR variant (UTR_3_PRIME) were the main single nucleotide polymorphisms (SNP) annotation information, and "INTRON", "UTR_3_PRIME", and downstream-gene variant (DOWNSTREAM) were the main insertion-deletion (InDel) annotation information. The predicted number of alternative splicing (AS) in all samples were mainly alternative 5' first exon (transcription start site)-the first exon splicing (TSS) and alternative 3' last exon (transcription terminal site)-the last exon splicing (TTS). Besides, there were 292 to 2801 annotated differentially expressed genes (DEGs) in breast muscle and 304 to 1950 annotated DEGs in leg muscle from different databases. It is worth noting that 75 DEGs in breast muscle and 49 DEGs in leg muscle were co-expressed at all developmental points of comparison, respectively. The RNA-Seq data were confirmed to be reliable by qPCR. The identified DEGs, such as CREBL2, RHEB, GDF6, SHISA2, MYLK2, ACTN3, RYR3, and STMN1, were specially highlighted, indicating their strong associations with muscle development in the Hanzhong Ma duck. KEGG pathway analysis suggested that regulation of actin cytoskeleton, oxidative phosphorylation, and focal adhesion were involved in the development of skeletal muscle. The findings from this study can contribute to future investigations of the growth and development mechanism in duck skeletal muscle.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaolin Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China; (Z.H.); (J.C.); (J.Z.); (L.G.); (H.Z.)
| |
Collapse
|
9
|
Taneja N, Neininger AC, Burnette DT. Coupling to substrate adhesions drives the maturation of muscle stress fibers into myofibrils within cardiomyocytes. Mol Biol Cell 2020; 31:1273-1288. [PMID: 32267210 PMCID: PMC7353145 DOI: 10.1091/mbc.e19-11-0652] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Forces generated by heart muscle contraction must be balanced by adhesion to the extracellular matrix (ECM) and to other cells for proper heart function. Decades of data have suggested that cell-ECM adhesions are important for sarcomere assembly. However, the relationship between cell-ECM adhesions and sarcomeres assembling de novo remains untested. Sarcomeres arise from muscle stress fibers (MSFs) that are translocating on the top (dorsal) surface of cultured cardiomyocytes. Using an array of tools to modulate cell-ECM adhesion, we established a strong positive correlation between the extent of cell-ECM adhesion and sarcomere assembly. On the other hand, we found a strong negative correlation between the extent of cell-ECM adhesion and the rate of MSF translocation, a phenomenon also observed in nonmuscle cells. We further find a conserved network architecture that also exists in nonmuscle cells. Taken together, our results show that cell-ECM adhesions mediate coupling between the substrate and MSFs, allowing their maturation into sarcomere-containing myofibrils.
Collapse
Affiliation(s)
- Nilay Taneja
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232
| | - Abigail C Neininger
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232
| | - Dylan T Burnette
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232
| |
Collapse
|
10
|
Sun S, Shi H, Moore S, Wang C, Ash-Shakoor A, Mather PT, Henderson JH, Ma Z. Progressive Myofibril Reorganization of Human Cardiomyocytes on a Dynamic Nanotopographic Substrate. ACS APPLIED MATERIALS & INTERFACES 2020; 12:21450-21462. [PMID: 32326701 DOI: 10.1021/acsami.0c03464] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Cardiomyocyte (CM) alignment with striated myofibril organization is developed during early cardiac organogenesis. Previous work has successfully achieved in vitro CM alignment using a variety of biomaterial scaffolds and substrates with static topographic features. However, the cellular processes that occur during the response of CMs to dynamic surface topographic changes, which may provide a model of in vivo developmental progress of CM alignment within embryonic myocardium, remains poorly understood. To gain insights into these cellular processes involved in the response of CMs to dynamic topographic changes, we developed a dynamic topographic substrate that employs a shape memory polymer coated with polyelectrolyte multilayers to produce a flat-to-wrinkle surface transition when triggered by a change in incubation temperature. Using this system, we investigated cellular morphological alignment and intracellular myofibril reorganization in response to the dynamic wrinkle formation. Hence, we identified the progressive cellular processes of human-induced pluripotent stem cell-CMs in a time-dependent manner, which could provide a foundation for a mechanistic model of cardiac myofibril reorganization in response to extracellular microenvironment changes.
Collapse
Affiliation(s)
- Shiyang Sun
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Huaiyu Shi
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Sarah Moore
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Chenyan Wang
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Ariel Ash-Shakoor
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Patrick T Mather
- Department of Chemical Engineering, Bucknell University, Lewisburg, Pennsylvania 17837, United States
| | - James H Henderson
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Zhen Ma
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| |
Collapse
|
11
|
Sarkar TJ, Quarta M, Mukherjee S, Colville A, Paine P, Doan L, Tran CM, Chu CR, Horvath S, Qi LS, Bhutani N, Rando TA, Sebastiano V. Transient non-integrative expression of nuclear reprogramming factors promotes multifaceted amelioration of aging in human cells. Nat Commun 2020; 11:1545. [PMID: 32210226 PMCID: PMC7093390 DOI: 10.1038/s41467-020-15174-3] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 02/20/2020] [Indexed: 01/11/2023] Open
Abstract
Aging is characterized by a gradual loss of function occurring at the molecular, cellular, tissue and organismal levels. At the chromatin level, aging associates with progressive accumulation of epigenetic errors that eventually lead to aberrant gene regulation, stem cell exhaustion, senescence, and deregulated cell/tissue homeostasis. Nuclear reprogramming to pluripotency can revert both the age and the identity of any cell to that of an embryonic cell. Recent evidence shows that transient reprogramming can ameliorate age-associated hallmarks and extend lifespan in progeroid mice. However, it is unknown how this form of rejuvenation would apply to naturally aged human cells. Here we show that transient expression of nuclear reprogramming factors, mediated by expression of mRNAs, promotes a rapid and broad amelioration of cellular aging, including resetting of epigenetic clock, reduction of the inflammatory profile in chondrocytes, and restoration of youthful regenerative response to aged, human muscle stem cells, in each case without abolishing cellular identity.
Collapse
Affiliation(s)
- Tapash Jay Sarkar
- 0000000419368956grid.168010.eInstitute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305 USA ,0000000419368956grid.168010.eDepartment of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA 94305 USA ,0000000419368956grid.168010.eDepartment of Applied Physics, Stanford University School of Humanities and Sciences, Stanford, CA 94305 USA
| | - Marco Quarta
- 0000000419368956grid.168010.eDepartment of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305 USA ,0000000419368956grid.168010.ePaul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305 USA ,0000 0004 0419 2556grid.280747.eCenter for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 USA ,0000 0004 0635 6745grid.486808.aMolecular Medicine Research Institute, Sunnyvale, CA 94085 USA
| | - Shravani Mukherjee
- Department of Orthopedic Surgery, Stanford University School of Medicine, Sanford, CA 94305 USA
| | - Alex Colville
- 0000000419368956grid.168010.eDepartment of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305 USA ,0000000419368956grid.168010.ePaul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305 USA ,0000 0004 0419 2556grid.280747.eCenter for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 USA ,0000000419368956grid.168010.eDepartment of Genetics, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Patrick Paine
- 0000000419368956grid.168010.eDepartment of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305 USA ,0000000419368956grid.168010.ePaul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305 USA ,0000 0004 0419 2556grid.280747.eCenter for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 USA ,0000 0004 0635 6745grid.486808.aMolecular Medicine Research Institute, Sunnyvale, CA 94085 USA
| | - Linda Doan
- 0000000419368956grid.168010.eDepartment of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305 USA ,0000000419368956grid.168010.ePaul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305 USA ,0000 0004 0419 2556grid.280747.eCenter for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 USA ,0000 0004 0635 6745grid.486808.aMolecular Medicine Research Institute, Sunnyvale, CA 94085 USA
| | - Christopher M. Tran
- 0000000419368956grid.168010.eDepartment of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305 USA ,0000000419368956grid.168010.ePaul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305 USA ,0000 0004 0419 2556grid.280747.eCenter for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 USA
| | - Constance R. Chu
- Department of Orthopedic Surgery, Stanford University School of Medicine, Sanford, CA 94305 USA ,0000 0004 0419 2556grid.280747.eVA Palo Alto Health Care System, Palo Alto, CA 94304 USA
| | - Steve Horvath
- 0000 0000 9632 6718grid.19006.3eDepartment of Human Genetics David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA ,0000 0000 9632 6718grid.19006.3eDepartment of Biostatistics, Fielding School of Public Health, UCLA, Los Angeles, CA 90095 USA
| | - Lei S. Qi
- 0000000419368956grid.168010.eDepartment of Bioengineering, Department of Chemical and Systems Biology, ChEM-H, Stanford University, Stanford, CA 94305 USA
| | - Nidhi Bhutani
- Department of Orthopedic Surgery, Stanford University School of Medicine, Sanford, CA 94305 USA
| | - Thomas A. Rando
- 0000000419368956grid.168010.eDepartment of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305 USA ,0000000419368956grid.168010.ePaul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305 USA ,0000 0004 0419 2556grid.280747.eCenter for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 USA
| | - Vittorio Sebastiano
- 0000000419368956grid.168010.eInstitute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305 USA ,0000000419368956grid.168010.eDepartment of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA 94305 USA
| |
Collapse
|
12
|
Luca E, Turcekova K, Hartung A, Mathes S, Rehrauer H, Krützfeldt J. Genetic deletion of microRNA biogenesis in muscle cells reveals a hierarchical non-clustered network that controls focal adhesion signaling during muscle regeneration. Mol Metab 2020; 36:100967. [PMID: 32240622 PMCID: PMC7139120 DOI: 10.1016/j.molmet.2020.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Decreased muscle mass is a major contributor to age-related morbidity, and strategies to improve muscle regeneration during ageing are urgently needed. Our aim was to identify the subset of relevant microRNAs (miRNAs) that partake in critical aspects of muscle cell differentiation, irrespective of computational predictions, genomic clustering or differential expression of the miRNAs. METHODS miRNA biogenesis was deleted in primary myoblasts using a tamoxifen-inducible CreLox system and combined with an add-back miRNA library screen. RNA-seq experiments, cellular signalling events, and glycogen synthesis, along with miRNA inhibitors, were performed in human primary myoblasts. Muscle regeneration in young and aged mice was assessed using the cardiotoxin (CTX) model. RESULTS We identified a hierarchical non-clustered miRNA network consisting of highly (miR-29a), moderately (let-7) and mildly active (miR-125b, miR-199a, miR-221) miRNAs that cooperate by directly targeting members of the focal adhesion complex. Through RNA-seq experiments comparing single versus combinatorial inhibition of the miRNAs, we uncovered a fundamental feature of this network, that miRNA activity inversely correlates to miRNA cooperativity. During myoblast differentiation, combinatorial inhibition of the five miRNAs increased activation of focal adhesion kinase (FAK), AKT, and p38 mitogen-activated protein kinase (MAPK), and improved myotube formation and insulin-dependent glycogen synthesis. Moreover, antagonizing the miRNA network in vivo following CTX-induced muscle regeneration enhanced muscle mass and myofiber formation in young and aged mice. CONCLUSION Our results provide novel insights into the dynamics of miRNA cooperativity and identify a miRNA network as therapeutic target for impaired focal adhesion signalling and muscle regeneration during ageing.
Collapse
Affiliation(s)
- Edlira Luca
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, 8091, Switzerland
| | - Katarina Turcekova
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, 8091, Switzerland; Competence Center Personalized Medicine UZH/ETH, ETH Zurich and University of Zurich, 8091, Switzerland
| | - Angelika Hartung
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, 8091, Switzerland
| | - Sebastian Mathes
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, 8091, Switzerland; Zurich Center for Integrative Human Physiology, University of Zurich, 8091, Switzerland
| | - Hubert Rehrauer
- Functional Genomics Center Zurich UZH/ETH, ETH Zurich and University of Zurich, 8091, Switzerland
| | - Jan Krützfeldt
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, 8091, Switzerland; Competence Center Personalized Medicine UZH/ETH, ETH Zurich and University of Zurich, 8091, Switzerland; Zurich Center for Integrative Human Physiology, University of Zurich, 8091, Switzerland.
| |
Collapse
|
13
|
Nascimento TL, Conte TC, Rissato TS, Luna MS, Soares AG, Moriscot AS, Yamanouye N, Miyabara EH. Radicicol enhances the regeneration of skeletal muscle injured by crotoxin via decrease of NF-kB activation. Toxicon 2019; 167:6-9. [PMID: 31173791 DOI: 10.1016/j.toxicon.2019.06.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 05/29/2019] [Accepted: 06/03/2019] [Indexed: 11/28/2022]
Abstract
This study evaluated cellular and molecular effects of radicicol, a heat shock protein (HSP) inducer, on the regeneration of skeletal muscle injured by crotoxin, the main toxin isolated from Crotalus durissus terrificus venom. Regenerating muscles treated with radicicol had decreased NF-kB activation. Differentiating myoblasts treated with radicicol showed reduced number of NF-kB positive nuclei and increased fusion index. The results suggest that radicicol enhances regeneration of muscle by attenuating NF-kB activation and increasing myogenic differentiation.
Collapse
Affiliation(s)
- T L Nascimento
- Department of Anatomy, Biomedical Sciences Institute, University of São Paulo. Av. Lineu Prestes 2415, 05508-000, São Paulo, SP, Brazil
| | - T C Conte
- Department of Anatomy, Biomedical Sciences Institute, University of São Paulo. Av. Lineu Prestes 2415, 05508-000, São Paulo, SP, Brazil
| | - T S Rissato
- Department of Anatomy, Biomedical Sciences Institute, University of São Paulo. Av. Lineu Prestes 2415, 05508-000, São Paulo, SP, Brazil
| | - M S Luna
- Pharmacology Laboratory, Butantan Institute, Av. Vital Brazil 1500, 05503-900, São Paulo, SP, Brazil
| | - A G Soares
- Department of Pharmacology, Biomedical Sciences Institute, University of São Paulo. Av Lineu Prestes 1524, 05508-900, São Paulo, SP, Brazil
| | - A S Moriscot
- Department of Anatomy, Biomedical Sciences Institute, University of São Paulo. Av. Lineu Prestes 2415, 05508-000, São Paulo, SP, Brazil
| | - N Yamanouye
- Pharmacology Laboratory, Butantan Institute, Av. Vital Brazil 1500, 05503-900, São Paulo, SP, Brazil
| | - E H Miyabara
- Department of Anatomy, Biomedical Sciences Institute, University of São Paulo. Av. Lineu Prestes 2415, 05508-000, São Paulo, SP, Brazil.
| |
Collapse
|
14
|
Xu B, Magli A, Anugrah Y, Koester SJ, Perlingeiro RCR, Shen W. Nanotopography-responsive myotube alignment and orientation as a sensitive phenotypic biomarker for Duchenne Muscular Dystrophy. Biomaterials 2018; 183:54-66. [PMID: 30149230 PMCID: PMC6239205 DOI: 10.1016/j.biomaterials.2018.08.047] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/09/2018] [Accepted: 08/20/2018] [Indexed: 01/08/2023]
Abstract
Duchenne Muscular Dystrophy (DMD) is a fatal genetic disorder currently having no cure. Here we report that culture substrates patterned with nanogrooves and functionalized with Matrigel (or laminin) present an engineered cell microenvironment to allow myotubes derived from non-diseased, less-affected DMD, and severely-affected DMD human induced pluripotent stem cells (hiPSCs) to exhibit prominent differences in alignment and orientation, providing a sensitive phenotypic biomarker to potentially facilitate DMD drug development and early diagnosis. We discovered that myotubes differentiated from myogenic progenitors derived from non-diseased hiPSCs align nearly perpendicular to nanogrooves, a phenomenon not reported previously. We further found that myotubes derived from hiPSCs of a dystrophin-null DMD patient orient randomly, and those from hiPSCs of a patient carrying partially functional dystrophin align approximately 14° off the alignment direction of non-diseased myotubes. Substrates engineered with micron-scale grooves and/or cell adhesion molecules only interacting with integrins all guide parallel myotube alignment to grooves and lose the ability to distinguish different cell types. Disruption of the interaction between the Dystrophin-Associated-Protein-Complex (DAPC) and laminin by heparin or anti-α-dystroglycan antibody IIH6 disenables myotubes to align perpendicular to nanogrooves, suggesting that this phenotype is controlled by the DAPC-mediated cytoskeleton-extracellular matrix linkage.
Collapse
Affiliation(s)
- Bin Xu
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Alessandro Magli
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Yoska Anugrah
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Steven J Koester
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Institute for Engineering in Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Rita C R Perlingeiro
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA; Institute for Engineering in Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Wei Shen
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA; Institute for Engineering in Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
15
|
Domenighetti AA, Mathewson MA, Pichika R, Sibley LA, Zhao L, Chambers HG, Lieber RL. Loss of myogenic potential and fusion capacity of muscle stem cells isolated from contractured muscle in children with cerebral palsy. Am J Physiol Cell Physiol 2018; 315:C247-C257. [PMID: 29694232 DOI: 10.1152/ajpcell.00351.2017] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cerebral palsy (CP) is the most common cause of pediatric neurodevelopmental and physical disability in the United States. It is defined as a group of motor disorders caused by a nonprogressive perinatal insult to the brain. Although the brain lesion is nonprogressive, there is a progressive, lifelong impact on skeletal muscles, which are shorter, spastic, and may develop debilitating contractures. Satellite cells are resident muscle stem cells that are indispensable for postnatal growth and regeneration of skeletal muscles. Here we measured the myogenic potential of satellite cells isolated from contractured muscles in children with CP. When compared with typically developing (TD) children, satellite cell-derived myoblasts from CP differentiated more slowly (slope: 0.013 (SD 0.013) CP vs. 0.091 (SD 0.024) TD over 24 h, P < 0.001) and fused less (fusion index: 21.3 (SD 8.6) CP vs. 81.3 (SD 7.7) TD after 48 h, P < 0.001) after exposure to low-serum conditions that stimulated myotube formation. This impairment was associated with downregulation of several markers important for myoblast fusion and myotube formation, including DNA methylation-dependent inhibition of promyogenic integrin-β 1D (ITGB1D) protein expression levels (-50% at 42 h), and ~25% loss of integrin-mediated focal adhesion kinase phosphorylation. The cytidine analog 5-Azacytidine (5-AZA), a demethylating agent, restored ITGB1D levels and promoted myogenesis in CP cultures. Our data demonstrate that muscle contractures in CP are associated with loss of satellite cell myogenic potential that is dependent on DNA methylation patterns affecting expression of genetic programs associated with muscle stem cell differentiation and muscle fiber formation.
Collapse
Affiliation(s)
- Andrea A Domenighetti
- The Shirley Ryan AbilityLab, Chicago, Illinois.,Department of Physical Medicine & Rehabilitation, Northwestern University , Chicago, Illinois.,Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, California
| | - Margie A Mathewson
- Bioengineering Department, University of California, San Diego, La Jolla, California
| | | | | | - Leyna Zhao
- ACEA Biosciences Incorporated, San Diego, California
| | | | - Richard L Lieber
- The Shirley Ryan AbilityLab, Chicago, Illinois.,Department of Physical Medicine & Rehabilitation, Northwestern University , Chicago, Illinois.,Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, California
| |
Collapse
|
16
|
Wang D, Gao CQ, Chen RQ, Jin CL, Li HC, Yan HC, Wang XQ. Focal adhesion kinase and paxillin promote migration and adhesion to fibronectin by swine skeletal muscle satellite cells. Oncotarget 2017; 7:30845-54. [PMID: 27127174 PMCID: PMC5058722 DOI: 10.18632/oncotarget.9010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 04/08/2016] [Indexed: 12/18/2022] Open
Abstract
The focal adhesion kinase (FAK) signaling pathway contributes to the cell migration and adhesion that is critical for wound healing and regeneration of damaged muscle, but its function in skeletal muscle satellite cells (SCs) is less clear. We compared the migration and adhesion of SCs derived from two species of pig (Lantang and Landrace) in vitro, and explored how FAK signaling modulates the two processes. The results showed that Lantang SCs had greater ability to migrate and adhere to fibronection (P < 0.05) than Landrace SCs. Compared to Landrace SCs, Lantang SCs expressed many more focal adhesion (FA) sites, which were indicated by the presence of p-paxillin (Tyr118), and exhibited less F-actin reorganization 24 h after seeding onto fibronectin. Levels of p-FAK (Tyr397) and p-paxillin (Tyr118) were greater (P < 0.05) in Lantang SCs than Landrace SCs after migration for 24 h. Similarly, Lantang SCs showed much higher levels of p-FAK (Tyr397), p-paxillin (Tyr118) and p-Akt (Ser473) than Landrace SCs 2 h after adhesion. Treatment with the FAK inhibitor PF-573228 (5 or 10 μmol/L) inhibited Lantang SC migration and adhesion to fibronectin (P < 0.05), decreased levels of p-paxillin (Tyr118) and p-Akt (Ser473) (P < 0.05), and suppressed the formation of FA sites on migrating SCs. Thus FAK appears to play a key role in the regulation of SC migration and adhesion necessary for muscle regeneration.
Collapse
Affiliation(s)
- Dan Wang
- College of Animal Science, South China Agricultural University/National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong Province, China
| | - Chun-Qi Gao
- College of Animal Science, South China Agricultural University/National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong Province, China
| | - Rong-Qiang Chen
- College of Animal Science, South China Agricultural University/National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong Province, China
| | - Cheng-Long Jin
- College of Animal Science, South China Agricultural University/National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong Province, China
| | - Hai-Chang Li
- Davis Heart and Lung Research Institute, Wexner Medical Center at the Ohio State University, Columbus, OH, USA
| | - Hui-Chao Yan
- College of Animal Science, South China Agricultural University/National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong Province, China
| | - Xiu-Qi Wang
- College of Animal Science, South China Agricultural University/National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong Province, China
| |
Collapse
|
17
|
Zubac D, Šimunič B. Skeletal Muscle Contraction Time and Tone Decrease After 8 Weeks of Plyometric Training. J Strength Cond Res 2017; 31:1610-1619. [PMID: 28538312 DOI: 10.1519/jsc.0000000000001626] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The aim of the study was to examine whether an improvement in jumping performance after 8 weeks of plyometric training (PT) runs in parallel with changes in lower-limb skeletal muscle contractile properties. Using noninvasive tensiomyography (TMG), we assessed contraction time (Tc) and the maximal amplitude of radial displacement (Dm) in 20 subjects (50% men; age 22.4 ± 4.7 years of age), randomly divided in PT group (N = 10; PLYO) and a control group (N = 10; CTRL). The PLYO performed 8 weeks of PT. Tensiomyography was measured in 5 leg skeletal muscles: vastus lateralis (VL), biceps femoris (BF), tibialis anterior (TA), gastrocnemius medialis (GM), and gastrocnemius lateralis (GL). Additionally, we evaluated countermovement jump (CMJ) height improvement on a ground force plate. Assessments were repeated before and after PT. After 8 weeks of PT, CMJ height increased by 12.2% in PLYO (p = 0.015), but not in CRTL. Contraction time, which is related to myosin heavy-chain type 1 (MHC-1) proportion, decreased in VL (-8.7%; p < 0.001), BF (-26.7%; p = 0.032), TA (-32.9%; p = 0.004), and GL (-25.8%; p = 0.044), but not in GM (-8.1%; p = 0.158). The estimated VL MHC-1 proportion decreased by -8.2% (p = 0.041). The maximal amplitude of radial displacement, inversely related to muscle tone, decreased in BF (-26.5%; p = 0.032), GM (-14.9%; p = 0.017), GL (-31.5%; p = 0.017), but not in TA (-16.8%; p = 0.113) and VL (-6.0%; p = 0.654). After PT, jumping performance increased, which was paralleled by decreased Tc and decreased muscle tone. Additionally, adaptations to contractile properties were muscle specific, which is important for future studies. It seems that adjustments were dose dependent, being higher in muscles with lower habitual load.
Collapse
Affiliation(s)
- Damir Zubac
- 1Faculty of Kinesiology, University of Split, Split, Croatia; and 2Science and Research Center, Institute for Kinesiology Research, University of Primorska, Koper, Slovenia
| | | |
Collapse
|
18
|
Lee HJ, Kao CY, Lin SC, Xu M, Xie X, Tsai SY, Tsai MJ. Dysregulation of nuclear receptor COUP-TFII impairs skeletal muscle development. Sci Rep 2017; 7:3136. [PMID: 28600496 PMCID: PMC5466650 DOI: 10.1038/s41598-017-03475-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 04/28/2017] [Indexed: 02/06/2023] Open
Abstract
Chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) has been shown to inhibit myogenesis and skeletal muscle metabolism in vitro. However, its precise role and in vivo function in muscle development has yet to be clearly defined. COUP-TFII protein expression level is high in undifferentiated progenitors and gradually declines during differentiation, raising an important question of whether downregulation of COUP-TFII expression is required for proper muscle cell differentiation. In this study, we generated a mouse model ectopically expressing COUP-TFII in myogenic precursors to maintain COUP-TFII activity during myogenesis and found that elevated COUP-TFII activity resulted in inefficient skeletal muscle development. Using in vitro cell culture and in vivo mouse models, we showed that COUP-TFII hinders myogenic development by repressing myoblast fusion. Mechanistically, the inefficient muscle cell fusion correlates well with the transcriptional repression of Npnt, Itgb1D and Cav3, genes important for cell-cell fusion. We further demonstrated that COUP-TFII also reduces the activation of focal adhesion kinase (FAK), an integrin downstream regulator which is essential for fusion process. Collectively, our studies highlight the importance of down-regulation of COUP-TFII signaling to allow for the induction of factors crucial for myoblast fusion.
Collapse
Affiliation(s)
- Hui-Ju Lee
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chung-Yang Kao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Shih-Chieh Lin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan, ROC
| | - Mafei Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xin Xie
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Sophia Y Tsai
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Ming-Jer Tsai
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
19
|
Deltex2 represses MyoD expression and inhibits myogenic differentiation by acting as a negative regulator of Jmjd1c. Proc Natl Acad Sci U S A 2017; 114:E3071-E3080. [PMID: 28351977 DOI: 10.1073/pnas.1613592114] [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] [Indexed: 02/08/2023] Open
Abstract
The myogenic regulatory factor MyoD has been implicated as a key regulator of myogenesis, and yet there is little information regarding its upstream regulators. We found that Deltex2 inhibits myogenic differentiation in vitro, and that skeletal muscle stem cells from Deltex2 knockout mice exhibit precocious myogenic differentiation and accelerated regeneration in response to injury. Intriguingly, Deltex2 inhibits myogenesis by suppressing MyoD transcription, and the Deltex2 knockout phenotype can be rescued by a loss-of-function allele for MyoD In addition, we obtained evidence that Deltex2 regulates MyoD expression by promoting the enrichment of histone 3 modified by dimethylation at lysine 9 at a key regulatory region of the MyoD locus. The enrichment is attributed to a Deltex2 interacting protein, Jmjd1c, whose activity is directly inhibited by Deltex2 and whose expression is required for MyoD expression in vivo and in vitro. Finally, we find that Deltex2 causes Jmjd1c monoubiquitination and inhibits its demethylase activity. Mutation of the monoubiquitination site in Jmjd1c abolishes the inhibitory effect of Deltex2 on Jmjd1c demethylase activity. These results reveal a mechanism by which a member of the Deltex family of proteins can inhibit cellular differentiation, and demonstrate a role of Deltex in the epigenetic regulation of myogenesis.
Collapse
|
20
|
Stebbings GK, Williams AG, Morse CI, Day SH. Polymorphisms in PTK2 are associated with skeletal muscle specific force: an independent replication study. Eur J Appl Physiol 2017; 117:713-720. [PMID: 28251396 DOI: 10.1007/s00421-017-3567-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 02/08/2017] [Indexed: 02/07/2023]
Abstract
PURPOSE The aim of the study was to investigate two single nucleotide polymorphisms (SNP) in PTK2 for associations with human muscle strength phenotypes in healthy men. METHODS Measurement of maximal isometric voluntary knee extension (MVCKE) torque, net MVCKE torque and vastus lateralis (VL) specific force, using established techniques, was completed on 120 Caucasian men (age = 20.6 ± 2.3 year; height = 1.79 ± 0.06 m; mass = 75.0 ± 10.0 kg; mean ± SD). All participants provided either a blood (n = 96) or buccal cell sample, from which DNA was isolated and genotyped for the PTK2 rs7843014 A/C and rs7460 A/T SNPs using real-time polymerase chain reaction. RESULTS Genotype frequencies for both SNPs were in Hardy-Weinberg equilibrium (X 2 ≤ 1.661, P ≥ 0.436). VL specific force was 8.3% higher in rs7843014 AA homozygotes than C-allele carriers (P = 0.017) and 5.4% higher in rs7460 AA homozygotes than T-allele carriers (P = 0.029). No associations between either SNP and net MVCKE torque (P ≥ 0.094) or peak MVCKE torque (P ≥ 0.107) were observed. CONCLUSIONS These findings identify a genetic contribution to the inter-individual variability within muscle specific force and provides the first independent replication, in a larger Caucasian cohort, of an association between these PTK2 SNPs and muscle specific force, thus extending our understanding of the influence of genetic variation on the intrinsic strength of muscle.
Collapse
Affiliation(s)
- Georgina K Stebbings
- MMU Sports Genomics Laboratory, Department of Exercise and Sport Science, Manchester Metropolitan University, Crewe, UK.
| | - A G Williams
- MMU Sports Genomics Laboratory, Department of Exercise and Sport Science, Manchester Metropolitan University, Crewe, UK.,Institute of Sport, Exercise and Health, University College London, London, UK
| | - C I Morse
- MMU Sports Genomics Laboratory, Department of Exercise and Sport Science, Manchester Metropolitan University, Crewe, UK
| | - S H Day
- MMU Sports Genomics Laboratory, Department of Exercise and Sport Science, Manchester Metropolitan University, Crewe, UK
| |
Collapse
|
21
|
Deletion of the Ste20-like kinase SLK in skeletal muscle results in a progressive myopathy and muscle weakness. Skelet Muscle 2017; 7:3. [PMID: 28153048 PMCID: PMC5288853 DOI: 10.1186/s13395-016-0119-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 12/26/2016] [Indexed: 12/16/2022] Open
Abstract
Background The Ste20-like kinase, SLK, plays an important role in cell proliferation and cytoskeletal remodeling. In fibroblasts, SLK has been shown to respond to FAK/Src signaling and regulate focal adhesion turnover through Paxillin phosphorylation. Full-length SLK has also been shown to be essential for embryonic development. In myoblasts, the overexpression of a dominant negative SLK is sufficient to block myoblast fusion. Methods In this study, we crossed the Myf5-Cre mouse model with our conditional SLK knockout model to delete SLK in skeletal muscle. A thorough analysis of skeletal muscle tissue was undertaken in order to identify defects in muscle development caused by the lack of SLK. Isometric force analysis was performed on adult knockout mice and compared to age-matched wild-type mice. Furthermore, cardiotoxin injections were performed followed by immunohistochemistry for myogenic markers to assess the efficiency muscle regeneration following SLK deletion. Results We show here that early deletion of SLK from the myogenic lineage does not markedly impair skeletal muscle development but delays the regenerative process. Interestingly, adult mice (~6 months) display an increase in the proportion of central nuclei and increased p38 activation. Furthermore, mice as young as 3 months old present with decreased force generation, suggesting that the loss of SLK impairs myofiber stability and function. Assessment of structural components revealed aberrant localization of focal adhesion proteins, such as FAK and paxillin. Our data show that the loss of SLK results in unstable myofibers resulting in a progressive myopathy. Additionally, the loss of SLK resulted in a delay in muscle regeneration following cardiotoxin injections. Conclusions Our results show that SLK is dispensable for muscle development and regeneration but is required for myofiber stability and optimal force generation. Electronic supplementary material The online version of this article (doi:10.1186/s13395-016-0119-1) contains supplementary material, which is available to authorized users.
Collapse
|
22
|
Regulation of Skeletal Myoblast Differentiation by Drebrin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1006:361-373. [DOI: 10.1007/978-4-431-56550-5_22] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
23
|
Sanger JW, Wang J, Fan Y, White J, Mi-Mi L, Dube DK, Sanger JM, Pruyne D. Assembly and Maintenance of Myofibrils in Striated Muscle. Handb Exp Pharmacol 2017; 235:39-75. [PMID: 27832381 DOI: 10.1007/164_2016_53] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
In this chapter, we present the current knowledge on de novo assembly, growth, and dynamics of striated myofibrils, the functional architectural elements developed in skeletal and cardiac muscle. The data were obtained in studies of myofibrils formed in cultures of mouse skeletal and quail myotubes, in the somites of living zebrafish embryos, and in mouse neonatal and quail embryonic cardiac cells. The comparative view obtained revealed that the assembly of striated myofibrils is a three-step process progressing from premyofibrils to nascent myofibrils to mature myofibrils. This process is specified by the addition of new structural proteins, the arrangement of myofibrillar components like actin and myosin filaments with their companions into so-called sarcomeres, and in their precise alignment. Accompanying the formation of mature myofibrils is a decrease in the dynamic behavior of the assembling proteins. Proteins are most dynamic in the premyofibrils during the early phase and least dynamic in mature myofibrils in the final stage of myofibrillogenesis. This is probably due to increased interactions between proteins during the maturation process. The dynamic properties of myofibrillar proteins provide a mechanism for the exchange of older proteins or a change in isoforms to take place without disassembling the structural integrity needed for myofibril function. An important aspect of myofibril assembly is the role of actin-nucleating proteins in the formation, maintenance, and sarcomeric arrangement of the myofibrillar actin filaments. This is a very active field of research. We also report on several actin mutations that result in human muscle diseases.
Collapse
Affiliation(s)
- Joseph W Sanger
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, 766 Irving Avenue, Syracuse, NY, 13224, USA.
| | - Jushuo Wang
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, 766 Irving Avenue, Syracuse, NY, 13224, USA
| | - Yingli Fan
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, 766 Irving Avenue, Syracuse, NY, 13224, USA
| | - Jennifer White
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, 766 Irving Avenue, Syracuse, NY, 13224, USA
| | - Lei Mi-Mi
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, 766 Irving Avenue, Syracuse, NY, 13224, USA
| | - Dipak K Dube
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, 766 Irving Avenue, Syracuse, NY, 13224, USA
| | - Jean M Sanger
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, 766 Irving Avenue, Syracuse, NY, 13224, USA
| | - David Pruyne
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, 766 Irving Avenue, Syracuse, NY, 13224, USA.
| |
Collapse
|
24
|
Graham ZA, Gallagher PM, Cardozo CP. Focal adhesion kinase and its role in skeletal muscle. J Muscle Res Cell Motil 2015; 36:305-15. [PMID: 26142360 PMCID: PMC4659753 DOI: 10.1007/s10974-015-9415-3] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 06/15/2015] [Indexed: 10/23/2022]
Abstract
Skeletal muscle has a remarkable ability to respond to different physical stresses. Loading muscle through exercise, either anaerobic or aerobic, can lead to increases in muscle size and function while, conversely, the absence of muscle loading stimulates rapid decreases in size and function. A principal mediator of this load-induced change is focal adhesion kinase (FAK), a downstream non-receptor tyrosine kinase that translates the cytoskeletal stress and strain signals transmitted across the cytoplasmic membrane by integrins to activate multiple anti-apoptotic and cell growth pathways. Changes in FAK expression and phosphorylation have been found to correlate to specific developmental states in myoblast differentiation, muscle fiber formation and muscle size in response to loading and unloading. With the capability to regulate costamere formation, hypertrophy and glucose metabolism, FAK is a molecule with diverse functions that are important in regulating muscle cell health.
Collapse
Affiliation(s)
- Zachary A Graham
- Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters Veterans Affairs Medical Center, 130 W. Kingsbridge Rd., Bronx, NY, 10468, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Christopher P Cardozo
- Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters Veterans Affairs Medical Center, 130 W. Kingsbridge Rd., Bronx, NY, 10468, USA.
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
25
|
Rico P, Rodrigo-Navarro A, Salmerón-Sánchez M. Borax-Loaded PLLA for Promotion of Myogenic Differentiation. Tissue Eng Part A 2015; 21:2662-72. [PMID: 26239605 DOI: 10.1089/ten.tea.2015.0044] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Boron is an essential metalloid, which plays a key role in plant and animal metabolisms. It has been reported that boron is involved in bone mineralization, has some uses in synthetic chemistry, and its potential has been only recently exploited in medicinal chemistry. However, in the area of tissue engineering, the use of boron is limited to works involving certain bioactive glasses. In this study, we engineer poly(l-lactic acid) (PLLA) substrates with sustained release of boron. Then, we analyze for the first time the uniqueness effects of boron in cell differentiation using murine C2C12 myoblasts and discuss a potential mechanism of action in cooperation with Ca(2+). Our results demonstrate that borax-loaded materials strongly enhance myotube formation at initial steps of myogenesis. Furthermore, we demonstrate that Ca(2+) plays an essential role in combination with borax as chelating or blocking Ca(2+) entry into the cell leads to a detrimental effect on myoblast differentiation observed on borax-loaded materials. This research identifies borax-loaded materials to trigger differentiation mechanisms and it establishes a new tool to engineer microenvironments with applications in regenerative medicine for muscular diseases.
Collapse
Affiliation(s)
- Patricia Rico
- 1 Center for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València , Valencia, Spain .,2 Biomedical Research Networking Center in Bioengineering , Biomaterials and Nanomedicine (CIBER-BBN), Valencia, Spain
| | - Aleixandre Rodrigo-Navarro
- 1 Center for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València , Valencia, Spain .,3 Division of Biomedical Engineering, School of Engineering, University of Glasgow , Glasgow, United Kingdom
| | - Manuel Salmerón-Sánchez
- 3 Division of Biomedical Engineering, School of Engineering, University of Glasgow , Glasgow, United Kingdom
| |
Collapse
|
26
|
Abstract
Muscle fibres are very specialised cells with a complex structure that requires a high level of organisation of the constituent proteins. For muscle contraction to function properly, there is a need for not only sarcomeres, the contractile structures of the muscle fibre, but also costameres. These are supramolecular structures associated with the sarcolemma that allow muscle adhesion to the extracellular matrix. They are composed of protein complexes that interact and whose functions include maintaining cell structure and signal transduction mediated by their constituent proteins. It is important to improve our understanding of these structures, as mutations in various genes that code for costamere proteins cause many types of muscular dystrophy. In this review, we provide a description of costameres detailing each of their constituent proteins, such as dystrophin, dystrobrevin, syntrophin, sarcoglycans, dystroglycans, vinculin, talin, integrins, desmin, plectin, etc. We describe as well the diseases associated with deficiency thereof, providing a general overview of their importance.
Collapse
|
27
|
Garatachea N, Fuku N, He ZH, Tian Y, Arai Y, Abe Y, Murakami H, Miyachi M, Yvert T, Venturini L, Santiago C, Santos-Lozano A, Rodríguez G, Ricevuti G, Pareja-Galeano H, Sanchis-Gomar F, Emanuele E, Hirose N, Lucia A. PTK2 rs7460 and rs7843014 polymorphisms and exceptional longevity: a functional replication study. Rejuvenation Res 2015; 17:430-8. [PMID: 24930376 DOI: 10.1089/rej.2014.1570] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Focal adhesion is critical for cell survival. The focal adhesion kinase (FAK, or PTK2) is an important component of the human interactome and thus is a potential longevity-related protein. Here we studied the association between two PTK2 gene single-nucleotide polymorphisms (SNPs) (rs7843014, rs7460) and exceptional longevity (EL). In addition to gaining insight into their functionality by determining luciferase gene reporter activity, we studied the genotype/allele frequency of these two SNPs among three different cohorts: (1) Spanish centenarians (n=175, 100-111 years, 144 women) and healthy controls (n=355, 20-50 years, 284 women); (2) Italian centenarians (n=79, 100-104 years, 40 women) and controls (n=316, 29-50 years, 156 women); and (3) Japanese centenarians (n=742, 100-116 years, 623 women) and healthy controls (n=499, 23-59 years, 356 women). Both SNPs had functional significance, with the A allele up-regulating luciferase activity compared to the other allele (rs7460 T allele and rs7843014 C allele, respectively). The A allele of both SNPs was negatively associated with EL in the Spanish cohort (rs7460, odds ratio [OR] adjusted by sex=0.40, 95% confidence intervals [CI] 0.3, 0.6, p<0.001); rs7843014, OR=0.37, 95% CI 0.3, 0.5, p<0.001). The OR of being a centenarian if having the rs7460-TT genotype was 6.68 (95% CI 4.1, 10.8, p<0.001). The rs7843014 CC genotype was also positively associated with EL (OR=7.58, 95% CI 4.6, 12.3, p<0.001]. No association was, however, found for the Italian or Japanese cohorts. Thus, two genotypes of the FAK gene, rs7460 TT and rs7843014 CC, are possibly associated with lower gene expression and might favor the likelihood of reaching EL in the Spanish population. Further research is needed to unveil the mechanisms by which FAK expression could perhaps influence the rate of aging.
Collapse
Affiliation(s)
- Nuria Garatachea
- 1 Faculty of Health and Sport Science, University of Zaragoza , Huesca, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Maartens AP, Brown NH. The many faces of cell adhesion during Drosophila muscle development. Dev Biol 2015; 401:62-74. [DOI: 10.1016/j.ydbio.2014.12.038] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 12/17/2014] [Accepted: 12/19/2014] [Indexed: 10/24/2022]
|
29
|
Early changes in costameric and mitochondrial protein expression with unloading are muscle specific. BIOMED RESEARCH INTERNATIONAL 2014; 2014:519310. [PMID: 25313365 PMCID: PMC4182083 DOI: 10.1155/2014/519310] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 08/23/2014] [Indexed: 11/25/2022]
Abstract
We hypothesised that load-sensitive expression of costameric proteins, which hold the sarcomere in place and position the mitochondria, contributes to the early adaptations of antigravity muscle to unloading and would depend on muscle fibre composition and chymotrypsin activity of the proteasome. Biopsies were obtained from vastus lateralis (VL) and soleus (SOL) muscles of eight men before and after 3 days of unilateral lower limb suspension (ULLS) and subjected to fibre typing and measures for costameric (FAK and FRNK), mitochondrial (NDUFA9, SDHA, UQCRC1, UCP3, and ATP5A1), and MHCI protein and RNA content. Mean cross-sectional area (MCSA) of types I and II muscle fibres in VL and type I fibres in SOL demonstrated a trend for a reduction after ULLS (0.05 ≤ P < 0.10). FAK phosphorylation at tyrosine 397 showed a 20% reduction in VL muscle (P = 0.029). SOL muscle demonstrated a specific reduction in UCP3 content (−23%; P = 0.012). Muscle-specific effects of ULLS were identified for linear relationships between measured proteins, chymotrypsin activity and fibre MCSA. The molecular modifications in costamere turnover and energy homoeostasis identify that aspects of atrophy and fibre transformation are detectable at the protein level in weight-bearing muscles within 3 days of unloading.
Collapse
|
30
|
Salanova M, Gelfi C, Moriggi M, Vasso M, Viganò A, Minafra L, Bonifacio G, Schiffl G, Gutsmann M, Felsenberg D, Cerretelli P, Blottner D. Disuse deterioration of human skeletal muscle challenged by resistive exercise superimposed with vibration: evidence from structural and proteomic analysis. FASEB J 2014; 28:4748-63. [PMID: 25122557 DOI: 10.1096/fj.14-252825] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In the present bed rest (BR) study, 23 volunteers were randomized into 3 subgroups: 60 d BR control (Ctr); BR with resistive exercise (RE; lower-limb load); and resistive vibration exercise (RVE; RE with superimposed vibration). The aim was to analyze by confocal and electron microscopy the effects of vibration on myofibril and filament integrity in soleus (Sol) and vastus lateralis (VL) muscle; differential proteomics of contractile, cytoskeletal, and costameric proteins (TN-C, ROCK1, and FAK); and expression of PGC1α and atrophy-related master genes MuRF1 and MuRF2. RVE (but not RE) preserved myofiber size and phenotype in Sol and VL by overexpressing MYBPC1 (42%, P ≤ 0.01), WDR1 (39%, P ≤ 0.01), sarcosin (84%, P ≤ 0.01), and CKM (20%, P ≤ 0.01) and prevented myofibrillar ultrastructural damage as detectable by MuRF1 expression. In Sol, cytoskeletal and contractile proteins were normalized by RVE, and TN-C increased (59%, P ≤ 0.01); the latter also with RE (108%, P ≤ 0.01). In VL, the outcomes of both RVE (acting on sarcosin and desmin) and RE (by way of troponinT-slow and MYL2) were similar. RVE appears to be a highly efficient countermeasure protocol against muscle atrophy and ultrastructural and molecular dysregulation induced by chronic disuse.
Collapse
Affiliation(s)
- Michele Salanova
- Center of Space Medicine Berlin, Neuromuscular Group, Institute of Anatomy, and
| | - Cecilia Gelfi
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy; Consiglio Nazionale delle Ricerche (CNR), Institute of Bioimaging and Molecular Physiology, Segrate, Italy; Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Donato, San Donato Milanese, Italy;
| | - Manuela Moriggi
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Michele Vasso
- Consiglio Nazionale delle Ricerche (CNR), Institute of Bioimaging and Molecular Physiology, Segrate, Italy; Institute of Bioimaging and Molecular Physiology, CNR-Laboratorio di Tecnologie Oncologiche (LATO), Cefalù, Italy; and
| | - Agnese Viganò
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Luigi Minafra
- Institute of Bioimaging and Molecular Physiology, CNR-Laboratorio di Tecnologie Oncologiche (LATO), Cefalù, Italy; and
| | - Gaetano Bonifacio
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Gudrun Schiffl
- Center of Space Medicine Berlin, Neuromuscular Group, Institute of Anatomy, and
| | - Martina Gutsmann
- Center of Space Medicine Berlin, Neuromuscular Group, Institute of Anatomy, and
| | - Dieter Felsenberg
- Center for Muscle and Bone Research (ZMK), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Paolo Cerretelli
- Consiglio Nazionale delle Ricerche (CNR), Institute of Bioimaging and Molecular Physiology, Segrate, Italy
| | - Dieter Blottner
- Center of Space Medicine Berlin, Neuromuscular Group, Institute of Anatomy, and
| |
Collapse
|
31
|
Living biointerfaces based on non-pathogenic bacteria to direct cell differentiation. Sci Rep 2014; 4:5849. [PMID: 25068919 PMCID: PMC5376178 DOI: 10.1038/srep05849] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 07/07/2014] [Indexed: 01/03/2023] Open
Abstract
Genetically modified Lactococcus lactis, non-pathogenic bacteria expressing the FNIII7-10 fibronectin fragment as a protein membrane have been used to create a living biointerface between synthetic materials and mammalian cells. This FNIII7-10 fragment comprises the RGD and PHSRN sequences of fibronectin to bind α5β1 integrins and triggers signalling for cell adhesion, spreading and differentiation. We used L. lactis strain to colonize material surfaces and produce stable biofilms presenting the FNIII7-10 fragment readily available to cells. Biofilm density is easily tunable and remains stable for several days. Murine C2C12 myoblasts seeded over mature biofilms undergo bipolar alignment and form differentiated myotubes, a process triggered by the FNIII7-10 fragment. This biointerface based on living bacteria can be further modified to express any desired biochemical signal, establishing a new paradigm in biomaterial surface functionalisation for biomedical applications.
Collapse
|
32
|
Vassilopoulos S, Gentil C, Lainé J, Buclez PO, Franck A, Ferry A, Précigout G, Roth R, Heuser JE, Brodsky FM, Garcia L, Bonne G, Voit T, Piétri-Rouxel F, Bitoun M. Actin scaffolding by clathrin heavy chain is required for skeletal muscle sarcomere organization. ACTA ACUST UNITED AC 2014; 205:377-93. [PMID: 24798732 PMCID: PMC4018784 DOI: 10.1083/jcb.201309096] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Clathrin heavy chain contributes to the formation and maintenance of the contractile apparatus in skeletal muscle through interactions with costameric proteins. The ubiquitous clathrin heavy chain (CHC), the main component of clathrin-coated vesicles, is well characterized for its role in intracellular membrane traffic and endocytosis from the plasma membrane (PM). Here, we demonstrate that in skeletal muscle CHC regulates the formation and maintenance of PM–sarcomere attachment sites also known as costameres. We show that clathrin forms large coated lattices associated with actin filaments and the muscle-specific isoform of α-actinin at the PM of differentiated myotubes. Depletion of CHC in myotubes induced a loss of actin and α-actinin sarcomeric organization, whereas CHC depletion in vivo induced a loss of contractile force due to the detachment of sarcomeres from the PM. Our results suggest that CHC contributes to the formation and maintenance of the contractile apparatus through interactions with costameric proteins and highlight an unconventional role for CHC in skeletal muscle that may be relevant to pathophysiology of neuromuscular disorders.
Collapse
Affiliation(s)
- Stéphane Vassilopoulos
- Institut National de la Santé et de la Recherche Médicale (INSERM) U974, 2 Centre National de la Recherche Scientifique (CNRS) UMR 7215, and 3 Université Pierre et Marie Curie-Paris 6, UM 76, Paris F-75013, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Salick MR, Napiwocki BN, Sha J, Knight GT, Chindhy SA, Kamp TJ, Ashton RS, Crone WC. Micropattern width dependent sarcomere development in human ESC-derived cardiomyocytes. Biomaterials 2014; 35:4454-64. [PMID: 24582552 PMCID: PMC4026015 DOI: 10.1016/j.biomaterials.2014.02.001] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 02/04/2014] [Indexed: 12/12/2022]
Abstract
In this study, human embryonic stem cell-derived cardiomyocytes were seeded onto controlled two-dimensional micropatterned features, and an improvement in sarcomere formation and cell alignment was observed in specific feature geometries. High-resolution photolithography techniques and microcontact printing were utilized to produce features of various rectangular geometries, with areas ranging from 2500 μm(2) to 160,000 μm(2). The microcontact printing method was used to pattern non-adherent poly(ethylene glycol) regions on gold coated glass slides. Matrigel and fibronectin extracellular matrix (ECM) proteins were layered onto the gold-coated glass slides, providing a controlled geometry for cell adhesion. We used small molecule-based differentiation and an antibiotic purification step to produce a pure population of immature cardiomyocytes from H9 human embryonic stem cells (hESCs). We then seeded this pure population of human cardiomyocytes onto the micropatterned features of various sizes and observed how the cardiomyocytes remodeled their myofilament structure in response to the feature geometries. Immunofluorescence was used to measure α-actinin expression, and phalloidin stains were used to detect actin presence in the patterned cells. Analysis of nuclear alignment was also used to determine how cell direction was influenced by the features. The seeded cells showed clear alignment with the features, dependent on the width rather than the overall aspect ratio of the features. It was determined that features with widths between 30 μm and 80 μm promoted highly aligned cardiomyocytes with a dramatic increase in sarcomere alignment relative to the long axis of the pattern. This creation of highly-aligned cell aggregates with robust sarcomere structures holds great potential in advancing cell-based pharmacological studies, and will help researchers to understand the means by which ECM geometries can affect myofilament structure and maturation in hESC-derived cardiomyocytes.
Collapse
Affiliation(s)
- Max R Salick
- Wisconsin Institutes for Discovery, 330 N Orchard St, Madison, WI 53715, USA; Department of Engineering Physics, University of Wisconsin - Madison, 1500 Engineering Drive, Madison, WI 53706, USA; Materials Science Program, University of Wisconsin - Madison, 1509 University Ave, Madison, WI 53706, USA
| | - Brett N Napiwocki
- Wisconsin Institutes for Discovery, 330 N Orchard St, Madison, WI 53715, USA; Department of Biomedical Engineering, University of Wisconsin - Madison, 1550 Engineering Drive, Madison, WI 53706, USA
| | - Jin Sha
- Wisconsin Institutes for Discovery, 330 N Orchard St, Madison, WI 53715, USA; School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai, China
| | - Gavin T Knight
- Wisconsin Institutes for Discovery, 330 N Orchard St, Madison, WI 53715, USA; Department of Biomedical Engineering, University of Wisconsin - Madison, 1550 Engineering Drive, Madison, WI 53706, USA
| | - Shahzad A Chindhy
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - Madison, 750 Highland Ave, Madison, WI 53706, USA
| | - Timothy J Kamp
- Wisconsin Institutes for Discovery, 330 N Orchard St, Madison, WI 53715, USA; Department of Medicine, School of Medicine and Public Health, University of Wisconsin - Madison, 750 Highland Ave, Madison, WI 53706, USA; WiCell Institute, 614 Walnut Street, Madison, WI 53726, USA; Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, 1300 University Ave, Madison, WI 53706, USA
| | - Randolph S Ashton
- Wisconsin Institutes for Discovery, 330 N Orchard St, Madison, WI 53715, USA; Department of Biomedical Engineering, University of Wisconsin - Madison, 1550 Engineering Drive, Madison, WI 53706, USA
| | - Wendy C Crone
- Wisconsin Institutes for Discovery, 330 N Orchard St, Madison, WI 53715, USA; Department of Engineering Physics, University of Wisconsin - Madison, 1500 Engineering Drive, Madison, WI 53706, USA; Materials Science Program, University of Wisconsin - Madison, 1509 University Ave, Madison, WI 53706, USA; Department of Biomedical Engineering, University of Wisconsin - Madison, 1550 Engineering Drive, Madison, WI 53706, USA.
| |
Collapse
|
34
|
Abstract
The function of muscle is to contract, which means to exert force on a substrate. The adaptations required for skeletal muscle differentiation, from a prototypic cell, involve specialization of housekeeping cytoskeletal contracting and supporting systems into crystalline arrays of proteins. Here I discuss the changes that all three cytoskeletal systems (microfilaments, intermediate filaments, and microtubules) undergo through myogenesis. I also discuss their interaction, through the membrane, to extracellular matrix and to other cells, where force will be exerted during contraction. The three cytoskeletal systems are necessary for the muscle cell and must exert complementary roles in the cell. Muscle is a responsive system, where structure and function are integrated: the structural adaptations it undergoes depend on force production. In this way, the muscle cytoskeleton is a portrait of its physiology. I review the cytoskeletal proteins and structures involved in muscle function and focus particularly on their role in myogenesis, the process by which this incredible muscle machine is made. Although the focus is on skeletal muscle, some of the discussion is applicable to cardiac and smooth muscle.
Collapse
|
35
|
Paxillin and focal adhesion kinase colocalise in human skeletal muscle and its associated microvasculature. Histochem Cell Biol 2014; 142:245-56. [PMID: 24671495 DOI: 10.1007/s00418-014-1212-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2014] [Indexed: 01/15/2023]
Abstract
Focal adhesion kinase (FAK) and paxillin are functionally linked hormonal- and mechano-sensitive proteins. We aimed to describe paxillin's subcellular distribution using widefield and confocal immunofluorescence microscopy and test the hypothesis that FAK and paxillin colocalise in human skeletal muscle and its associated microvasculature. Percutaneous muscle biopsies were collected from the m. vastus lateralis of seven healthy males, and 5-μm cryosections were stained with anti-paxillin co-incubated with anti-dystrophin to identify the sarcolemma, anti-myosin heavy chain type I for fibre-type differentiation, anti-dihydropyridine receptor to identify T-tubules, lectin UEA-I to identify the endothelium of microvessels and anti-α-smooth muscle actin to identify vascular smooth muscle cells (VSMC). Colocalisation of anti-paxillin with anti-dystrophin or anti-FAK was quantified using Pearson's correlation coefficient on confocal microscopy images. Paxillin was primarily present in (sub)sarcolemmal regions of skeletal muscle fibres where it colocalised with dystrophin (r = 0.414 ± 0.026). The (sub)sarcolemmal paxillin immunofluorescence intensity was ~2.4-fold higher than in sarcoplasmic regions (P < 0.001) with sarcoplasmic paxillin immunofluorescence intensity ~10 % higher in type I than in type II fibres (P < 0.01). In some longitudinally orientated fibres, paxillin formed striations that corresponded to the I-band region. Paxillin immunostaining was highest in endothelial and VSMC and distributed heterogeneously in both cell types. FAK and paxillin colocalised at (sub)sarcolemmal regions and within the microvasculature (r = 0.367 ± 0.036). The first images of paxillin in human skeletal muscle suggest paxillin is present in (sub)sarcolemmal and I-band regions of muscle fibres and within the microvascular endothelium and VSMC. Colocalisation of FAK and paxillin supports their suggested role in hormonal and mechano-sensitive signalling.
Collapse
|
36
|
Lee EJ, Malik A, Pokharel S, Ahmad S, Mir BA, Cho KH, Kim J, Kong JC, Lee DM, Chung KY, Kim SH, Choi I. Identification of genes differentially expressed in myogenin knock-down bovine muscle satellite cells during differentiation through RNA sequencing analysis. PLoS One 2014; 9:e92447. [PMID: 24647404 PMCID: PMC3960249 DOI: 10.1371/journal.pone.0092447] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 02/21/2014] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The expression of myogenic regulatory factors (MRFs) consisting of MyoD, Myf5, myogenin (MyoG) and MRF4 characterizes various phases of skeletal muscle development including myoblast proliferation, cell-cycle exit, cell fusion and the maturation of myotubes to form myofibers. Although it is well known that the function of MyoG cannot be compensated for other MRFs, the molecular mechanism by which MyoG controls muscle cell differentiation is still unclear. Therefore, in this study, RNA-Seq technology was applied to profile changes in gene expression in response to MyoG knock-down (MyoGkd) in primary bovine muscle satellite cells (MSCs). RESULTS About 61-64% of the reads of over 42 million total reads were mapped to more than 13,000 genes in the reference bovine genome. RNA-Seq analysis identified 8,469 unique genes that were differentially expressed in MyoGkd. Among these genes, 230 were up-regulated and 224 were down-regulated by at least four-fold. DAVID Functional Annotation Cluster (FAC) and pathway analysis of all up- and down-regulated genes identified overrepresentation for cell cycle and division, DNA replication, mitosis, organelle lumen, nucleoplasm and cytosol, phosphate metabolic process, phosphoprotein phosphatase activity, cytoskeleton and cell morphogenesis, signifying the functional implication of these processes and pathways during skeletal muscle development. The RNA-Seq data was validated by real time RT-PCR analysis for eight out of ten genes as well as five marker genes investigated. CONCLUSIONS This study is the first RNA-Seq based gene expression analysis of MyoGkd undertaken in primary bovine MSCs. Computational analysis of the differentially expressed genes has identified the significance of genes such as SAP30-like (SAP30L), Protein lyl-1 (LYL1), various matrix metalloproteinases, and several glycogenes in myogenesis. The results of the present study widen our knowledge of the molecular basis of skeletal muscle development and reveal the vital regulatory role of MyoG in retaining muscle cell differentiation.
Collapse
Affiliation(s)
- Eun Ju Lee
- School of Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
- Bovine Genome Resources Bank, Yeungnam University, Gyeongsan, Republic of Korea
| | - Adeel Malik
- School of Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Smritee Pokharel
- School of Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Sarafraz Ahmad
- School of Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Bilal Ahmad Mir
- School of Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Kyung Hyun Cho
- School of Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Jihoe Kim
- School of Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Joon Chan Kong
- Biomedical Manufacturing Technology Center, Korea Institute of Industrial Technology, Yeongcheon-si, Republic of Korea
| | - Dong-Mok Lee
- Biomedical Manufacturing Technology Center, Korea Institute of Industrial Technology, Yeongcheon-si, Republic of Korea
| | - Ki Yong Chung
- Hanwoo Experiment Station, National Institute of Animal Science, RDA, Pyeongchang, Republic of Korea
| | - Sang Hoon Kim
- Department of Biology, Kyung Hee University, Seoul, Republic of Korea
| | - Inho Choi
- School of Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
- Bovine Genome Resources Bank, Yeungnam University, Gyeongsan, Republic of Korea
- * E-mail:
| |
Collapse
|
37
|
Li R, Narici MV, Erskine RM, Seynnes OR, Rittweger J, Pišot R, Šimunič B, Flück M. Costamere remodeling with muscle loading and unloading in healthy young men. J Anat 2013; 223:525-36. [PMID: 24010829 PMCID: PMC3916893 DOI: 10.1111/joa.12101] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2013] [Indexed: 11/28/2022] Open
Abstract
Costameres are mechano-sensory sites of focal adhesion in the sarcolemma that provide a structural anchor for myofibrils. Their turnover is regulated by integrin-associated focal adhesion kinase (FAK). We hypothesized that changes in content of costamere components (beta 1 integrin, FAK, meta-vinculin, gamma-vinculin) with increased and reduced loading of human anti-gravity muscle would: (i) relate to changes in muscle size and molecular parameters of muscle size regulation [p70S6K, myosin heavy chain (MHC)1 and MHCIIA]; (ii) correspond to adjustments in activity and expression of FAK, and its negative regulator, FRNK; and (iii) reflect the temporal response to reduced and increased loading. Unloading induced a progressive decline in thickness of human vastus lateralis muscle after 8 and 34 days of bedrest (−4% and −14%, respectively; n = 9), contrasting the increase in muscle thickness after 10 and 27 days of resistance training (+5% and +13%; n = 6). Changes in muscle thickness were correlated with changes in cross-sectional area of type I muscle fibers (r = 0.66) and beta 1 integrin content (r = 0.76) at the mid-point of altered loading. Changes in meta-vinculin and FAK-pY397 content were correlated (r = 0.85) and differed, together with the changes of beta 1 integrin, MHCI, MHCII and p70S6K, between the mid- and end-point of resistance training. By contrast, costamere protein level changes did not differ between time points of bedrest. The findings emphasize the role of FAK-regulated costamere turnover in the load-dependent addition and removal of myofibrils, and argue for two phases of muscle remodeling with resistance training, which do not manifest at the macroscopic level.
Collapse
Affiliation(s)
- Ruowei Li
- Institute for Biomedical Research into Human Movement and Health, Manchester Metropolitan University, Manchester, UK
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Klossner S, Li R, Ruoss S, Durieux AC, Flück M. Quantitative changes in focal adhesion kinase and its inhibitor, FRNK, drive load-dependent expression of costamere components. Am J Physiol Regul Integr Comp Physiol 2013; 305:R647-57. [PMID: 23904105 DOI: 10.1152/ajpregu.00007.2013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Costameres are mechanosensory sites of focal adhesion in the sarcolemma that reinforce the muscle-fiber composite and provide an anchor for myofibrillogenesis. We hypothesized that elevated content of the integrin-associated regulator of costamere turnover in culture, focal adhesion kinase (FAK), drives changes in costamere component content in antigravity muscle in a load-dependent way in correspondence with altered muscle weight. The content of FAK in soleus muscle being phosphorylated at autoregulatory tyrosine 397 (FAK-pY397) was increased after 20 s of stretch. FAK-pY397 content remained elevated after 24 h of stretch-overload due to upregulated FAK content. Overexpression of FAK in soleus muscle fibers by means of gene electrotransfer increased the β1-integrin (+56%) and meta-vinculin (+88%) content. α7-Integrin (P = 0.46) and γ-vinculin (P = 0.18) content was not altered after FAK overexpression. Co-overexpression of the FAK inhibitor FAK-related nonkinase (FRNK) reduced FAK-pY397 content by 33% and increased the percentage of fast-type fibers that arose in connection with hybrid fibers with gene transfer. Transplantation experiments confirmed the association of FRNK expression with slow-to-fast fiber transformation. Seven days of unloading blunted the elevation of FAK-pY397, β1-integrin, and meta-vinculin content with FAK overexpression, and this was reversed by 1 day of reloading. The results highlight that the expression of components for costameric attachment sites of myofibrils is under load- and fiber type-related control via FAK and its inhibitor FRNK.
Collapse
|
39
|
Storbeck CJ, Al-Zahrani KN, Sriram R, Kawesa S, O'Reilly P, Daniel K, McKay M, Kothary R, Tsilfidis C, Sabourin LA. Distinct roles for Ste20-like kinase SLK in muscle function and regeneration. Skelet Muscle 2013; 3:16. [PMID: 23815977 PMCID: PMC3733878 DOI: 10.1186/2044-5040-3-16] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 05/02/2013] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Cell growth and terminal differentiation are controlled by complex signaling systems that regulate the tissue-specific expression of genes controlling cell fate and morphogenesis. We have previously reported that the Ste20-like kinase SLK is expressed in muscle tissue and is required for cell motility. However, the specific function of SLK in muscle tissue is still poorly understood. METHODS To gain further insights into the role of SLK in differentiated muscles, we expressed a kinase-inactive SLK from the human skeletal muscle actin promoter. Transgenic muscles were surveyed for potential defects. Standard histological procedures and cardiotoxin-induced regeneration assays we used to investigate the role of SLK in myogenesis and muscle repair. RESULTS High levels of kinase-inactive SLK in muscle tissue produced an overall decrease in SLK activity in muscle tissue, resulting in altered muscle organization, reduced litter sizes, and reduced breeding capacity. The transgenic mice did not show any differences in fiber-type distribution but displayed enhanced regeneration capacity in vivo and more robust differentiation in vitro. CONCLUSIONS Our results show that SLK activity is required for optimal muscle development in the embryo and muscle physiology in the adult. However, reduced kinase activity during muscle repair enhances regeneration and differentiation. Together, these results suggest complex and distinct roles for SLK in muscle development and function.
Collapse
|
40
|
Ballester-Beltrán J, Lebourg M, Salmerón-Sánchez M. Dorsal and ventral stimuli in sandwich-like microenvironments. Effect on cell differentiation. Biotechnol Bioeng 2013; 110:3048-58. [DOI: 10.1002/bit.24972] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 05/29/2013] [Accepted: 05/31/2013] [Indexed: 12/16/2022]
Affiliation(s)
- José Ballester-Beltrán
- Center for Biomaterials and Tissue Engineering; Universitat Politècnica de València; Valencia 46022 Spain
| | - Myriam Lebourg
- Center for Biomaterials and Tissue Engineering; Universitat Politècnica de València; Valencia 46022 Spain
- CIBER de Bioingeniería; Biomateriales y Nanomedicina; Valencia 46022 Spain
| | - Manuel Salmerón-Sánchez
- Division of Biomedical Engineering, School of Engineering; University of Glasgow; Rankine Building, Oakfield Avenue Glasgow G12 8LT UK
| |
Collapse
|
41
|
De Palma S, Leone R, Grumati P, Vasso M, Polishchuk R, Capitanio D, Braghetta P, Bernardi P, Bonaldo P, Gelfi C. Changes in muscle cell metabolism and mechanotransduction are associated with myopathic phenotype in a mouse model of collagen VI deficiency. PLoS One 2013; 8:e56716. [PMID: 23437220 PMCID: PMC3577731 DOI: 10.1371/journal.pone.0056716] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 01/14/2013] [Indexed: 02/06/2023] Open
Abstract
This study identifies metabolic and protein phenotypic alterations in gastrocnemius, tibialis anterior and diaphragm muscles of Col6a1−/− mice, a model of human collagen VI myopathies. All three muscles of Col6a1−/− mice show some common changes in proteins involved in metabolism, resulting in decreased glycolysis and in changes of the TCA cycle fluxes. These changes lead to a different fate of α-ketoglutarate, with production of anabolic substrates in gastrocnemius and tibialis anterior, and with lipotoxicity in diaphragm. The metabolic changes are associated with changes of proteins involved in mechanotransduction at the myotendineous junction/costameric/sarcomeric level (TN-C, FAK, ROCK1, troponin I fast) and in energy metabolism (aldolase, enolase 3, triose phosphate isomerase, creatine kinase, adenylate kinase 1, parvalbumin, IDH1 and FASN). Together, these change may explain Ca2+ deregulation, impaired force development, increased muscle-relaxation-time and fiber damage found in the mouse model as well as in patients. The severity of these changes differs in the three muscles (gastrocnemius<tibialis anterior<diaphragm) and correlates to the mass-to-tendon (myotendineous junction) ratio and to muscle morphology.
Collapse
Affiliation(s)
- Sara De Palma
- Department of Biomedical Sciences for Health, University of Milan, Segrate (MI), Italy
- Institute of Molecular Bioimaging and Physiology, National Research Council (CNR), Segrate (MI), Italy
| | - Roberta Leone
- Department of Biomedical Sciences for Health, University of Milan, Segrate (MI), Italy
| | - Paolo Grumati
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Michele Vasso
- Department of Biomedical Sciences for Health, University of Milan, Segrate (MI), Italy
- Institute of Molecular Bioimaging and Physiology, National Research Council (CNR), Segrate (MI), Italy
| | - Roman Polishchuk
- Telethon Institute of Genetics and Medicine, Institute of Protein Biochemistry, Italian National Research Council (CNR), Naples, Italy
| | - Daniele Capitanio
- Department of Biomedical Sciences for Health, University of Milan, Segrate (MI), Italy
| | - Paola Braghetta
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Paolo Bernardi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Paolo Bonaldo
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Cecilia Gelfi
- Department of Biomedical Sciences for Health, University of Milan, Segrate (MI), Italy
- Institute of Molecular Bioimaging and Physiology, National Research Council (CNR), Segrate (MI), Italy
- * E-mail:
| |
Collapse
|
42
|
Biophysical Forces Modulate the Costamere and Z-Disc for Sarcomere Remodeling in Heart Failure. BIOPHYSICS OF THE FAILING HEART 2013. [DOI: 10.1007/978-1-4614-7678-8_7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
43
|
Alternative polyadenylation mediates microRNA regulation of muscle stem cell function. Cell Stem Cell 2012; 10:327-36. [PMID: 22385659 DOI: 10.1016/j.stem.2012.01.017] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2011] [Revised: 12/14/2011] [Accepted: 01/26/2012] [Indexed: 01/09/2023]
Abstract
Pax3, a key myogenic regulator, is transiently expressed during activation of adult muscle stem cells, or satellite cells (SCs), and is also expressed in a subset of quiescent SCs (QSCs), but only in specific muscles. The mechanisms regulating these variations in expression are not well understood. Here we show that Pax3 levels are regulated by miR-206, a miRNA with a previously demonstrated role in myogenic differentiation. In most QSCs and activated SCs, miR-206 expression suppresses Pax3 expression. Paradoxically, QSCs that express high levels of Pax3 also express high levels of miR-206. In these QSCs, Pax3 transcripts are subject to alternative polyadenylation, resulting in transcripts with shorter 3' untranslated regions (3'UTRs) that render them resistant to regulation by miR-206. Similar alternate polyadenylation of the Pax3 transcript also occurs in myogenic progenitors during development. Our findings may reflect a general role of alternative polyadenylation in circumventing miRNA-mediated regulation of stem cell function.
Collapse
|
44
|
Immunofluorescent visualisation of focal adhesion kinase in human skeletal muscle and its associated microvasculature. Histochem Cell Biol 2012; 138:617-26. [PMID: 22752263 DOI: 10.1007/s00418-012-0980-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2012] [Indexed: 10/28/2022]
Abstract
Within animal skeletal muscle, focal adhesion kinase (FAK) has been associated with load-dependent molecular and metabolic adaptation including the regulation of insulin sensitivity. This study aimed to generate the first visual images of the localisation of FAK within human skeletal muscle fibres and its associated microvasculature using widefield and confocal immunofluorescence microscopy. Percutaneous muscle biopsies, taken from five lean, active males, were frozen and 5-μm cryosections were incubated with FAK antibodies for visualisation in muscle fibres and the microvasculature. Anti-myosin heavy chain type I was used for fibre-type differentiation. Muscle sections were also incubated with anti-dihydropyridine receptor (DHPR) to investigate co-localisation of FAK with the t-tubules. FITC-conjugated Ulex europaeus Agglutinin I stained the endothelium of the capillaries, whilst anti-smooth muscle actin stained the vascular smooth muscle of arterioles. Fibre-type differences in the intensity of FAK immunofluorescence were determined with image analysis software. In transversely and longitudinally orientated fibres, FAK was localised at the sarcolemmal regions. In longitudinally orientated fibres, FAK staining also showed uniform striations across the fibre and co-staining with DHPR suggests FAK associates with the t-tubules. There was no fibre-type difference in sarcoplasmic FAK content. Within the capillary endothelium and arteriolar smooth muscle, FAK was distributed heterogeneously as clusters. This is the first study to visualise FAK in human skeletal muscle microvasculature and within the (sub)sarcolemmal and t-tubule regions using immunofluorescence microscopy. This technique will be an important tool for investigating the role of FAK in the intracellular signalling of human skeletal muscle and the endothelium of its associated microvasculature.
Collapse
|
45
|
Friedrich BM, Fischer-Friedrich E, Gov NS, Safran SA. Sarcomeric pattern formation by actin cluster coalescence. PLoS Comput Biol 2012; 8:e1002544. [PMID: 22685394 PMCID: PMC3369942 DOI: 10.1371/journal.pcbi.1002544] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 03/26/2012] [Indexed: 01/10/2023] Open
Abstract
Contractile function of striated muscle cells depends crucially on the almost crystalline order of actin and myosin filaments in myofibrils, but the physical mechanisms that lead to myofibril assembly remains ill-defined. Passive diffusive sorting of actin filaments into sarcomeric order is kinetically impossible, suggesting a pivotal role of active processes in sarcomeric pattern formation. Using a one-dimensional computational model of an initially unstriated actin bundle, we show that actin filament treadmilling in the presence of processive plus-end crosslinking provides a simple and robust mechanism for the polarity sorting of actin filaments as well as for the correct localization of myosin filaments. We propose that the coalescence of crosslinked actin clusters could be key for sarcomeric pattern formation. In our simulations, sarcomere spacing is set by filament length prompting tight length control already at early stages of pattern formation. The proposed mechanism could be generic and apply both to premyofibrils and nascent myofibrils in developing muscle cells as well as possibly to striated stress-fibers in non-muscle cells. Muscle contraction driving voluntary movements and the beating of the heart relies on the contraction of highly regular bundles of actin and myosin filaments, which share a periodic, sarcomeric pattern. We know little about the mechanisms by which these ‘biological crystals’ are assembled and it is a general question how order on a scale of 100 micrometers can emerge from the interactions of micrometer-sized building blocks, such as actin and myosin filaments. In our paper, we consider a computational model for a bundle of actin filaments and discuss physical mechanisms by which periodic order emerges spontaneously. Mutual crosslinking of actin filaments results in the formation and coalescence of growing actin clusters. Active elongation and shrinkage dynamics of actin filaments generates polymerization forces and causes local actin flow that can act like a conveyor belt to sort myosin filaments in place.
Collapse
Affiliation(s)
- Benjamin M Friedrich
- Department of Materials and Interfaces, Weizmann Institute of Science, Rehovot, Israel.
| | | | | | | |
Collapse
|
46
|
Parker MH, Loretz C, Tyler AE, Snider L, Storb R, Tapscott SJ. Inhibition of CD26/DPP-IV enhances donor muscle cell engraftment and stimulates sustained donor cell proliferation. Skelet Muscle 2012; 2:4. [PMID: 22340947 PMCID: PMC3299591 DOI: 10.1186/2044-5040-2-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Accepted: 02/16/2012] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Transplantation of myogenic stem cells possesses great potential for long-term repair of dystrophic muscle. In murine-to-murine transplantation experiments, CXCR4 expression marks a population of adult murine satellite cells with robust engraftment potential in mdx mice, and CXCR4-positive murine muscle-derived SP cells home more effectively to dystrophic muscle after intra-arterial delivery in mdx5cv mice. Together, these data suggest that CXCR4 plays an important role in donor cell engraftment. Therefore, we sought to translate these results to a clinically relevant canine-to-canine allogeneic transplant model for Duchenne muscular dystrophy (DMD) and determine if CXCR4 is important for donor cell engraftment. METHODS In this study, we used a canine-to-murine xenotransplantation model to quantitatively compare canine muscle cell engraftment, and test the most effective cell population and modulating factor in a canine model of DMD using allogeneic transplantation experiments. RESULTS We show that CXCR4 expressing cells are important for donor muscle cell engraftment, yet FACS sorted CXCR4-positive cells display decreased engraftment efficiency. However, diprotin A, a positive modulator of CXCR4-SDF-1 binding, significantly enhanced engraftment and stimulated sustained proliferation of donor cells in vivo. Furthermore, the canine-to-murine xenotransplantation model accurately predicted results in canine-to-canine muscle cell transplantation. CONCLUSIONS Therefore, these results establish the efficacy of diprotin A in stimulating muscle cell engraftment, and highlight the pre-clinical utility of a xenotransplantation model in assessing the relative efficacy of muscle stem cell populations.
Collapse
Affiliation(s)
- Maura H Parker
- Program in Transplantation Biology, Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, Mailstop D1-100, Seattle, WA, 98109-1024, USA.
| | | | | | | | | | | |
Collapse
|
47
|
Erskine RM, Williams AG, Jones DA, Stewart CE, Degens H. Do PTK2 gene polymorphisms contribute to the interindividual variability in muscle strength and the response to resistance training? A preliminary report. J Appl Physiol (1985) 2012; 112:1329-34. [PMID: 22323655 DOI: 10.1152/japplphysiol.01137.2011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The protein tyrosine kinase-2 (PTK2) gene encodes focal adhesion kinase, a structural protein involved in lateral transmission of muscle fiber force. We investigated whether single-nucleotide polymorphisms (SNPs) of the PTK2 gene were associated with various indexes of human skeletal muscle strength and the interindividual variability in the strength responses to resistance training. We determined unilateral knee extension single repetition maximum (1-RM), maximum isometric voluntary contraction (MVC) knee joint torque, and quadriceps femoris muscle specific force (maximum force per unit physiological cross-sectional area) before and after 9 wk of knee extension resistance training in 51 untrained young men. All participants were genotyped for the PTK2 intronic rs7843014 A/C and 3'-untranslated region (UTR) rs7460 A/T SNPs. There were no genotype associations with baseline measures or posttraining changes in 1-RM or MVC. Although the training-induced increase in specific force was similar for all PTK2 genotypes, baseline specific force was higher in PTK2 rs7843014 AA and rs7460 TT homozygotes than in the respective rs7843014 C- (P = 0.016) and rs7460 A-allele (P = 0.009) carriers. These associations between muscle specific force and PTK2 SNPs suggest that interindividual differences exist in the way force is transmitted from the muscle fibers to the tendon. Therefore, our results demonstrate for the first time the impact of genetic variation on the intrinsic strength of human skeletal muscle.
Collapse
Affiliation(s)
- Robert M Erskine
- Institute for Performance Research, Department of Exercise and Sport Science, Manchester Metropolitan University, Crewe, United Kingdom.
| | | | | | | | | |
Collapse
|
48
|
Ballester-Beltrán J, Cantini M, Lebourg M, Rico P, Moratal D, García AJ, Salmerón-Sánchez M. Effect of topological cues on material-driven fibronectin fibrillogenesis and cell differentiation. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2012; 23:195-204. [PMID: 22201030 DOI: 10.1007/s10856-011-4532-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 12/10/2011] [Indexed: 05/31/2023]
Abstract
Fibronectin (FN) assembles into fibrillar networks by cells through an integrin-dependent mechanism. We have recently shown that simple FN adsorption onto poly(ethyl acrylate) surfaces (PEA), but not control polymer (poly(methyl acrylate), PMA), also triggered FN organization into a physiological fibrillar network. FN fibrils exhibited enhanced biological activities in terms of myogenic differentiation compared to individual FN molecules. In the present study, we investigate the influence of topological cues on the material-driven FN assembly and the myogenic differentiation process. Aligned and random electrospun fibers were prepared. While FN fibrils assembled on the PEA fibers as they do on the smooth surface, the characteristic distribution of globular FN molecules observed on flat PMA transformed into non-connected FN fibrils on electrospun PMA, which significantly enhanced cell differentiation. The direct relationship between the fibrillar organization of FN at the material interface and the myogenic process was further assessed by preparing FN gradients on smooth PEA and PMA films. Isolated FN molecules observed at one edge of the substrate gradually interconnected with each other, eventually forming a fully developed network of FN fibrils on PEA. In contrast, FN adopted a globular-like conformation along the entire length of the PMA surface, and the FN gradient consisted only of increased density of adsorbed FN. Correspondingly, the percentage of differentiated cells increased monotonically along the FN gradient on PEA but not on PMA. This work demonstrates an interplay between material chemistry and topology in modulating material-driven FN fibrillogenesis and cell differentiation.
Collapse
Affiliation(s)
- José Ballester-Beltrán
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain
| | | | | | | | | | | | | |
Collapse
|
49
|
Flueck M, Eyeang-Békalé N, Héraud A, Girard A, Gimpl M, Seynnes OR, Rittweger J, Niebauer J, Mueller E, Narici M. Load-sensitive adhesion factor expression in the elderly with skiing: relation to fiber type and muscle strength. Scand J Med Sci Sports 2011; 21 Suppl 1:29-38. [PMID: 21679321 DOI: 10.1111/j.1600-0838.2011.01339.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We hypothesized that 12 weeks of downhill skiing mitigates the functional deficits of knee extensor muscles in elderly subjects due to the specific recruitment of fast motor units during forceful turns on the slope. Downhill skiing led to a 1.4-fold increase in the mean cross-sectional area of slow (P=0.04)- and fast (P=0.08)-type muscle fibers. Fold changes in the expression of the structural component of focal adhesions, gamma-vinculin, were correlated with alterations in concentric force (r=0.64). Hypertrophy of fast fibers was more pronounced in women than in men (1.7 vs 1.1). Gender-specific structural-functional adjustments of knee extensor muscles and attached patellar tendon were reflected by altered expression of pro- vs de-adhesive proteins and a number of correlations. The de-adhesive protein tenascin-C was selectively increased in women compared with men (1.7 vs 1.1) while the content of the adhesive collagen XII was specifically reduced in women. The pro-adhesive focal adhesion kinase showed a specific increase in men compared with women (1.9 vs 1.1). Our findings indicate that quantitatively matched adaptations in slow and fast motor units of extensor muscle underlie the preventive effect of skiing against sarcopenia and support that hypertrophy and reinforcement of fiber adhesion operate in the improvement of muscle strength.
Collapse
Affiliation(s)
- M Flueck
- Institute for Biomedical Research into Human Movement and Health, Manchester Metropolitan University, Manchester, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Protein tyrosine phosphatase-like A regulates myoblast proliferation and differentiation through MyoG and the cell cycling signaling pathway. Mol Cell Biol 2011; 32:297-308. [PMID: 22106411 DOI: 10.1128/mcb.05484-11] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Protein tyrosine phosphatase-like A (PTPLa) has been implicated in skeletal myogenesis and cardiogenesis. Mutations in PTPLa correlated with arrhythmogenic right ventricular dysplasia in humans and congenital centronuclear myopathy with severe hypotonia in dogs. The molecular mechanisms of PTPLa in myogenesis are unknown. In this report, we demonstrate that PTPLa is required for myoblast growth and differentiation. The cells lacking PTPLa remained immature and failed to differentiate into mature myotubes. The repressed MyoG expression was responsible for the impaired myoblast differentiation. Meanwhile, impeded cell growth, with an obvious S-phase arrest and compromised G(2)/M transition, was observed in PTPLa-deficient myoblasts. Further study demonstrated that the upregulation of cyclin D1 and cyclin E2 complexes, along with a compromised G(2)/M transition due to the decreased CDK1 (cyclin-dependent kinase 1) activity and upregulated p21, contributed to the mutant cell S-phase arrest and eventually led to the retarded cell growth. Finally, the transcriptional regulation of the PTPLa gene was explored. We identified PTPLa as a new target gene of the serum response factor (SRF). Skeletal- and cardiac-muscle-specific SRF knockouts resulted in significant decreases in PTPLa expression, suggesting a conserved transcriptional regulation of the PTPLa gene in mice.
Collapse
|