1
|
Qiu M, Zhang X, Liao L, Zhang N, Liu M. Regulatory Role of Nfix Gene in Sheep Skeletal Muscle Cell Development and Its Interaction Mechanism with MSTN. Int J Mol Sci 2024; 25:11988. [PMID: 39596059 PMCID: PMC11593348 DOI: 10.3390/ijms252211988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Skeletal muscle development is crucial for livestock production, and understanding the molecular mechanisms involved is essential for enhancing muscle growth in sheep. This study aimed to investigate the role of Nfix, a member of the nuclear factor I (NFI) family, in regulating muscle development in sheep, filling a significant gap in the current understanding of Nfix deficiency and its impact on skeletal muscle growth, as no similar studies have been reported in this species. Bioinformatic analysis, including temporal analysis of transcriptome data, identified Nfix as a potential target gene for muscle growth regulation. The effects of Nfix overexpression and knockout on the proliferation and differentiation of sheep skeletal muscle cells were investigated. Changes in the expression of associated marker genes were assessed to explore the regulatory link between Nfix and the myostatin (MSTN) gene. Additionally, target miRNAs for Nfix and MSTN were predicted using online databases such as miRWalk, resulting in the construction of an Nfix-miRNA-MSTN interactive regulatory network. The findings revealed that Nfix promotes the proliferation and differentiation of sheep skeletal muscle cells, with further analysis indicating that Nfix may regulate muscle cell development by modulating MSTN expression. This study provides preliminary insights into the function of Nfix in sheep skeletal muscle development and its regulatory interactions, addressing a critical knowledge gap regarding Nfix deficiency and its implications for muscle growth. These findings contribute to a better understanding of muscle biology in sheep and provide a theoretical foundation for future research into the regulatory mechanisms governing muscle development.
Collapse
Affiliation(s)
- Meiyu Qiu
- Institute of Biotechnology, Xinjiang Academy of Animal Science, Xinjiang Key Laboratory of Animal Biotechnology, Urumqi 830026, China
| | - Xuemei Zhang
- Institute of Biotechnology, Xinjiang Academy of Animal Science, Xinjiang Key Laboratory of Animal Biotechnology, Urumqi 830026, China
| | - Li Liao
- Institute of Biotechnology, Xinjiang Academy of Animal Science, Xinjiang Key Laboratory of Animal Biotechnology, Urumqi 830026, China
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China
| | - Ning Zhang
- Institute of Biotechnology, Xinjiang Academy of Animal Science, Xinjiang Key Laboratory of Animal Biotechnology, Urumqi 830026, China
| | - Mingjun Liu
- Institute of Biotechnology, Xinjiang Academy of Animal Science, Xinjiang Key Laboratory of Animal Biotechnology, Urumqi 830026, China
| |
Collapse
|
2
|
Vanhoutte D, Schips TG, Minerath RA, Huo J, Kavuri NSS, Prasad V, Lin SC, Bround MJ, Sargent MA, Adams CM, Molkentin JD. Thbs1 regulates skeletal muscle mass in a TGFβ-Smad2/3-ATF4-dependent manner. Cell Rep 2024; 43:114149. [PMID: 38678560 PMCID: PMC11217783 DOI: 10.1016/j.celrep.2024.114149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 03/22/2024] [Accepted: 04/09/2024] [Indexed: 05/01/2024] Open
Abstract
Loss of muscle mass is a feature of chronic illness and aging. Here, we report that skeletal muscle-specific thrombospondin-1 transgenic mice (Thbs1 Tg) have profound muscle atrophy with age-dependent decreases in exercise capacity and premature lethality. Mechanistically, Thbs1 activates transforming growth factor β (TGFβ)-Smad2/3 signaling, which also induces activating transcription factor 4 (ATF4) expression that together modulates the autophagy-lysosomal pathway (ALP) and ubiquitin-proteasome system (UPS) to facilitate muscle atrophy. Indeed, myofiber-specific inhibition of TGFβ-receptor signaling represses the induction of ATF4, normalizes ALP and UPS, and partially restores muscle mass in Thbs1 Tg mice. Similarly, myofiber-specific deletion of Smad2 and Smad3 or the Atf4 gene antagonizes Thbs1-induced muscle atrophy. More importantly, Thbs1-/- mice show significantly reduced levels of denervation- and caloric restriction-mediated muscle atrophy, along with blunted TGFβ-Smad3-ATF4 signaling. Thus, Thbs1-mediated TGFβ-Smad3-ATF4 signaling in skeletal muscle regulates tissue rarefaction, suggesting a target for atrophy-based muscle diseases and sarcopenia with aging.
Collapse
Affiliation(s)
- Davy Vanhoutte
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Tobias G Schips
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Rachel A Minerath
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jiuzhou Huo
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Naga Swathi Sree Kavuri
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Vikram Prasad
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Suh-Chin Lin
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Michael J Bround
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Michelle A Sargent
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Christopher M Adams
- Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Jeffery D Molkentin
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
3
|
Granados A, Zamperoni M, Rapone R, Moulin M, Boyarchuk E, Bouyioukos C, Del Maestro L, Joliot V, Negroni E, Mohamed M, Piquet S, Bigot A, Le Grand F, Albini S, Ait-Si-Ali S. SETDB1 modulates the TGFβ response in Duchenne muscular dystrophy myotubes. SCIENCE ADVANCES 2024; 10:eadj8042. [PMID: 38691608 PMCID: PMC11062573 DOI: 10.1126/sciadv.adj8042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 03/28/2024] [Indexed: 05/03/2024]
Abstract
Overactivation of the transforming growth factor-β (TGFβ) signaling in Duchenne muscular dystrophy (DMD) is a major hallmark of disease progression, leading to fibrosis and muscle dysfunction. Here, we investigated the role of SETDB1 (SET domain, bifurcated 1), a histone lysine methyltransferase involved in muscle differentiation. Our data show that, following TGFβ induction, SETDB1 accumulates in the nuclei of healthy myotubes while being already present in the nuclei of DMD myotubes where TGFβ signaling is constitutively activated. Transcriptomics revealed that depletion of SETDB1 in DMD myotubes leads to down-regulation of TGFβ target genes coding for secreted factors involved in extracellular matrix remodeling and inflammation. Consequently, SETDB1 silencing in DMD myotubes abrogates the deleterious effect of their secretome on myoblast differentiation by impairing myoblast pro-fibrotic response. Our findings indicate that SETDB1 potentiates the TGFβ-driven fibrotic response in DMD muscles, providing an additional axis for therapeutic intervention.
Collapse
Affiliation(s)
- Alice Granados
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, UMR7216, F-75013 Paris, France
| | - Maeva Zamperoni
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, UMR7216, F-75013 Paris, France
| | - Roberta Rapone
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, UMR7216, F-75013 Paris, France
| | - Maryline Moulin
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, UMR7216, F-75013 Paris, France
| | - Ekaterina Boyarchuk
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, UMR7216, F-75013 Paris, France
| | - Costas Bouyioukos
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, UMR7216, F-75013 Paris, France
| | - Laurence Del Maestro
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, UMR7216, F-75013 Paris, France
| | - Véronique Joliot
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, UMR7216, F-75013 Paris, France
| | - Elisa Negroni
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Myriame Mohamed
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, UMR7216, F-75013 Paris, France
| | - Sandra Piquet
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, UMR7216, F-75013 Paris, France
| | - Anne Bigot
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Fabien Le Grand
- Université Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Institut NeuroMyoGène, Pathophysiology and Genetics of Neuron and Muscle (PGNM) Unit, 69008 Lyon, France
| | - Sonia Albini
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, UMR7216, F-75013 Paris, France
| | - Slimane Ait-Si-Ali
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, UMR7216, F-75013 Paris, France
| |
Collapse
|
4
|
Hong J, Raza SHA, Liu M, Li M, Ruan J, Jia J, Ge C, Cao W. Association analysis of transcriptome and quasi-targeted metabolomics reveals the regulation mechanism underlying broiler muscle tissue development at different levels of dietary guanidinoacetic acid. Front Vet Sci 2024; 11:1384028. [PMID: 38725583 PMCID: PMC11080945 DOI: 10.3389/fvets.2024.1384028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/09/2024] [Indexed: 05/12/2024] Open
Abstract
The development and characteristics of muscle fibers in broilers are critical determinants that influence their growth performance, as well as serve as essential prerequisites for the production of high-quality chicken meat. Guanidinoacetic acid (GAA) is a crucial endogenous substance in animal creatine synthesis, and its utilization as a feed additive has been demonstrated the capabilities to enhance animal performance, optimize muscle yield, and augment carcass quality. The objective of this study was to investigate the regulation and molecular mechanism underlying muscle development in broilers at different levels of GAA via multiple omics analysis. The 90 Cobb broilers, aged 1 day, were randomly allocated into three treatments consisting of five replicates of six chickens each. The control group was provided with a basal diet, while the Normal GAA and High GAA groups received a basal diet supplemented with 1.2 g/kg and 3.6 g/kg of GAA, respectively. After a feeding period of 42 days, the pectoralis muscles were collected for histomorphological observation, transcriptome and metabolomic analysis. The results demonstrated that the addition of 1.2 g/kg GAA in the diet led to an augmentation in muscle fiber diameter and up-regulation of IGF1, IHH, ASB2, and ANKRD2 gene expression. However, a high dose of 3.6 g/kg GAA in the diet potentially reversed the beneficial effects on chicken breast development by excessively activating the TGF-β signaling pathway and reducing nucleotide metabolite content. These findings would provide a theoretical foundation for enhancing the performance and meat quality of broilers by incorporating GAA as a feed additive.
Collapse
Affiliation(s)
- Jieyun Hong
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Sayed Haidar Abbas Raza
- Guangdong Provincial Key Laboratory of Food Quality and Safety/Nation-Local Joint Engineering Research Center for Machining and Safety of Livestock and Poultry Products, South China Agricultural University, Guangzhou, China
| | - Mengqian Liu
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Mengyuan Li
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Jinrui Ruan
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Junjing Jia
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, China
| | - Changrong Ge
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, China
| | - Weina Cao
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
5
|
Liao Z, Lan H, Jian X, Huang J, Wang H, Hu J, Liao H. Myofiber directs macrophages IL-10-Vav1-Rac1 efferocytosis pathway in inflamed muscle following CTX myoinjury by activating the intrinsic TGF-β signaling. Cell Commun Signal 2023; 21:168. [PMID: 37403092 DOI: 10.1186/s12964-023-01163-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 05/10/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND To explore the role of skeletal muscle specific TGF-β signaling on macrophages efferocytosis in inflamed muscle caused by Cardiotoxin (CTX) injection. METHODS CTX myoinjury was manipulated in TGF-βr2flox/flox (control) mice or transgenic mice with TGF-β receptor 2 (TGF-βr2) being specifically deleted in skeletal muscle (SM TGF-βr2-/-). Gene levels of TGF-β signal molecules, special inflammatory mediators in damaged muscle or in cultured and differentiated myogenic precursor cells (MPC-myotubes) were monitored by transcriptome microarray or qRT-PCR. TGF-β pathway molecules, myokines and embryonic myosin heavy chain in regenerating myofibers, the phenotype and efferocytosis of macrophages were evaluated by immunofluorescence, immunoblotting, Luminex, or FACS analysis. In vitro apoptotic cells were prepared by UV-irradiation. RESULTS In control mice, TGF-β-Smad2/3 signaling were significantly up-regulated in regenerating centronuclear myofibers after CTX-myoinjury. More severe muscle inflammation was caused by the deficiency of muscle TGF-β signaling, with the increased number of M1, but the decreased number of M2 macrophages. Notably, the deficiency of TGF-β signaling in myofibers dramatically affected on the ability of macrophages to conduct efferocytosis, marked by the decreased number of Annexin-V-F4/80+Tunel+ macrophages in inflamed muscle, and the impaired uptake of macrophages to PKH67+ apoptotic cells transferred into damaged muscle. Further, our study suggested that, the intrinsic TGF-β signaling directed IL-10-Vav1-Rac1 efferocytosis signaling in muscle macrophages. CONCLUSIONS Our data demonstrate that muscle inflammation can be suppressed potentially by activating the intrinsic TGF-β signaling in myofibers to promote IL-10 dependent-macrophages efferocytosis. Video Abstract.
Collapse
Affiliation(s)
- Zhaohong Liao
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering; Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan, 528000, China
| | - Haiqiang Lan
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering; Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xiaoting Jian
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering; Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jingwen Huang
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering; Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Han Wang
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering; Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jijie Hu
- Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Hua Liao
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering; Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
6
|
Gallardo FS, Córdova-Casanova A, Bock-Pereda A, Rebolledo DL, Ravasio A, Casar JC, Brandan E. Denervation Drives YAP/TAZ Activation in Muscular Fibro/Adipogenic Progenitors. Int J Mol Sci 2023; 24:ijms24065585. [PMID: 36982659 PMCID: PMC10059792 DOI: 10.3390/ijms24065585] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/17/2023] Open
Abstract
Loss of motoneuron innervation (denervation) is a hallmark of neurodegeneration and aging of the skeletal muscle. Denervation induces fibrosis, a response attributed to the activation and expansion of resident fibro/adipogenic progenitors (FAPs), i.e., multipotent stromal cells with myofibroblast potential. Using in vivo and in silico approaches, we revealed FAPs as a novel cell population that activates the transcriptional coregulators YAP/TAZ in response to skeletal muscle denervation. Here, we found that denervation induces the expression and transcriptional activity of YAP/TAZ in whole muscle lysates. Using the PdgfraH2B:EGFP/+ transgenic reporter mice to trace FAPs, we demonstrated that denervation leads to increased YAP expression that accumulates within FAPs nuclei. Consistently, re-analysis of published single-nucleus RNA sequencing (snRNA-seq) data indicates that FAPs from denervated muscles have a higher YAP/TAZ signature level than control FAPs. Thus, our work provides the foundations to address the functional role of YAP/TAZ in FAPs in a neurogenic pathological context, which could be applied to develop novel therapeutic approaches for the treatment of muscle disorders triggered by motoneuron degeneration.
Collapse
Affiliation(s)
- Felipe S. Gallardo
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago 7750000, Chile
| | - Adriana Córdova-Casanova
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago 7750000, Chile
| | - Alexia Bock-Pereda
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago 7750000, Chile
| | - Daniela L. Rebolledo
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas 6213515, Chile
| | - Andrea Ravasio
- Institute for Biological and Medical Engineering, School of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Juan Carlos Casar
- Departamento de Neurología, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Enrique Brandan
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago 7750000, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510602, Chile
- Correspondence:
| |
Collapse
|
7
|
Xiao J, Huang J, Jian X, Wang H, Lan H, Liao Z, Gu R, Hu J, Liao H. IRE1α arm of unfolded protein response in muscle-specific TGF-β signaling-mediated regulation of muscle cell immunological properties. Cell Mol Biol Lett 2023; 28:15. [PMID: 36849929 PMCID: PMC9972623 DOI: 10.1186/s11658-023-00429-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 02/06/2023] [Indexed: 03/01/2023] Open
Abstract
Endoplasmic reticulum stress (ERS) and the unfolded protein response (UPR) are involved in various muscle pathological states. The IRE1α arm of UPR can affect immunological properties of myofiber through restraining p38 mitogen-activated protein kinases (MAPK) activation under inflammatory milieu. However, the relevant pathway molecules regulating the initiation of the IRE1α arm in myofiber remain unclear. In this work, expression of transforming growth factor-beta (TGF-β) and TGF-β receptor II (TGF-βr2), and UPR pathway activation were examined in cardiotoxin (CTX)-damaged mouse muscle, which revealed the activation of TGF-β signaling and UPR in CTX-damaged muscle and in regenerating myofibers. Using control or transgenic mice with TGF-βr2 deleted in skeletal muscle (SM TGF-βr2-/-) and the derived primary differentiating myogenic precursor cells (MPCs) treated with/without ERS activator or inhibitor, IRE1α pathway inhibitor, or TGF-β signaling activator, this study further revealed an essential role of intrinsic TGF-β signaling in regulating muscle cell to express inflammation-related molecules including H-2Kb, H2-Eα, TLR3, and special myokines. TGF-β signaling prompted UPR IRE1α arm and restrained p38 MAPK activation in myofiber under inflammatory milieu. This study uncovers a previously unrecognized function of TGF-β signaling acting as an upstream factor controlling myofiber immune capacities in the inflamed state through the UPR-IRE1α-p38 MAPK pathway.
Collapse
Affiliation(s)
- Jiangwei Xiao
- grid.284723.80000 0000 8877 7471Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515 China
| | - Jingwen Huang
- grid.284723.80000 0000 8877 7471Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515 China
| | - Xiaoting Jian
- grid.284723.80000 0000 8877 7471Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515 China
| | - Han Wang
- grid.284723.80000 0000 8877 7471Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515 China
| | - Haiqiang Lan
- grid.284723.80000 0000 8877 7471Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515 China
| | - Zhaohong Liao
- grid.284723.80000 0000 8877 7471Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515 China
| | - Ruicai Gu
- grid.266902.90000 0001 2179 3618Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, OK USA
| | - Jijie Hu
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Hua Liao
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
8
|
Wang Y, Lu J, Liu Y. Skeletal Muscle Regeneration in Cardiotoxin-Induced Muscle Injury Models. Int J Mol Sci 2022; 23:ijms232113380. [PMID: 36362166 PMCID: PMC9657523 DOI: 10.3390/ijms232113380] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Skeletal muscle injuries occur frequently in daily life and exercise. Understanding the mechanisms of regeneration is critical for accelerating the repair and regeneration of muscle. Therefore, this article reviews knowledge on the mechanisms of skeletal muscle regeneration after cardiotoxin-induced injury. The process of regeneration is similar in different mouse strains and is inhibited by aging, obesity, and diabetes. Exercise, microcurrent electrical neuromuscular stimulation, and mechanical loading improve regeneration. The mechanisms of regeneration are complex and strain-dependent, and changes in functional proteins involved in the processes of necrotic fiber debris clearance, M1 to M2 macrophage conversion, SC activation, myoblast proliferation, differentiation and fusion, and fibrosis and calcification influence the final outcome of the regenerative activity.
Collapse
|
9
|
Murach KA, Liu Z, Jude B, Figueiredo VC, Wen Y, Khadgi S, Lim S, Morena da Silva F, Greene NP, Lanner JT, McCarthy JJ, Vechetti IJ, von Walden F. Multi-transcriptome analysis following an acute skeletal muscle growth stimulus yields tools for discerning global and MYC regulatory networks. J Biol Chem 2022; 298:102515. [PMID: 36150502 PMCID: PMC9583450 DOI: 10.1016/j.jbc.2022.102515] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/15/2022] [Accepted: 09/17/2022] [Indexed: 02/01/2023] Open
Abstract
Myc is a powerful transcription factor implicated in epigenetic reprogramming, cellular plasticity, and rapid growth as well as tumorigenesis. Cancer in skeletal muscle is extremely rare despite marked and sustained Myc induction during loading-induced hypertrophy. Here, we investigated global, actively transcribed, stable, and myonucleus-specific transcriptomes following an acute hypertrophic stimulus in mouse plantaris. With these datasets, we define global and Myc-specific dynamics at the onset of mechanical overload-induced muscle fiber growth. Data collation across analyses reveals an under-appreciated role for the muscle fiber in extracellular matrix remodeling during adaptation, along with the contribution of mRNA stability to epigenetic-related transcript levels in muscle. We also identify Runx1 and Ankrd1 (Marp1) as abundant myonucleus-enriched loading-induced genes. We observed that a strong induction of cell cycle regulators including Myc occurs with mechanical overload in myonuclei. Additionally, in vivo Myc-controlled gene expression in the plantaris was defined using a genetic muscle fiber-specific doxycycline-inducible Myc-overexpression model. We determined Myc is implicated in numerous aspects of gene expression during early-phase muscle fiber growth. Specifically, brief induction of Myc protein in muscle represses Reverbα, Reverbβ, and Myh2 while increasing Rpl3, recapitulating gene expression in myonuclei during acute overload. Experimental, comparative, and in silico analyses place Myc at the center of a stable and actively transcribed, loading-responsive, muscle fiber-localized regulatory hub. Collectively, our experiments are a roadmap for understanding global and Myc-mediated transcriptional networks that regulate rapid remodeling in postmitotic cells. We provide open webtools for exploring the five RNA-seq datasets as a resource to the field.
Collapse
Affiliation(s)
- Kevin A. Murach
- Department of Health, Human Performance, and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, USA,Cell and Molecular Biology Graduate Program, University of Arkansas, Fayetteville, Arkansas, USA,For correspondence: Kevin A. Murach; Ivan J. Vechetti; Ferdinand von Walden
| | - Zhengye Liu
- Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - Baptiste Jude
- Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden,Department of Women’s and Children’s Health, Karolinska Institute, Solna, Sweden
| | - Vandre C. Figueiredo
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA,Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Yuan Wen
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA,Department of Physical Therapy, University of Kentucky, Lexington, Kentucky, USA
| | - Sabin Khadgi
- Department of Health, Human Performance, and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, USA
| | - Seongkyun Lim
- Department of Health, Human Performance, and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, USA,Cachexia Research Laboratory, University of Arkansas, Fayetteville, Arkansas, USA
| | - Francielly Morena da Silva
- Department of Health, Human Performance, and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, USA,Cachexia Research Laboratory, University of Arkansas, Fayetteville, Arkansas, USA
| | - Nicholas P. Greene
- Department of Health, Human Performance, and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, USA,Cell and Molecular Biology Graduate Program, University of Arkansas, Fayetteville, Arkansas, USA,Cachexia Research Laboratory, University of Arkansas, Fayetteville, Arkansas, USA
| | - Johanna T. Lanner
- Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - John J. McCarthy
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA,Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Ivan J. Vechetti
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Nebraska, USA,For correspondence: Kevin A. Murach; Ivan J. Vechetti; Ferdinand von Walden
| | - Ferdinand von Walden
- Department of Women’s and Children’s Health, Karolinska Institute, Solna, Sweden,For correspondence: Kevin A. Murach; Ivan J. Vechetti; Ferdinand von Walden
| |
Collapse
|
10
|
Mahdy MAA, Akl MA, Madkour FA. Effect of chitosan and curcumin nanoparticles against skeletal muscle fibrosis at early regenerative stage of glycerol-injured rat muscles. BMC Musculoskelet Disord 2022; 23:670. [PMID: 35836166 PMCID: PMC9281067 DOI: 10.1186/s12891-022-05633-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 07/06/2022] [Indexed: 11/10/2022] Open
Abstract
Introduction Chitosan and curcumin are natural products that have a wide range of beneficial effects including wound healing. However, their high molecular weight and poor water solubility limit their applications. Aims Therefore, the current study aims to evaluate the effects of chitosan (Cs) and curcumin (Cn) nanoparticles (NPs) on fibrosis and regeneration of glycerol-injured muscle. Methods Muscle injury was induced by intramuscular injection of glycerol into the tibialis anterior muscle of rats. Cs-NPs and Cn-NPs were administered at different doses intraperitoneally after injury. Injured muscles were collected at day 7 after injury, and muscle fibrosis and regeneration were assessed. Results The present results revealed that Cs-NPs and Cn-NPs treatment significantly decreased fibrosis index and increased the average myotube diameter with shifting of the distribution of myotube diameters towards larger diameters in a dose-dependent manner. Immunohistochemical analysis revealed that Cs-NPs and Cn-NPs treatment significantly decreased the number of CD-68+ cells and Col-1+ area. Results showed that Cn-NPs had a higher protective effect, in the form of attenuating muscle fibrosis and inflammation, and enhancing muscle regeneration, than that of Cs-NPs. Conclusions To our knowledge, this is the first study to document the effects of Cs-NPs in injured muscles. The results of study might be a novel approach to attenuate muscle fibrosis in humans using curcumin and chitosan nanoparticles.
Collapse
Affiliation(s)
- Mohamed A A Mahdy
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, South Valley University, Qena, 83523, Egypt.
| | - Mohamed A Akl
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Fatma A Madkour
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, South Valley University, Qena, 83523, Egypt
| |
Collapse
|
11
|
Huang T, Huang J, Liao Z, Lan H, Jian X, Gu R, Ouyang J, Hu J, Liao H. Regenerating myofiber directs Tregs and Th17 responses in inflamed muscle through the intrinsic TGF-β signaling-mediated IL-6 production. Am J Physiol Endocrinol Metab 2022; 323:E92-E106. [PMID: 35532076 DOI: 10.1152/ajpendo.00247.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Transforming growth factor-β (TGF-β) is considered to be an important immune regulatory cytokine. However, it remains unknown whether and how the muscle fiber specific-TGF-β signaling is directly involved in intramuscular inflammatory regulation by affecting T cells. Here, we addressed these in a mouse tibialis anterior muscle Cardiotoxin injection-induced injury repair model in muscle creatine kinase (MCK)-Cre control or transgenic mice with TGF-β receptor II (TGF-βr2) being specifically deleted in muscle cells (SM TGF-βr2-/-). In control mice, TGF-β2 and TGF-βr2 were found significantly upregulated in muscle after the acute injury. In mutant mice, deficiency of TGF-β signaling in muscle cells caused more serious muscle inflammation, with the increased infiltration of macrophages and CD4+ T cells at the degeneration stage (D4) and the early stage of regeneration (D7) after myoinjury. Notably, the loss of TGF-β signaling in myofibers dramatically affected CD4+ T cell function and delayed T cells withdrawal at the later stage of muscle regeneration (D10 and D15), marked by the elevated Th17, but the impaired Tregs response. Furthermore, in vivo and in vitro, the intrinsic TGF-β signaling affected immune behaviors of muscle cells and directed CD4+ T cells differentiation by impairing IL-6 production and release. It suggests that local muscle inflammation can be inhibited potentially by directly activating the TGF-β signaling pathway in muscle cells to suppress Th17, but induce Tregs responses. Thus, according to the results of this study, we found a new idea for the control of local acute inflammation in skeletal muscle.NEW & NOTEWORTHY Myofiber mediates muscle inflammatory response through activating the intrinsic TGF-β signaling. The specific TGF-β signaling activation contributes to myofiber IL-6 production and directs muscle-specific Th17 and Treg cell responses.
Collapse
Affiliation(s)
- Tao Huang
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering; Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
- Department of Anatomy, School of Basic Medical Science, Guizhou Medical University, Guizhou, China
| | - JingWen Huang
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering; Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - ZhaoHong Liao
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering; Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - HaiQiang Lan
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering; Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - XiaoTing Jian
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering; Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - RuiCai Gu
- Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma
| | - Jun Ouyang
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering; Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Jijie Hu
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hua Liao
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering; Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| |
Collapse
|
12
|
Hillege MMG, Shi A, Galli RA, Wu G, Bertolino P, Hoogaars WMH, Jaspers RT. Lack of Tgfbr1 and Acvr1b synergistically stimulates myofibre hypertrophy and accelerates muscle regeneration. eLife 2022; 11:77610. [PMID: 35323108 PMCID: PMC9005187 DOI: 10.7554/elife.77610] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/05/2022] [Indexed: 12/02/2022] Open
Abstract
In skeletal muscle, transforming growth factor-β (TGF-β) family growth factors, TGF-β1 and myostatin, are involved in atrophy and muscle wasting disorders. Simultaneous interference with their signalling pathways may improve muscle function; however, little is known about their individual and combined receptor signalling. Here, we show that inhibition of TGF-β signalling by simultaneous muscle-specific knockout of TGF-β type I receptors Tgfbr1 and Acvr1b in mice, induces substantial hypertrophy, while such effect does not occur by single receptor knockout. Hypertrophy is induced by increased phosphorylation of Akt and p70S6K and reduced E3 ligases expression, while myonuclear number remains unaltered. Combined knockout of both TGF-β type I receptors increases the number of satellite cells, macrophages and improves regeneration post cardiotoxin-induced injury by stimulating myogenic differentiation. Extra cellular matrix gene expression is exclusively elevated in muscle with combined receptor knockout. Tgfbr1 and Acvr1b are synergistically involved in regulation of myofibre size, regeneration, and collagen deposition.
Collapse
Affiliation(s)
- Michèle M G Hillege
- Department of Human Movement, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Andi Shi
- Department of Human Movement, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Ricardo A Galli
- Department of Human Movement, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Gang Wu
- Department of Oral and Maxillofacial Surgery/Pathology, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Philippe Bertolino
- Centre de Recherche en Cancérologie de Lyon, Université de Lyon, UMR INSERM U1052, CNRS 5286, Lyon, France
| | - Willem M H Hoogaars
- Department of Human Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Richard T Jaspers
- Department of Human Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
13
|
Amino Acids and IGF1 Regulation of Fish Muscle Growth Revealed by Transcriptome and microRNAome Integrative Analyses of Pacu ( Piaractus mesopotamicus) Myotubes. Int J Mol Sci 2022; 23:ijms23031180. [PMID: 35163102 PMCID: PMC8835699 DOI: 10.3390/ijms23031180] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 12/04/2022] Open
Abstract
Amino acids (AA) and IGF1 have been demonstrated to play essential roles in protein synthesis and fish muscle growth. The myoblast cell culture is useful for studying muscle regulation, and omics data have contributed enormously to understanding its molecular biology. However, to our knowledge, no study has performed the large-scale sequencing of fish-cultured muscle cells stimulated with pro-growth signals. In this work, we obtained the transcriptome and microRNAome of pacu (Piaractus mesopotamicus)-cultured myotubes treated with AA or IGF1. We identified 1228 and 534 genes differentially expressed by AA and IGF1. An enrichment analysis showed that AA treatment induced chromosomal changes, mitosis, and muscle differentiation, while IGF1 modulated IGF/PI3K signaling, metabolic alteration, and matrix structure. In addition, potential molecular markers were similarly modulated by both treatments. Muscle-miRNAs (miR-1, -133, -206 and -499) were up-regulated, especially in AA samples, and we identified molecular networks with omics integration. Two pairs of genes and miRNAs demonstrated a high-level relationship, and involvement in myogenesis and muscle growth: marcksb and miR-29b in AA, and mmp14b and miR-338-5p in IGF1. Our work helps to elucidate fish muscle physiology and metabolism, highlights potential molecular markers, and creates a perspective for improvements in aquaculture and in in vitro meat production.
Collapse
|
14
|
Rebolledo DL, Lipson KE, Brandan E. Driving fibrosis in neuromuscular diseases: Role and regulation of Connective tissue growth factor (CCN2/CTGF). Matrix Biol Plus 2021; 11:100059. [PMID: 34435178 PMCID: PMC8377001 DOI: 10.1016/j.mbplus.2021.100059] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 02/07/2023] Open
Abstract
Connective tissue growth factor or cellular communication network 2 (CCN2/CTGF) is a matricellular protein member of the CCN family involved in several crucial biological processes. In skeletal muscle, CCN2/CTGF abundance is elevated in human muscle biopsies and/or animal models for diverse neuromuscular pathologies, including muscular dystrophies, neurodegenerative disorders, muscle denervation, and muscle overuse. In this context, CCN2/CTGF is deeply involved in extracellular matrix (ECM) modulation, acting as a strong pro-fibrotic factor that promotes excessive ECM accumulation. Reducing CCN2/CTGF levels or biological activity in pathological conditions can decrease fibrosis, improve muscle architecture and function. In this work, we summarize information about the role of CCN2/CTGF in fibrosis associated with neuromuscular pathologies and the mechanisms and signaling pathways that regulate their expression in skeletal muscle.
Collapse
Affiliation(s)
- Daniela L Rebolledo
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Punta Arenas, Chile
| | | | - Enrique Brandan
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Chile.,Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile.,Fundación Ciencia y Vida, Santiago, Chile
| |
Collapse
|
15
|
Wharton's Jelly-Derived Mesenchymal Stem Cells Reduce Fibrosis in a Mouse Model of Duchenne Muscular Dystrophy by Upregulating microRNA 499. Biomedicines 2021; 9:biomedicines9091089. [PMID: 34572277 PMCID: PMC8469349 DOI: 10.3390/biomedicines9091089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 08/23/2021] [Indexed: 01/10/2023] Open
Abstract
The aim of this study was to evaluate the therapeutic effects and mechanisms of Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) in an animal model of Duchenne muscular dystrophy (DMD). Mdx mice (3-5 months old) were administered five different doses of WJ-MSCs through their tail veins. A week after injection, grip strength measurements, creatine kinase (CK) assays, immunohistochemistry, and western blots were performed for comparison between healthy mice, mdx control mice, and WJ-MSC-injected mdx mice. WJ-MSCs exerted dose-dependent multisystem therapeutic effects in mdx mice, by decreasing CK, recovering normal behavior, regenerating muscle, and reducing apoptosis and fibrosis in skeletal muscle. We also confirmed that miR-499-5p is significantly downregulated in mdx mice, and that intravenous injection of WJ-MSCs enhanced its expression, leading to anti-fibrotic effects via targeting TGFβR 1 and 3. Thus, WJ-MSCs may represent novel allogeneic "off-the-shelf" cellular products for the treatment of DMD and possibly other muscle disorders.
Collapse
|
16
|
Vanhoutte D, Schips TG, Vo A, Grimes KM, Baldwin TA, Brody MJ, Accornero F, Sargent MA, Molkentin JD. Thbs1 induces lethal cardiac atrophy through PERK-ATF4 regulated autophagy. Nat Commun 2021; 12:3928. [PMID: 34168130 PMCID: PMC8225674 DOI: 10.1038/s41467-021-24215-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 05/31/2021] [Indexed: 02/05/2023] Open
Abstract
The thrombospondin (Thbs) family of secreted matricellular proteins are stress- and injury-induced mediators of cellular attachment dynamics and extracellular matrix protein production. Here we show that Thbs1, but not Thbs2, Thbs3 or Thbs4, induces lethal cardiac atrophy when overexpressed. Mechanistically, Thbs1 binds and activates the endoplasmic reticulum stress effector PERK, inducing its downstream transcription factor ATF4 and causing lethal autophagy-mediated cardiac atrophy. Antithetically, Thbs1-/- mice develop greater cardiac hypertrophy with pressure overload stimulation and show reduced fasting-induced atrophy. Deletion of Thbs1 effectors/receptors, including ATF6α, CD36 or CD47 does not diminish Thbs1-dependent cardiac atrophy. However, deletion of the gene encoding PERK in Thbs1 transgenic mice blunts the induction of ATF4 and autophagy, and largely corrects the lethal cardiac atrophy. Finally, overexpression of PERK or ATF4 using AAV9 gene-transfer similarly promotes cardiac atrophy and lethality. Hence, we identified Thbs1-mediated PERK-eIF2α-ATF4-induced autophagy as a critical regulator of cardiomyocyte size in the stressed heart.
Collapse
Affiliation(s)
- Davy Vanhoutte
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Tobias G Schips
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Janssen Pharmaceuticals, Spring House, PA, USA
| | - Alexander Vo
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kelly M Grimes
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Tanya A Baldwin
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew J Brody
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Federica Accornero
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Michelle A Sargent
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jeffery D Molkentin
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
17
|
Rebolledo DL, Acuña MJ, Brandan E. Role of Matricellular CCN Proteins in Skeletal Muscle: Focus on CCN2/CTGF and Its Regulation by Vasoactive Peptides. Int J Mol Sci 2021; 22:5234. [PMID: 34063397 PMCID: PMC8156781 DOI: 10.3390/ijms22105234] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/28/2021] [Accepted: 05/12/2021] [Indexed: 02/08/2023] Open
Abstract
The Cellular Communication Network (CCN) family of matricellular proteins comprises six proteins that share conserved structural features and play numerous biological roles. These proteins can interact with several receptors or soluble proteins, regulating cell signaling pathways in various tissues under physiological and pathological conditions. In the skeletal muscle of mammals, most of the six CCN family members are expressed during embryonic development or in adulthood. Their roles during the adult stage are related to the regulation of muscle mass and regeneration, maintaining vascularization, and the modulation of skeletal muscle fibrosis. This work reviews the CCNs proteins' role in skeletal muscle physiology and disease, focusing on skeletal muscle fibrosis and its regulation by Connective Tissue Growth factor (CCN2/CTGF). Furthermore, we review evidence on the modulation of fibrosis and CCN2/CTGF by the renin-angiotensin system and the kallikrein-kinin system of vasoactive peptides.
Collapse
Affiliation(s)
- Daniela L. Rebolledo
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas 6213515, Chile
| | - María José Acuña
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O Higgins, Santiago 8370854, Chile
| | - Enrique Brandan
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Fundación Ciencia & Vida, Santiago 7810000, Chile
| |
Collapse
|
18
|
Pryce BR, Labrèche C, Hamoudi D, Abou-Hamad J, Al-Zahrani KN, Hodgins JJ, Boulanger-Piette A, Bossé S, Balog-Alvarez C, Frénette J, Ardolino M, Kornegay JN, Sabourin LA. Muscle-specific deletion of SLK/Stk2 enhances p38 activity and myogenesis in mdx mice. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1868:118917. [PMID: 33259860 DOI: 10.1016/j.bbamcr.2020.118917] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/23/2020] [Accepted: 11/25/2020] [Indexed: 01/17/2023]
Abstract
Duchenne's muscular dystrophy (DMD) is a severe muscle wasting disorder characterized by the loss of dystrophin expression, muscle necrosis, inflammation and fibrosis. Ongoing muscle regeneration is impaired by persistent cytokine stress, further decreasing muscle function. Patients with DMD rarely survive beyond their early 20s, with cardiac and respiratory dysfunction being the primary cause of death. Despite an increase in our understanding of disease progression as well as promising preclinical animal models for therapeutic intervention, treatment options for muscular dystrophy remain limited and novel therapeutic targets are required. Many reports suggest that the TGFβ signalling pathway is activated in dystrophic muscle and contributes to the pathology of DMD in part by impairing the differentiation of myoblasts into mature myofibers. Here, we show that in vitro knockdown of the Ste20-like kinase, SLK, can partially restore myoblast differentiation downstream of TGFβ in a Smad2/3 independent manner. In an mdx model, we demonstrate that SLK is expressed at high levels in regenerating myofibers. Muscle-specific deletion of SLK reduced leukocyte infiltration, increased myogenin and utrophin expression and enhanced differentiation. This was accompanied by resistance to eccentric contraction-induced injury in slow fiber type-enriched soleus muscles. Finally, we found that these effects were partially dependent on the upregulation of p38 signalling. Collectively, these results demonstrate that SLK downregulation can restore some aspects of disease progression in DMD.
Collapse
Affiliation(s)
- Benjamin R Pryce
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Cédrik Labrèche
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Dounia Hamoudi
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Centre Hospitalier de L'Université Laval, Université Laval, Quebec City, Quebec, Canada
| | - John Abou-Hamad
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Khalid N Al-Zahrani
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Jonathan J Hodgins
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Antoine Boulanger-Piette
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Centre Hospitalier de L'Université Laval, Université Laval, Quebec City, Quebec, Canada
| | - Sabrina Bossé
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Centre Hospitalier de L'Université Laval, Université Laval, Quebec City, Quebec, Canada
| | - Cindy Balog-Alvarez
- Department of Veterinary Integrative Biosciences, Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, TX, USA
| | - Jérôme Frénette
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Centre Hospitalier de L'Université Laval, Université Laval, Quebec City, Quebec, Canada; Département de Réadaptation, Faculté de Médecine, Université Laval, Quebec City, Quebec, Canada
| | - Michele Ardolino
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Joe N Kornegay
- Department of Veterinary Integrative Biosciences, Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, TX, USA
| | - Luc A Sabourin
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
| |
Collapse
|
19
|
Murach KA, Vechetti IJ, Van Pelt DW, Crow SE, Dungan CM, Figueiredo VC, Kosmac K, Fu X, Richards CI, Fry CS, McCarthy JJ, Peterson CA. Fusion-Independent Satellite Cell Communication to Muscle Fibers During Load-Induced Hypertrophy. FUNCTION 2020; 1:zqaa009. [PMID: 32864621 PMCID: PMC7448100 DOI: 10.1093/function/zqaa009] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/29/2020] [Accepted: 06/29/2020] [Indexed: 01/06/2023] Open
Abstract
The "canonical" function of Pax7+ muscle stem cells (satellite cells) during hypertrophic growth of adult muscle fibers is myonuclear donation via fusion to support increased transcriptional output. In recent years, however, emerging evidence suggests that satellite cells play an important secretory role in promoting load-mediated growth. Utilizing genetically modified mouse models of delayed satellite cell fusion and in vivo extracellular vesicle (EV) tracking, we provide evidence for satellite cell communication to muscle fibers during hypertrophy. Myogenic progenitor cell-EV-mediated communication to myotubes in vitro influences extracellular matrix (ECM)-related gene expression, which is congruent with in vivo overload experiments involving satellite cell depletion, as well as in silico analyses. Satellite cell-derived EVs can transfer a Cre-induced, cytoplasmic-localized fluorescent reporter to muscle cells as well as microRNAs that regulate ECM genes such as matrix metalloproteinase 9 (Mmp9), which may facilitate growth. Delayed satellite cell fusion did not limit long-term load-induced muscle hypertrophy indicating that early fusion-independent communication from satellite cells to muscle fibers is an underappreciated aspect of satellite cell biology. We cannot exclude the possibility that satellite cell-mediated myonuclear accretion is necessary to maintain prolonged growth, specifically in the later phases of adaptation, but these data collectively highlight how EV delivery from satellite cells can directly contribute to mechanical load-induced muscle fiber hypertrophy, independent of cell fusion to the fiber.
Collapse
Affiliation(s)
- Kevin A Murach
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Ivan J Vechetti
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Douglas W Van Pelt
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Samuel E Crow
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Cory M Dungan
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Vandre C Figueiredo
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Kate Kosmac
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Xu Fu
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Christopher I Richards
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Christopher S Fry
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - John J McCarthy
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Charlotte A Peterson
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
20
|
Stepien DM, Hwang C, Marini S, Pagani CA, Sorkin M, Visser ND, Huber AK, Edwards NJ, Loder SJ, Vasquez K, Aguilar CA, Kumar R, Mascharak S, Longaker MT, Li J, Levi B. Tuning Macrophage Phenotype to Mitigate Skeletal Muscle Fibrosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:2203-2215. [PMID: 32161098 PMCID: PMC8080967 DOI: 10.4049/jimmunol.1900814] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 01/20/2020] [Indexed: 12/16/2022]
Abstract
Myeloid cells are critical to the development of fibrosis following muscle injury; however, the mechanism of their role in fibrosis formation remains unclear. In this study, we demonstrate that myeloid cell-derived TGF-β1 signaling is increased in a profibrotic ischemia reperfusion and cardiotoxin muscle injury model. We found that myeloid-specific deletion of Tgfb1 abrogates the fibrotic response in this injury model and reduces fibro/adipogenic progenitor cell proliferation while simultaneously enhancing muscle regeneration, which is abrogated by adaptive transfer of normal macrophages. Similarly, a murine TGFBRII-Fc ligand trap administered after injury significantly reduced muscle fibrosis and improved muscle regeneration. This study ultimately demonstrates that infiltrating myeloid cell TGF-β1 is responsible for the development of traumatic muscle fibrosis, and its blockade offers a promising therapeutic target for preventing muscle fibrosis after ischemic injury.
Collapse
Affiliation(s)
- David M Stepien
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Charles Hwang
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Simone Marini
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109
| | - Chase A Pagani
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Michael Sorkin
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Noelle D Visser
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Amanda K Huber
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Nicole J Edwards
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Shawn J Loder
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Kaetlin Vasquez
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Carlos A Aguilar
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
- Biomedical Engineering Department, Biointerfaces Institute and Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109
| | - Ravi Kumar
- Acceleron Pharmaceuticals, Cambridge MA 02139
| | - Shamik Mascharak
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford University, Stanford, CA 94305; and
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford University, Stanford, CA 94305; and
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford University, Stanford, CA 94305
| | - Jun Li
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109
| | - Benjamin Levi
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109;
| |
Collapse
|
21
|
Contreras O, Soliman H, Theret M, Rossi FMV, Brandan E. TGF-β-driven downregulation of the Wnt/β-Catenin transcription factor TCF7L2/TCF4 in PDGFRα+ fibroblasts. J Cell Sci 2020; 133:jcs.242297. [DOI: 10.1242/jcs.242297] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 04/29/2020] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) are multipotent progenitors essential for organogenesis, tissue homeostasis, regeneration, and scar formation. Tissue injury upregulates TGF-β signaling, which modulates myofibroblast fate, extracellular matrix remodeling, and fibrosis. However, the molecular determinants of MSCs differentiation and survival remain poorly understood. The canonical Wnt Tcf/Lef transcription factors regulate development and stemness, but the mechanisms by which injury-induced cues modulate their expression remain underexplored. Here, we studied the cell-specific gene expression of Tcf/Lef and, more specifically, we investigated whether damage-induced TGF-β impairs the expression and function of TCF7L2, using several models of MSCs, including skeletal muscle fibro-adipogenic progenitors. We show that Tcf/Lefs are differentially expressed and that TGF-β reduces the expression of TCF7L2 in MSCs but not in myoblasts. We also found that the ubiquitin-proteasome system regulates TCF7L2 proteostasis and participates in TGF-β-mediated TCF7L2 protein downregulation. Finally, we show that TGF-β requires HDACs activity to repress the expression of TCF7L2. Thus, our work found a novel interplay between TGF-β and Wnt canonical signaling cascades in PDGFRα+ fibroblasts and suggests that this mechanism could be targeted in tissue repair and regeneration.
Collapse
Affiliation(s)
- Osvaldo Contreras
- Departamento de Biología Celular y Molecular and Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
- Biomedical Research Centre, Department of Medical Genetics and School of Biomedical Engineering, University of British Columbia, V6T 1Z3 Vancouver, BC, Canada
- Present address: Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, 2010, Australia
| | - Hesham Soliman
- Biomedical Research Centre, Department of Medical Genetics and School of Biomedical Engineering, University of British Columbia, V6T 1Z3 Vancouver, BC, Canada
- Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Marine Theret
- Biomedical Research Centre, Department of Medical Genetics and School of Biomedical Engineering, University of British Columbia, V6T 1Z3 Vancouver, BC, Canada
| | - Fabio M. V. Rossi
- Biomedical Research Centre, Department of Medical Genetics and School of Biomedical Engineering, University of British Columbia, V6T 1Z3 Vancouver, BC, Canada
| | - Enrique Brandan
- Departamento de Biología Celular y Molecular and Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
- Fundación Ciencia & Vida, Santiago, Chile
| |
Collapse
|
22
|
Barzilai-Tutsch H, Dewulf M, Lamaze C, Butler Browne G, Pines M, Halevy O. A promotive effect for halofuginone on membrane repair and synaptotagmin-7 levels in muscle cells of dysferlin-null mice. Hum Mol Genet 2019; 27:2817-2829. [PMID: 29771357 DOI: 10.1093/hmg/ddy185] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 05/09/2018] [Indexed: 11/14/2022] Open
Abstract
In the absence of dysferlin, skeletal muscle cells fail to reseal properly after injury, resulting in slow progress of the dysferlinopathy muscular dystrophy (MD). Halofuginone, a leading agent in preventing fibrosis in MDs, was tested for its effects on membrane resealing post-injury. A hypo-osmotic shock assay on myotubes derived from wild-type (Wt) and dysferlin-null (dysf-/-) mice revealed that pre-treatment with halofuginone reduces the percentage of membrane-ruptured myotubes only in dysf-/- myotubes. In laser-induced injury of isolated myofibers, halofuginone decreased the amount of FM1-43 at the injury site of dysf-/- myofibers while having no effect on Wt myofibers. These results implicate halofuginone in ameliorating muscle-cell membrane integrity in dysf-/- mice. Halofuginone increased lysosome scattering across the cytosol of dysf-/- primary myoblasts, in a protein kinase/extracellular signal-regulated protein kinase and phosphoinositide 3 kinase/Akt-dependent manner, in agreement with an elevation in lysosomal exocytotic activity in these cells. A spatial- and age-dependent synaptotagmin-7 (Syt-7) expression pattern was shown in dysf-/- versus Wt mice, suggesting that these pattern alterations are related to the disease progress and that sytnaptotagmin-7 may be compensating for the lack of dysferlin at least with regard to membrane resealing post-injury. While halofuginone did not affect patch-repair-complex key proteins, it further enhanced Syt-7 levels and its spread across the cytosol in dysf-/- myofibers and muscle tissue, and increased its co-localization with lysosomes. Together, the data imply a novel role for halofuginone in membrane-resealing events with Syt-7 possibly taking part in these events.
Collapse
Affiliation(s)
- Hila Barzilai-Tutsch
- Department of Animal Sciences, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Melissa Dewulf
- Membrane Dynamics and Mechanics of Intracellular Signaling Laboratory, Institut Curie-Centre de Recherche, PSL Research University, INSERM U1143, Centre national de la recherche scientifique, UMR 3666, Paris, France
| | - Christophe Lamaze
- Membrane Dynamics and Mechanics of Intracellular Signaling Laboratory, Institut Curie-Centre de Recherche, PSL Research University, INSERM U1143, Centre national de la recherche scientifique, UMR 3666, Paris, France
| | - Gillian Butler Browne
- Center for Research in Myology, CNRS FRE 3617, UPMC Univ Paris 06, UM76, INSERM U974, Sorbonne Universités, Paris, France
| | - Mark Pines
- The Volcani Center, Institute of Animal Science, Bet Dagan 52505, Israel
| | - Orna Halevy
- Department of Animal Sciences, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| |
Collapse
|
23
|
Petrosino JM, Leask A, Accornero F. Genetic manipulation of CCN2/CTGF unveils cell-specific ECM-remodeling effects in injured skeletal muscle. FASEB J 2019; 33:2047-2057. [PMID: 30216109 PMCID: PMC6338641 DOI: 10.1096/fj.201800622rr] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/20/2018] [Indexed: 01/03/2023]
Abstract
In skeletal muscle, extracellular matrix (ECM) remodeling can either support the complete regeneration of injured muscle or facilitate pathologic fibrosis and muscle degeneration. Muscular dystrophy (MD) is a group of genetic disorders that results in a progressive decline in muscle function and is characterized by the abundant deposition of fibrotic tissue. Unlike acute injury, where ECM remodeling is acute and transient, in MD, remodeling persists until fibrosis obstructs the regenerative efforts of diseased muscles. Thus, understanding how ECM is deposited and organized is critical in the context of muscle repair. Connective tissue growth factor (CTGF or CCN2) is a matricellular protein expressed by multiple cell types in response to tissue injury. Although used as a general marker of fibrosis, the cell type-dependent role of CTGF in dystrophic muscle has not been elucidated. To address this question, a conditional Ctgf myofiber and fibroblast-knockout mouse lines were generated and crossed to a dystrophic background. Only myofiber-selective inhibition of CTGF protected δ-sarcoglycan-null ( Sgcd-/-) mice from the dystrophic phenotype, and it did so by affecting collagen organization in a way that allowed for improvements in dystrophic muscle regeneration and function. To confirm that muscle-specific CTGF functions to mediate collagen organization, we generated mice with transgenic muscle-specific overexpression of CTGF. Again, genetic modulation of CTGF in muscle was not sufficient to drive fibrosis, but altered collagen content and organization after injury. Our results show that the myofibers are critical mediators of the deleterious effects associated with CTGF in MD and acutely injured skeletal muscle.-Petrosino, J. M., Leask, A., Accornero, F. Genetic manipulation of CCN2/CTGF unveils cell-specific ECM-remodeling effects in injured skeletal muscle.
Collapse
Affiliation(s)
- Jennifer M. Petrosino
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Andrew Leask
- Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Federica Accornero
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
24
|
Lighthouse JK, Burke RM, Velasquez LS, Dirkx RA, Aiezza A, Moravec CS, Alexis JD, Rosenberg A, Small EM. Exercise promotes a cardioprotective gene program in resident cardiac fibroblasts. JCI Insight 2019; 4:92098. [PMID: 30626739 DOI: 10.1172/jci.insight.92098] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 11/20/2018] [Indexed: 02/06/2023] Open
Abstract
Exercise and heart disease both induce cardiac remodeling, but only disease causes fibrosis and compromises heart function. The cardioprotective benefits of exercise have been attributed to changes in cardiomyocyte physiology, but the impact of exercise on cardiac fibroblasts (CFs) is unknown. Here, RNA-sequencing reveals rapid divergence of CF transcriptional programs during exercise and disease. Among the differentially expressed programs, NRF2-dependent antioxidant genes - including metallothioneins (Mt1 and Mt2) - are induced in CFs during exercise and suppressed by TGF-β/p38 signaling in disease. In vivo, mice lacking Mt1/2 exhibit signs of cardiac dysfunction in exercise, including cardiac fibrosis, vascular rarefaction, and functional decline. Mechanistically, exogenous MTs derived from fibroblasts are taken up by cultured cardiomyocytes, reducing oxidative damage-dependent cell death. Importantly, suppression of MT expression is conserved in human heart failure. Taken together, this study defines the acute transcriptional response of CFs to exercise and disease and reveals a cardioprotective mechanism that is lost in disease.
Collapse
Affiliation(s)
- Janet K Lighthouse
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Ryan M Burke
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Lissette S Velasquez
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Ronald A Dirkx
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Alessandro Aiezza
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | | | | | - Alex Rosenberg
- Department of Allergy, Immunology, and Rheumatology Research, and
| | - Eric M Small
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA.,Department of Medicine.,Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
25
|
Contreras O, Cruz-Soca M, Theret M, Soliman H, Tung LW, Groppa E, Rossi FM, Brandan E. The cross-talk between TGF-β and PDGFRα signaling pathways regulates stromal fibro/adipogenic progenitors’ fate. J Cell Sci 2019; 132:jcs.232157. [DOI: 10.1242/jcs.232157] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 08/12/2019] [Indexed: 12/15/2022] Open
Abstract
Fibro/adipogenic progenitors (FAPs) are tissue-resident mesenchymal stromal cells (MSCs) required for proper skeletal muscle development, regeneration, and maintenance. However, FAPs are also responsible for fibro-fatty scar deposition following chronic damage. We aimed to study a functional cross-talk between TGF-β and PDGFRα signaling pathways in FAPs’ fate. Here, we show that the number of FAPs correlates with TGF-β levels and with extracellular matrix deposition during regeneration and repair. Interestingly, the expression of PDGFRα changed dynamically in the stromal/fibroblast lineage after injury. Furthermore, PDGFRα-dependent immediate early gene expression changed during regeneration and repair. We also found that TGF-β signaling reduces PDGFRα expression in FAPs, mouse dermal fibroblasts, and in two related mesenchymal/fibroblast cell lines. Moreover, TGF-β promotes myofibroblast differentiation of FAPs but inhibits their adipogenicity. Accordingly, TGF-β impairs the expression of PDGFRα-dependent immediate early genes in a TGF-BR1-dependent manner. Finally, pharmacological inhibition of PDGFRα activity with AG1296 impaired TGF-β-induced extracellular matrix remodeling, Smad2 signaling, myofibroblast differentiation, and migration of MSCs. Thus, our work establishes a functional cross-talk between TGF-β and PDGFRα signaling pathways that is involved in regulating the biology of FAPs/MSCs.
Collapse
Affiliation(s)
- Osvaldo Contreras
- Departamento de Biología Celular y Molecular and Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Biomedical Research Centre, Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Meilyn Cruz-Soca
- Departamento de Biología Celular y Molecular and Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marine Theret
- Biomedical Research Centre, Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Hesham Soliman
- Biomedical Research Centre, Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Lin Wei Tung
- Biomedical Research Centre, Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Elena Groppa
- Biomedical Research Centre, Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Fabio M. Rossi
- Biomedical Research Centre, Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Enrique Brandan
- Departamento de Biología Celular y Molecular and Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
26
|
Yao X, Yu T, Xi F, Xu Y, Ma L, Pan X, Chen S, Han M, Yin Y, Dai X, Xu G, Zhang H, Yang G, Xie L. BAMBI shuttling between cytosol and membrane is required for skeletal muscle development and regeneration. Biochem Biophys Res Commun 2018; 509:125-132. [PMID: 30580997 DOI: 10.1016/j.bbrc.2018.12.082] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 12/12/2018] [Indexed: 01/12/2023]
Abstract
Bone morphogenetic protein and activin membrane-bound inhibitor (BAMBI) gene encodes a transmembrane protein and is involved in multiple physiological and pathological processes, such as inflammatory response, tumor development and progression, cell proliferation and differentiation. A previous study suggested that BAMBI may interact with the Wnt/β-catenin signaling pathway via promoting β-catenin nuclear translocation associated with C2C12 myogenic myoblast differentiation. However, its biological function in skeletal muscle still remains unknown and requires further characterization. The present work sought to investigate its biological function in skeletal muscle, especially the physiological roles of BAMBI during skeletal muscle growth and regeneration. Our current work suggests that BAMBI protein is highly expressed in skeletal muscle and is only detected in cytosolic fraction in the resting muscle. Moreover, BAMBI protein is co-localized in fast-twitch (glycolytic) fibers, but not in slow-twitch (oxidative) fibers. Comparing with the cytosolic trapping in resting muscle, BAMBI protein is enriched on cellular membrane during the muscle growth and regeneration, suggesting that BAMBI-mediated a significant signaling pathway may be an essential part of muscle growth and regeneration.
Collapse
Affiliation(s)
- Xiangping Yao
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China; State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangzhou, 510070, China; Guangdong Bide Biotech CO. L.T.D., Guangzhou, Guangdong, China
| | - Taiyong Yu
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Fengxue Xi
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yanting Xu
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Lu Ma
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Xiaohan Pan
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangzhou, 510070, China; Guangdong Bide Biotech CO. L.T.D., Guangzhou, Guangdong, China
| | - Shujie Chen
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangzhou, 510070, China; Guangdong Bide Biotech CO. L.T.D., Guangzhou, Guangdong, China
| | - Mulan Han
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangzhou, 510070, China; Guangdong Bide Biotech CO. L.T.D., Guangzhou, Guangdong, China
| | - Yulong Yin
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangzhou, 510070, China
| | - Xiaoshuang Dai
- BGI Institute of Applied Agriculture, BGI-Shenzhen, Shenzhen, 518120, China
| | - Guohuan Xu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangzhou, 510070, China; Guangdong Bide Biotech CO. L.T.D., Guangzhou, Guangdong, China
| | - Huabing Zhang
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China.
| | - Gongshe Yang
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangzhou, 510070, China; Guangdong Bide Biotech CO. L.T.D., Guangzhou, Guangdong, China.
| |
Collapse
|
27
|
Dowling JJ, D. Gonorazky H, Cohn RD, Campbell C. Treating pediatric neuromuscular disorders: The future is now. Am J Med Genet A 2018; 176:804-841. [PMID: 28889642 PMCID: PMC5900978 DOI: 10.1002/ajmg.a.38418] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 07/31/2017] [Indexed: 12/12/2022]
Abstract
Pediatric neuromuscular diseases encompass all disorders with onset in childhood and where the primary area of pathology is in the peripheral nervous system. These conditions are largely genetic in etiology, and only those with a genetic underpinning will be presented in this review. This includes disorders of the anterior horn cell (e.g., spinal muscular atrophy), peripheral nerve (e.g., Charcot-Marie-Tooth disease), the neuromuscular junction (e.g., congenital myasthenic syndrome), and the muscle (myopathies and muscular dystrophies). Historically, pediatric neuromuscular disorders have uniformly been considered to be without treatment possibilities and to have dire prognoses. This perception has gradually changed, starting in part with the discovery and widespread application of corticosteroids for Duchenne muscular dystrophy. At present, several exciting therapeutic avenues are under investigation for a range of conditions, offering the potential for significant improvements in patient morbidities and mortality and, in some cases, curative intervention. In this review, we will present the current state of treatment for the most common pediatric neuromuscular conditions, and detail the treatment strategies with the greatest potential for helping with these devastating diseases.
Collapse
Affiliation(s)
- James J. Dowling
- Division of NeurologyHospital for Sick ChildrenTorontoOntarioCanada
- Program for Genetics and Genome BiologyHospital for Sick ChildrenTorontoOntarioCanada
- Departments of Paediatrics and Molecular GeneticsUniversity of TorontoTorontoOntarioCanada
| | | | - Ronald D. Cohn
- Program for Genetics and Genome BiologyHospital for Sick ChildrenTorontoOntarioCanada
- Departments of Paediatrics and Molecular GeneticsUniversity of TorontoTorontoOntarioCanada
| | - Craig Campbell
- Department of PediatricsClinical Neurological SciencesEpidemiologyWestern UniversityLondonOntarioCanada
| |
Collapse
|
28
|
DiMario JX. KLF10 Gene Expression Modulates Fibrosis in Dystrophic Skeletal Muscle. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1263-1275. [PMID: 29458012 DOI: 10.1016/j.ajpath.2018.01.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 01/22/2018] [Accepted: 01/30/2018] [Indexed: 02/06/2023]
Abstract
Dystrophic skeletal muscle is characterized by fibrotic accumulation of extracellular matrix components that compromise muscle structure, function, and capacity for regeneration. Tissue fibrosis is often initiated and sustained through transforming growth factor-β (TGF-β) signaling, and Krüppel-like factor 10 (KLF10) is an immediate early gene that is transcriptionally activated in response to TGF-β signaling. It encodes a transcriptional regulator that mediates the effects of TGF-β signaling in a variety of cell types. This report presents results of investigation of the effects of loss of KLF10 gene expression in wild-type and dystrophic (mdx) skeletal muscle. On the basis of RT-PCR, Western blot, and histological analyses of mouse tibialis anterior and diaphragm muscles, collagen type I (Col1a1) and fibronectin gene expression and protein deposition were increased in KLF10-/- mice, contributing to increased fibrosis. KLF10-/- mice displayed increased expression of genes encoding SMAD2, SMAD3, and SMAD7, particularly in diaphragm muscle. SMAD4 gene expression was unchanged. Expression of the extracellular matrix remodeling genes, MMP2 and TIMP1, was also increased in KLF10-deficient mouse muscle. Histological analyses and assays of hydroxyproline content indicated that the loss of KLF10 increased fibrosis. Dystrophic KLF10-null mice also had reduced grip strength. The effects of loss of KLF10 gene expression were most pronounced in dystrophic diaphragm muscle, suggesting that KLF10 moderates the fibrotic effects of TGF-β signaling in chronically damaged regenerating muscle.
Collapse
Affiliation(s)
- Joseph X DiMario
- Department of Cell Biology and Anatomy, Rosalind Franklin University of Medicine and Science, Chicago Medical School, North Chicago, Illinois.
| |
Collapse
|
29
|
Jin Q, Qiao C, Li J, Li J, Xiao X. Neonatal Systemic AAV-Mediated Gene Delivery of GDF11 Inhibits Skeletal Muscle Growth. Mol Ther 2018; 26:1109-1117. [PMID: 29503194 DOI: 10.1016/j.ymthe.2018.01.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 01/10/2018] [Accepted: 01/16/2018] [Indexed: 12/19/2022] Open
Abstract
Growth and differentiation factor 11 (GDF11; BMP11) is a circulating cytokine in the transforming growth factor beta (TGF-β) superfamily. Treatment with recombinant GDF11 (rGDF11) protein has previously been shown to reverse skeletal muscle dysfunction in aged mice. However, the actions of GDF11 in skeletal muscle are still not fully understood. Because GDF11 activates the TGF-β-SMAD2/3 pathway, we hypothesized that GDF11 overexpression would inhibit skeletal muscle growth. To test this hypothesis, we generated recombinant adeno-associated virus serotype 9 (AAV9) vectors harboring the gene for either human GDF11 (AAV9-GDF11) or human IgG1 Fc-fused GDF11 propeptide (AAV9-GDF11Pro-Fc-1) to study the effects of GDF11 overexpression or blockade on skeletal muscle growth and function in vivo. After intravenous administration of AAV9-GDF11 into neonatal C57BL/6J mice, we observed sustained limb muscle growth inhibition along with reductions in forelimb grip strength and treadmill running endurance at 16 weeks. Conversely, treatment with AAV9-GDF11Pro-Fc-1 led to increased limb muscle mass and forelimb grip strength after 28 weeks, although a difference in the total body mass/muscle mass ratio was not observed between treatment and control groups. In sum, our results suggest GDF11 overexpression has an inhibitory effect on skeletal muscle growth.
Collapse
Affiliation(s)
- Quan Jin
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chunping Qiao
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jianbin Li
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Juan Li
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xiao Xiao
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
30
|
Past, Present, and Future Perspective of Targeting Myostatin and Related Signaling Pathways to Counteract Muscle Atrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1088:153-206. [DOI: 10.1007/978-981-13-1435-3_8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
31
|
Quattrocelli M, Capote J, Ohiri JC, Warner JL, Vo AH, Earley JU, Hadhazy M, Demonbreun AR, Spencer MJ, McNally EM. Genetic modifiers of muscular dystrophy act on sarcolemmal resealing and recovery from injury. PLoS Genet 2017; 13:e1007070. [PMID: 29065150 PMCID: PMC5669489 DOI: 10.1371/journal.pgen.1007070] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 11/03/2017] [Accepted: 10/11/2017] [Indexed: 12/17/2022] Open
Abstract
Genetic disruption of the dystrophin complex produces muscular dystrophy characterized by a fragile muscle plasma membrane leading to excessive muscle degeneration. Two genetic modifiers of Duchenne Muscular Dystrophy implicate the transforming growth factor β (TGFβ) pathway, osteopontin encoded by the SPP1 gene and latent TGFβ binding protein 4 (LTBP4). We now evaluated the functional effect of these modifiers in the context of muscle injury and repair to elucidate their mechanisms of action. We found that excess osteopontin exacerbated sarcolemmal injury, and correspondingly, that loss of osteopontin reduced injury extent both in isolated myofibers and in muscle in vivo. We found that ablation of osteopontin was associated with reduced expression of TGFβ and TGFβ-associated pathways. We identified that increased TGFβ resulted in reduced expression of Anxa1 and Anxa6, genes encoding key components of the muscle sarcolemma resealing process. Genetic manipulation of Ltbp4 in dystrophic muscle also directly modulated sarcolemmal resealing, and Ltbp4 alleles acted in concert with Anxa6, a distinct modifier of muscular dystrophy. These data provide a model in which a feed forward loop of TGFβ and osteopontin directly impacts the capacity of muscle to recover from injury, and identifies an intersection of genetic modifiers on muscular dystrophy.
Collapse
MESH Headings
- Animals
- Annexin A1/genetics
- Annexin A1/metabolism
- Annexin A6/genetics
- Annexin A6/metabolism
- Female
- Gene Expression Regulation
- Genes, Modifier
- Latent TGF-beta Binding Proteins/physiology
- Male
- Mice
- Mice, Inbred DBA
- Mice, Knockout
- Muscle, Skeletal/injuries
- Muscle, Skeletal/physiology
- Muscular Dystrophy, Animal/genetics
- Muscular Dystrophy, Animal/metabolism
- Muscular Dystrophy, Animal/pathology
- Osteopontin/genetics
- Osteopontin/metabolism
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Recovery of Function
- Sarcolemma/physiology
Collapse
Affiliation(s)
- Mattia Quattrocelli
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Joanna Capote
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Joyce C. Ohiri
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - James L. Warner
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Andy H. Vo
- Committee on Development, Regeneration, and Stem Cell Biology, The University of Chicago, Chicago, Illinois, United States of America
| | - Judy U. Earley
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Michele Hadhazy
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Alexis R. Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Melissa J. Spencer
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Elizabeth M. McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| |
Collapse
|
32
|
Khalil H, Kanisicak O, Prasad V, Correll RN, Fu X, Schips T, Vagnozzi RJ, Liu R, Huynh T, Lee SJ, Karch J, Molkentin JD. Fibroblast-specific TGF-β-Smad2/3 signaling underlies cardiac fibrosis. J Clin Invest 2017; 127:3770-3783. [PMID: 28891814 DOI: 10.1172/jci94753] [Citation(s) in RCA: 619] [Impact Index Per Article: 77.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 07/26/2017] [Indexed: 02/06/2023] Open
Abstract
The master cytokine TGF-β mediates tissue fibrosis associated with inflammation and tissue injury. TGF-β induces fibroblast activation and differentiation into myofibroblasts that secrete extracellular matrix proteins. Canonical TGF-β signaling mobilizes Smad2 and Smad3 transcription factors that control fibrosis by promoting gene expression. However, the importance of TGF-β-Smad2/3 signaling in fibroblast-mediated cardiac fibrosis has not been directly evaluated in vivo. Here, we examined pressure overload-induced cardiac fibrosis in fibroblast- and myofibroblast-specific inducible Cre-expressing mouse lines with selective deletion of the TGF-β receptors Tgfbr1/2, Smad2, or Smad3. Fibroblast-specific deletion of Tgfbr1/2 or Smad3, but not Smad2, markedly reduced the pressure overload-induced fibrotic response as well as fibrosis mediated by a heart-specific, latency-resistant TGF-β mutant transgene. Interestingly, cardiac fibroblast-specific deletion of Tgfbr1/2, but not Smad2/3, attenuated the cardiac hypertrophic response to pressure overload stimulation. Mechanistically, loss of Smad2/3 from tissue-resident fibroblasts attenuated injury-induced cellular expansion within the heart and the expression of fibrosis-mediating genes. Deletion of Smad2/3 or Tgfbr1/2 from cardiac fibroblasts similarly inhibited the gene program for fibrosis and extracellular matrix remodeling, although deletion of Tgfbr1/2 uniquely altered expression of an array of regulatory genes involved in cardiomyocyte homeostasis and disease compensation. These findings implicate TGF-β-Smad2/3 signaling in activated tissue-resident cardiac fibroblasts as principal mediators of the fibrotic response.
Collapse
Affiliation(s)
- Hadi Khalil
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Onur Kanisicak
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Vikram Prasad
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Robert N Correll
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Xing Fu
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Tobias Schips
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Ronald J Vagnozzi
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Ruijie Liu
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA.,Department of Biomedical Sciences, Grand Valley State University, Allendale, Michigan, USA
| | - Thanh Huynh
- Department of Molecular Biology and Genetics, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Se-Jin Lee
- Department of Molecular Biology and Genetics, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jason Karch
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jeffery D Molkentin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA.,Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
33
|
Quattrocelli M, Salamone IM, Page PG, Warner JL, Demonbreun AR, McNally EM. Intermittent Glucocorticoid Dosing Improves Muscle Repair and Function in Mice with Limb-Girdle Muscular Dystrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2520-2535. [PMID: 28823869 DOI: 10.1016/j.ajpath.2017.07.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/03/2017] [Accepted: 07/13/2017] [Indexed: 12/17/2022]
Abstract
The muscular dystrophies are genetically diverse. Shared pathological features among muscular dystrophies include breakdown, or loss of muscle, and accompanying fibrotic replacement. Novel strategies are needed to enhance muscle repair and function and to slow this pathological remodeling. Glucocorticoid steroids, like prednisone, are known to delay loss of ambulation in patients with Duchenne muscular dystrophy but are accompanied by prominent adverse effects. However, less is known about the effects of steroid administration in other types of muscular dystrophies, including limb-girdle muscular dystrophies (LGMDs). LGMD 2B is caused by loss of dysferlin, a membrane repair protein, and LGMD 2C is caused by loss of the dystrophin-associated protein, γ-sarcoglycan. Herein, we assessed the efficacy of steroid dosing on sarcolemmal repair, muscle function, histopathology, and the regenerative capacity of primary muscle cells. We found that in murine models of LGMD 2B and 2C, daily prednisone dosing reduced muscle damage and fibroinflammatory infiltration. However, daily prednisone dosing also correlated with increased muscle adipogenesis and atrophic remodeling. Conversely, intermittent dosing of prednisone, provided once weekly, enhanced muscle repair and did not induce atrophy or adipogenesis, and was associated with improved muscle function. These data indicate that dosing frequency of glucocorticoid steroids affects muscle remodeling in non-Duchenne muscular dystrophies, suggesting a positive outcome associated with intermittent steroid dosing in LGMD 2B and 2C muscle.
Collapse
Affiliation(s)
- Mattia Quattrocelli
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Isabella M Salamone
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Patrick G Page
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - James L Warner
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Alexis R Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
34
|
Mahdy MAA, Warita K, Hosaka YZ. Effects of transforming growth factor-β1 treatment on muscle regeneration and adipogenesis in glycerol-injured muscle. Anim Sci J 2017; 88:1811-1819. [PMID: 28585769 DOI: 10.1111/asj.12845] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 04/03/2017] [Indexed: 12/27/2022]
Abstract
Transforming growth factor (TGF)-β1 is associated with fibrosis in many organs. Recent studies demonstrated that delivery of TGF-β1 into chemically injured muscle enhances fibrosis. In this study, we investigated the effects of exogenous TGF-β1 on muscle regeneration and adipogenesis in glycerol-injured muscle of normal mice. Tibialis anterior (TA) muscles were injured by glycerol injection. TGF-β1 was either co-injected with glycerol, as an 'early treatment' group, or injected at day 4 after glycerol, as a 'late treatment' group and the TA muscles were collected at day 7 after initial injury. Myotube density was significantly lower in the early treatment group than in the glycerol-injured group (without TGF-β1 treatment). Moreover, the Oil red O-positive area was significantly smaller in the early treatment group than in the late treatment group and glycerol-injured group. Furthermore, TGF-β1 treatment increased endomysial fibrosis and induced immunostaining of α-smooth muscle actin. The greater inhibitory effects of early TGF-β1 treatment than that of late TGF-β1 treatment during regeneration in glycerol-injured muscle suggest a more potent effect of TGF-β1 on the initial stage of muscle regeneration and adipogenesis. Combination of TGF-β1 with glycerol might be an alternative to enhance muscle fibrosis for future studies.
Collapse
Affiliation(s)
- Mohamed A A Mahdy
- Laboratory of Basic Veterinary Science, United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan.,Department of Veterinary Anatomy, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Katsuhiko Warita
- Laboratory of Basic Veterinary Science, United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan.,Department of Veterinary Anatomy, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Yoshinao Z Hosaka
- Laboratory of Basic Veterinary Science, United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan.,Department of Veterinary Anatomy, Faculty of Agriculture, Tottori University, Tottori, Japan
| |
Collapse
|
35
|
Fibrosis development in early-onset muscular dystrophies: Mechanisms and translational implications. Semin Cell Dev Biol 2017; 64:181-190. [DOI: 10.1016/j.semcdb.2016.09.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 09/22/2016] [Accepted: 09/22/2016] [Indexed: 02/06/2023]
|
36
|
Quattrocelli M, Spencer MJ, McNally EM. Outside in: The matrix as a modifier of muscular dystrophy. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2017; 1864:572-579. [PMID: 28011285 PMCID: PMC5262521 DOI: 10.1016/j.bbamcr.2016.12.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/14/2016] [Accepted: 12/19/2016] [Indexed: 02/07/2023]
Abstract
Muscular dystrophies are genetic conditions leading to muscle degeneration and often, impaired regeneration. Duchenne Muscular Dystrophy is a prototypical form of muscular dystrophy, and like other forms of genetically inherited muscle diseases, pathological progression is variable. Variability in muscular dystrophy can arise from differences in the manner in which the primary mutation impacts the affected protein's function; however, clinical heterogeneity also derives from secondary mutations in other genes that can enhance or reduce pathogenic features of disease. These genes, called genetic modifiers, regulate the pathophysiological context of dystrophic degeneration and regeneration. Understanding the mechanistic links between genetic modifiers and dystrophic progression sheds light on pathologic remodeling, and provides novel avenues to therapeutically intervene to reduce muscle degeneration. Based on targeted genetic approaches and unbiased genomewide screens, several modifiers have been identified for muscular dystrophy, including extracellular agonists of signaling cascades. This review will focus on identification and possible mechanisms of recently identified modifiers for muscular dystrophy, including osteopontin, latent TGFβ binding protein 4 (LTBP4) and Jagged1. Moreover, we will review the investigational approaches that aim to target modifier pathways and thereby counteract dystrophic muscle wasting.
Collapse
Affiliation(s)
| | - Melissa J Spencer
- Dept of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | |
Collapse
|
37
|
Delaney K, Kasprzycka P, Ciemerych MA, Zimowska M. The role of TGF-β1 during skeletal muscle regeneration. Cell Biol Int 2017; 41:706-715. [PMID: 28035727 DOI: 10.1002/cbin.10725] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 12/26/2016] [Indexed: 02/06/2023]
Abstract
The injury of adult skeletal muscle initiates series of well-coordinated events that lead to the efficient repair of the damaged tissue. Any disturbances during muscle myolysis or reconstruction may result in the unsuccessful regeneration, characterised by strong inflammatory response and formation of connective tissue, that is, fibrosis. The switch between proper regeneration of skeletal muscle and development of fibrosis is controlled by various factors. Amongst them are those belonging to the transforming growth factor β family. One of the TGF-β family members is TGF-β1, a multifunctional cytokine involved in the regulation of muscle repair via satellite cells activation, connective tissue formation, as well as regulation of the immune response intensity. Here, we present the role of TGF-β1 in myogenic differentiation and muscle repair. The understanding of the mechanisms controlling these processes can contribute to the better understanding of skeletal muscle atrophy and diseases which consequence is fibrosis disrupting muscle function.
Collapse
Affiliation(s)
- Kamila Delaney
- Faculty of Biology, Department of Cytology, Institute of Zoology, University of Warsaw, 1 Miecznikowa St., 02-096 Warsaw, Poland
| | - Paulina Kasprzycka
- Faculty of Biology, Department of Cytology, Institute of Zoology, University of Warsaw, 1 Miecznikowa St., 02-096 Warsaw, Poland
| | - Maria Anna Ciemerych
- Faculty of Biology, Department of Cytology, Institute of Zoology, University of Warsaw, 1 Miecznikowa St., 02-096 Warsaw, Poland
| | - Malgorzata Zimowska
- Faculty of Biology, Department of Cytology, Institute of Zoology, University of Warsaw, 1 Miecznikowa St., 02-096 Warsaw, Poland
| |
Collapse
|
38
|
Chadwick JA, Bhattacharya S, Lowe J, Weisleder N, Rafael-Fortney JA. Renin-angiotensin-aldosterone system inhibitors improve membrane stability and change gene-expression profiles in dystrophic skeletal muscles. Am J Physiol Cell Physiol 2016; 312:C155-C168. [PMID: 27881412 PMCID: PMC5336592 DOI: 10.1152/ajpcell.00269.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/21/2016] [Accepted: 11/22/2016] [Indexed: 01/16/2023]
Abstract
Angiotensin-converting enzyme inhibitors (ACEi) and mineralocorticoid receptor (MR) antagonists are FDA-approved drugs that inhibit the renin-angiotensin-aldosterone system (RAAS) and are used to treat heart failure. Combined treatment with the ACEi lisinopril and the nonspecific MR antagonist spironolactone surprisingly improves skeletal muscle, in addition to heart function and pathology in a Duchenne muscular dystrophy (DMD) mouse model. We recently demonstrated that MR is present in all limb and respiratory muscles and functions as a steroid hormone receptor in differentiated normal human skeletal muscle fibers. The goals of the current study were to begin to define cellular and molecular mechanisms mediating the skeletal muscle efficacy of RAAS inhibitor treatment. We also compared molecular changes resulting from RAAS inhibition with those resulting from the current DMD standard-of-care glucocorticoid treatment. Direct assessment of muscle membrane integrity demonstrated improvement in dystrophic mice treated with lisinopril and spironolactone compared with untreated mice. Short-term treatments of dystrophic mice with specific and nonspecific MR antagonists combined with lisinopril led to overlapping gene-expression profiles with beneficial regulation of metabolic processes and decreased inflammatory gene expression. Glucocorticoids increased apoptotic, proteolytic, and chemokine gene expression that was not changed by RAAS inhibitors in dystrophic mice. Microarray data identified potential genes that may underlie RAAS inhibitor treatment efficacy and the side effects of glucocorticoids. Direct effects of RAAS inhibitors on membrane integrity also contribute to improved pathology of dystrophic muscles. Together, these data will inform clinical development of MR antagonists for treating skeletal muscles in DMD.
Collapse
Affiliation(s)
- Jessica A Chadwick
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio; and
| | - Sayak Bhattacharya
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio; and.,Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Jeovanna Lowe
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio; and
| | - Noah Weisleder
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio; and.,Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Jill A Rafael-Fortney
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio; and
| |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW Recently, genetic pathways that modify the clinical severity of Duchenne muscular dystrophy (DMD) have been identified. The pathways uncovered as modifiers are useful to predict prognosis and also elucidate molecular signatures that can be manipulated therapeutically. RECENT FINDINGS Modifiers have been identified using combinations of transcriptome and genome profiling. Osteopontin, encoded by the SPP1 gene, was found using gene expression profiling. Latent TGFβ binding protein 4, encoding latent TGFβ binding protein 4 was initially discovered using a genome-wide screen in mice and then validated in cohorts of DMD patients. These two pathways converge in that they both regulate TGFβ. A third modifier, Anxa6 that specifies annexin A6, is a calcium binding protein that has been identified using mouse models, and regulates the injury pathway and sarcolemmal resealing. SUMMARY Genetic modifiers can serve as biomarkers for outcomes in DMD. Modifiers can alter strength and ambulation in muscular dystrophy, and these same features can be used as endpoints used in clinical trials. Moreover, because genetic modifiers can influence outcomes, these genetic markers should be considered when stratifying results in muscular dystrophy.
Collapse
Affiliation(s)
- Andy H Vo
- Committee on Development, Regeneration and Stem Cell Biology, Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, The University of Chicago, Chicago, Illinois, USA
| | | |
Collapse
|
40
|
Korneenko TV, Pestov NB, Ahmad N, Okkelman IA, Dmitriev RI, Shakhparonov MI, Modyanov NN. Evolutionary diversification of the BetaM interactome acquired through co-option of the ATP1B4 gene in placental mammals. Sci Rep 2016; 6:22395. [PMID: 26939788 PMCID: PMC4778017 DOI: 10.1038/srep22395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 02/11/2016] [Indexed: 11/16/2022] Open
Abstract
ATP1B4 genes represent a rare instance of orthologous vertebrate gene co-option that radically changed properties of the encoded BetaM proteins, which function as Na,K-ATPase subunits in lower vertebrates and birds. Eutherian BetaM has lost its ancestral function and became a muscle-specific resident of the inner nuclear membrane. Our earlier work implicated BetaM in regulation of gene expression through direct interaction with the transcriptional co-regulator SKIP. To gain insight into evolution of BetaM interactome we performed expanded screening of eutherian and avian cDNA libraries using yeast-two-hybrid and split-ubiquitin systems. The inventory of identified BetaM interactors includes lamina-associated protein LAP-1, myocyte nuclear envelope protein Syne1, BetaM itself, heme oxidases HMOX1 and HMOX2; transcription factor LZIP/CREB3, ERGIC3, PHF3, reticulocalbin-3, and β-sarcoglycan. No new interactions were found for chicken BetaM and human Na,K-ATPase β1, β2 and β3 isoforms, indicating the uniqueness of eutherian BetaM interactome. Analysis of truncated forms of BetaM indicates that residues 72-98 adjacent to the membrane in nucleoplasmic domain are important for the interaction with SKIP. These findings demonstrate that evolutionary alterations in structural and functional properties of eutherian BetaM proteins are associated with the increase in its interactome complexity.
Collapse
Affiliation(s)
- Tatyana V Korneenko
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117871, Russia.,Department of Physiology and Pharmacology and Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, 3000 Arlington Ave, Toledo, OH 43614, USA
| | - Nikolay B Pestov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117871, Russia.,Department of Physiology and Pharmacology and Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, 3000 Arlington Ave, Toledo, OH 43614, USA
| | - Nisar Ahmad
- Department of Physiology and Pharmacology and Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, 3000 Arlington Ave, Toledo, OH 43614, USA
| | - Irina A Okkelman
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117871, Russia
| | - Ruslan I Dmitriev
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117871, Russia
| | | | - Nikolai N Modyanov
- Department of Physiology and Pharmacology and Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, 3000 Arlington Ave, Toledo, OH 43614, USA
| |
Collapse
|
41
|
Lamar KM, Miller T, Dellefave-Castillo L, McNally EM. Genotype-Specific Interaction of Latent TGFβ Binding Protein 4 with TGFβ. PLoS One 2016; 11:e0150358. [PMID: 26918958 PMCID: PMC4769137 DOI: 10.1371/journal.pone.0150358] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 02/12/2016] [Indexed: 01/06/2023] Open
Abstract
Latent TGFβ binding proteins are extracellular matrix proteins that bind latent TGFβ to form the large latent complex. Nonsynonymous polymorphisms in LTBP4, a member of the latent TGFβ binding protein gene family, have been linked to several human diseases, underscoring the importance of TGFβ regulation for a range of phenotypes. Because of strong linkage disequilibrium across the LTBP4 gene, humans have two main LTBP4 alleles that differ at four amino acid positions, referred to as IAAM and VTTT for the encoded residues. VTTT is considered the “risk” allele and associates with increased intracellular TGFβ signaling and more deleterious phenotypes in muscular dystrophy and other diseases. We now evaluated LTBP4 nsSNPs in dilated cardiomyopathy, a distinct disorder associated with TGFβ signaling. We stratified based on self-identified ethnicity and found that the LTBP4 VTTT allele is associated with increased risk of dilated cardiomyopathy in European Americans extending the diseases that associate with LTBP4 genotype. However, the association of LTBP4 SNPs with dilated cardiomyopathy was not observed in African Americans. To elucidate the mechanism by which LTBP4 genotype exerts this differential effect, TGFβ’s association with LTBP4 protein was examined. LTBP4 protein with the IAAM residues bound more latent TGFβ compared to the LTBP4 VTTT protein. Together these data provide support that LTBP4 genotype exerts its effect through differential avidity for TGFβ accounting for the differences in TGFβ signaling attributed to these two alleles.
Collapse
Affiliation(s)
- Kay-Marie Lamar
- Department of Human Genetics, The University of Chicago, Chicago, Illinois, United States of America
| | - Tamari Miller
- Department of Human Genetics, The University of Chicago, Chicago, Illinois, United States of America
| | - Lisa Dellefave-Castillo
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Elizabeth M. McNally
- Department of Human Genetics, The University of Chicago, Chicago, Illinois, United States of America
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
42
|
de Greef JC, Hamlyn R, Jensen BS, O'Campo Landa R, Levy JR, Kobuke K, Campbell KP. Collagen VI deficiency reduces muscle pathology, but does not improve muscle function, in the γ-sarcoglycan-null mouse. Hum Mol Genet 2016; 25:1357-69. [PMID: 26908621 PMCID: PMC4787905 DOI: 10.1093/hmg/ddw018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 01/18/2016] [Indexed: 01/19/2023] Open
Abstract
Muscular dystrophy is characterized by progressive skeletal muscle weakness and dystrophic muscle exhibits degeneration and regeneration of muscle cells, inflammation and fibrosis. Skeletal muscle fibrosis is an excessive deposition of components of the extracellular matrix including an accumulation of Collagen VI. We hypothesized that a reduction of Collagen VI in a muscular dystrophy model that presents with fibrosis would result in reduced muscle pathology and improved muscle function. To test this hypothesis, we crossed γ-sarcoglycan-null mice, a model of limb-girdle muscular dystrophy type 2C, with a Col6a2-deficient mouse model. We found that the resulting γ-sarcoglycan-null/Col6a2Δex5 mice indeed exhibit reduced muscle pathology compared with γ-sarcoglycan-null mice. Specifically, fewer muscle fibers are degenerating, fiber size varies less, Evans blue dye uptake is reduced and serum creatine kinase levels are lower. Surprisingly, in spite of this reduction in muscle pathology, muscle function is not significantly improved. In fact, grip strength and maximum isometric tetanic force are even lower in γ-sarcoglycan-null/Col6a2Δex5 mice than in γ-sarcoglycan-null mice. In conclusion, our results reveal that Collagen VI-mediated fibrosis contributes to skeletal muscle pathology in γ-sarcoglycan-null mice. Importantly, however, our data also demonstrate that a reduction in skeletal muscle pathology does not necessarily lead to an improvement of skeletal muscle function, and this should be considered in future translational studies.
Collapse
Affiliation(s)
- Jessica C de Greef
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology and Department of Internal Medicine, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242, USA
| | - Rebecca Hamlyn
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology and Department of Internal Medicine, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242, USA
| | - Braden S Jensen
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology and Department of Internal Medicine, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242, USA
| | - Raul O'Campo Landa
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology and Department of Internal Medicine, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242, USA
| | - Jennifer R Levy
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology and Department of Internal Medicine, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242, USA
| | - Kazuhiro Kobuke
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology and Department of Internal Medicine, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242, USA
| | - Kevin P Campbell
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology and Department of Internal Medicine, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242, USA
| |
Collapse
|
43
|
Tjondrokoesoemo A, Schips T, Kanisicak O, Sargent MA, Molkentin JD. Genetic overexpression of Serpina3n attenuates muscular dystrophy in mice. Hum Mol Genet 2016; 25:1192-202. [PMID: 26744329 DOI: 10.1093/hmg/ddw005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 01/05/2016] [Indexed: 01/06/2023] Open
Abstract
Muscular dystrophy (MD) is associated with mutations in genes that stabilize the myofiber plasma membrane, such as through the dystrophin-glycoprotein complex (DGC). Instability of this complex or defects in membrane repair/integrity leads to calcium influx and myofiber necrosis leading to progressive dystrophic disease. MD pathogenesis is also associated with increased skeletal muscle protease levels and activity that could augment weakening of the sarcolemma through greater degradation of cellular attachment complexes. Here, we observed a compensatory increase in the serine protease inhibitor Serpina3n in mouse models of MD and after acute muscle tissue injury. Serpina3n muscle-specific transgenic mice were generated to model this increase in expression, which reduced the activity of select proteases in dystrophic skeletal muscle and protected muscle from both acute injury with cardiotoxin and from chronic muscle disease in the mdx or Sgcd(-/-) MD genetic backgrounds. The Serpina3n transgene mitigated muscle degeneration and fibrosis, reduced creatine kinase serum levels, restored running capacity on a treadmill and reduced muscle membrane leakiness in vivo that is characteristic of mdx and Sgcd(-/-) mice. Mechanistically, we show that increased Serpina3n promotes greater sarcolemma membrane integrity and stability in dystrophic mouse models in association with increased membrane residence of the integrins, the DGC/utrophin-glycoprotein complex of proteins and annexin A1. Hence, Serpina3n blocks endogenous increases in the activity of select skeletal muscle resident proteases during injury or dystrophic disease, which stabilizes the sarcolemma leading to less myofiber degeneration and increased regeneration. These results suggest the use of select protease inhibitors as a strategy for treating MD.
Collapse
Affiliation(s)
| | - Tobias Schips
- Department of Pediatrics, University of Cincinnati and
| | | | | | - Jeffery D Molkentin
- Department of Pediatrics, University of Cincinnati and Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way, MLC7020, Cincinnati, OH 45229, USA
| |
Collapse
|
44
|
Moyer AL, Wagner KR. Mammalian Mss51 is a skeletal muscle-specific gene modulating cellular metabolism. J Neuromuscul Dis 2015; 2:371-385. [PMID: 26634192 PMCID: PMC4664537 DOI: 10.3233/jnd-150119] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND The transforming growth factor β (TGF-β) signaling pathways modulate skeletal muscle growth, regeneration, and cellular metabolism. Several recent gene expression studies have shown that inhibition of myostatin and TGF-β1 signaling consistently leads to a significant reduction in expression of Mss51, also named Zmynd17. The function of mammalian Mss51 is unknown although a putative homolog in yeast is a mitochondrial translational activator. OBJECTIVE The objective of this work was to characterize mammalian Mss51. METHODS Quantitative RT-PCR and immunoblot of subcellular fractionation were used to determine expression patterns and localization of Mss51. The CRISPR/Cas9 system was used to reduce expression of Mss51 in C2C12 myoblasts and the function of Mss51 was evaluated in assays of proliferation, differentiation and cellular metabolism. RESULTS Mss51 was predominantly expressed in skeletal muscle and in those muscles dominated by fast-twitch fibers. In vitro, its expression was upregulated upon differentiation of C2C12 myoblasts into myotubes. Expression of Mss51 was modulated in response to altered TGF-β family signaling. In human muscle, Mss51 localized to the mitochondria. Its genetic disruption resulted in increased levels of cellular ATP, β-oxidation, glycolysis, and oxidative phosphorylation. CONCLUSIONS Mss51 is a novel, skeletal muscle-specific gene and a key target of myostatin and TGF-β1 signaling. Unlike myostatin, TGF-β1 and IGF-1, Mss51 does not regulate myoblast proliferation or differentiation. Rather, Mss51 appears to be one of the effectors of these growth factors on metabolic processes including fatty acid oxidation, glycolysis and oxidative phosphorylation.
Collapse
Affiliation(s)
- Adam L. Moyer
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kathryn R. Wagner
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|