1
|
Saeed M, Haque A, Shoaib A, Danish Rizvi SM. Exploring novel natural compound-based therapies for Duchenne muscular dystrophy management: insights from network pharmacology, QSAR modeling, molecular dynamics, and free energy calculations. Front Pharmacol 2024; 15:1395014. [PMID: 39415830 PMCID: PMC11481126 DOI: 10.3389/fphar.2024.1395014] [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: 03/02/2024] [Accepted: 05/31/2024] [Indexed: 10/19/2024] Open
Abstract
Muscular dystrophies encompass a heterogeneous group of rare neuromuscular diseases characterized by progressive muscle degeneration and weakness. Among these, Duchenne muscular dystrophy (DMD) stands out as one of the most severe forms. The present study employs an integrative approach combining network pharmacology, quantitative structure-activity relationship (QSAR) modeling, molecular dynamics (MD) simulations, and free energy calculations to identify potential therapeutic targets and natural compounds for DMD. Upon analyzing the GSE38417 dataset, it was found that individuals with DMD exhibited 290 upregulated differentially expressed genes (DEGs) compared to healthy controls. By utilizing gene ontology (GO) and protein-protein interaction (PPI) network analysis, this study provides insights into the functional roles of the identified DEGs, identifying ten hub genes that play a critical role in the pathology of DMD. These key genes include DMD, TTN, PLEC, DTNA, PKP2, SLC24A, FBXO32, SNTA1, SMAD3, and NOS1. Furthermore, through the use of ligand-based pharmacophore modeling and virtual screening, three natural compounds were identified as potential inhibitors. Among these, compounds 3874518 and 12314417 have demonstrated significant promise as an inhibitor of the SMAD3 protein, a crucial factor in the fibrotic and inflammatory mechanisms associated with DMD. The therapeutic potential of the compounds was further supported by molecular dynamics simulation and Molecular Mechanics/Generalized Born Surface Area (MM/GBSA) analysis. These findings suggest that the compounds are viable candidates for experimental validation against DMD.
Collapse
Affiliation(s)
- Mohd Saeed
- Department of Biology, College of Sciences, University of Ha’il, Ha’il, Saudi Arabia
| | - Ashanul Haque
- Department of Chemistry, College of Sciences, University of Ha’il, Ha’il, Saudi Arabia
| | - Ambreen Shoaib
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Syed Mohd Danish Rizvi
- Department of Pharmaceutics, College of Pharmacy, University of Ha’il, Ha’il, Saudi Arabia
| |
Collapse
|
2
|
Li N, Xiahou Z, Li Z, Zhang Z, Song Y, Wang Y. Identification of hub genes and therapeutic siRNAs to develop novel adjunctive therapy for Duchenne muscular dystrophy. BMC Musculoskelet Disord 2024; 25:386. [PMID: 38762732 PMCID: PMC11102231 DOI: 10.1186/s12891-024-07206-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/15/2024] [Indexed: 05/20/2024] Open
Abstract
OBJECTIVE Duchenne muscular dystrophy (DMD) is a devastating X-linked neuromuscular disorder caused by various defects in the dystrophin gene and still no universal therapy. This study aims to identify the hub genes unrelated to excessive immune response but responsible for DMD progression and explore therapeutic siRNAs, thereby providing a novel treatment. METHODS Top ten hub genes for DMD were identified from GSE38417 dataset by using GEO2R and PPI networks based on Cytoscape analysis. The hub genes unrelated to excessive immune response were identified by GeneCards, and their expression was further verified in mdx and C57 mice at 2 and 4 months (M) by (RT-q) PCR and western blotting. Therapeutic siRNAs were deemed as those that could normalize the expression of the validated hub genes in transfected C2C12 cells. RESULTS 855 up-regulated and 324 down-regulated DEGs were screened from GSE38417 dataset. Five of the top 10 hub genes were considered as the candidate genes unrelated to excessive immune response, and three of these candidates were consistently and significantly up-regulated in mdx mice at 2 M and 4 M when compared with age-matched C57 mice, including Col1a2, Fbn1 and Fn1. Furthermore, the three validated up-regulated candidate genes can be significantly down-regulated by three rational designed siRNA (p < 0.0001), respectively. CONCLUSION COL1A2, FBN1 and FN1 may be novel biomarkers for DMD, and the siRNAs designed in our study were help to develop adjunctive therapy for Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Na Li
- Department of Aerospace Medical Training, School of Aerospace Medicine, Air Force Medical University, Xi'an, China
- School of Sports Science, Beijing Sport University, Beijing, China
| | - Zhikai Xiahou
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| | - Zhuo Li
- School of Sports Science, Beijing Sport University, Beijing, China
| | - Zilian Zhang
- School of Sports Science, Beijing Sport University, Beijing, China
| | - Yafeng Song
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China.
| | - Yongchun Wang
- Department of Aerospace Medical Training, School of Aerospace Medicine, Air Force Medical University, Xi'an, China.
| |
Collapse
|
3
|
Wang XF, Vigouroux R, Syonov M, Baglaenko Y, Nikolakopoulou AM, Ringuette D, Rus H, DiStefano PV, Dufour S, Shabanzadeh AP, Lee S, Mueller BK, Charish J, Harada H, Fish JE, Wither J, Wälchli T, Cloutier JF, Zlokovic BV, Carlen PL, Monnier PP. The liver and muscle secreted HFE2-protein maintains central nervous system blood vessel integrity. Nat Commun 2024; 15:1037. [PMID: 38310100 PMCID: PMC10838306 DOI: 10.1038/s41467-024-45303-1] [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: 09/15/2022] [Accepted: 01/19/2024] [Indexed: 02/05/2024] Open
Abstract
Liver failure causes breakdown of the Blood CNS Barrier (BCB) leading to damages of the Central-Nervous-System (CNS), however the mechanisms whereby the liver influences BCB-integrity remain elusive. One possibility is that the liver secretes an as-yet to be identified molecule(s) that circulate in the serum to directly promote BCB-integrity. To study BCB-integrity, we developed light-sheet imaging for three-dimensional analysis. We show that liver- or muscle-specific knockout of Hfe2/Rgmc induces BCB-breakdown, leading to accumulation of toxic-blood-derived fibrinogen in the brain, lower cortical neuron numbers, and behavioral deficits in mice. Soluble HFE2 competes with its homologue RGMa for binding to Neogenin, thereby blocking RGMa-induced downregulation of PDGF-B and Claudin-5 in endothelial cells, triggering BCB-disruption. HFE2 administration in female mice with experimental autoimmune encephalomyelitis, a model for multiple sclerosis, prevented paralysis and immune cell infiltration by inhibiting RGMa-mediated BCB alteration. This study has implications for the pathogenesis and potential treatment of diseases associated with BCB-dysfunction.
Collapse
Affiliation(s)
- Xue Fan Wang
- Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard St.,, Toronto, M5T 2O8, ON, Canada
- Institute of Biomedical and Biomaterial Engineering, University of Toronto, 1 King's College circle,, Toronto, M5S 1A8, ON, Canada
| | - Robin Vigouroux
- Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard St.,, Toronto, M5T 2O8, ON, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College circle,, Toronto, M5S 1A8, ON, Canada
| | - Michal Syonov
- Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard St.,, Toronto, M5T 2O8, ON, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College circle,, Toronto, M5S 1A8, ON, Canada
| | - Yuriy Baglaenko
- Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard St.,, Toronto, M5T 2O8, ON, Canada
| | - Angeliki M Nikolakopoulou
- Department of Physiology and Neuroscience, The Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Dene Ringuette
- Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard St.,, Toronto, M5T 2O8, ON, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College circle,, Toronto, M5S 1A8, ON, Canada
| | - Horea Rus
- University of Maryland, School of Medicine, Department of Neurology, Baltimore, MD, 21201, USA
| | - Peter V DiStefano
- Toronto General Hospital Research Institute, University Health Network, 101 College St. Rm 3-308, Toronto, M5L 1L7, ON, Canada
| | - Suzie Dufour
- Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard St.,, Toronto, M5T 2O8, ON, Canada
| | - Alireza P Shabanzadeh
- Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard St.,, Toronto, M5T 2O8, ON, Canada
| | - Seunggi Lee
- Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard St.,, Toronto, M5T 2O8, ON, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College circle,, Toronto, M5S 1A8, ON, Canada
| | | | - Jason Charish
- Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard St.,, Toronto, M5T 2O8, ON, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College circle,, Toronto, M5S 1A8, ON, Canada
| | - Hidekiyo Harada
- Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard St.,, Toronto, M5T 2O8, ON, Canada
| | - Jason E Fish
- Toronto General Hospital Research Institute, University Health Network, 101 College St. Rm 3-308, Toronto, M5L 1L7, ON, Canada
| | - Joan Wither
- Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard St.,, Toronto, M5T 2O8, ON, Canada
| | - Thomas Wälchli
- Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard St.,, Toronto, M5T 2O8, ON, Canada
- Group of CNS Angiogenesis and Neurovascular Link, and Physician-Scientist Program, Institute for Regenerative Medicine, Neuroscience Center Zurich, and Division of Neurosurgery, University and University Hospital Zurich, Zurich, Switzerland
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University Health Network, Toronto, Canada
| | - Jean-François Cloutier
- The Neuro - Montreal Neurological Institute and Hospital, 3801 Rue Université, Montréal, QC, H3A 2B4, Canada
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, The Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Peter L Carlen
- Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard St.,, Toronto, M5T 2O8, ON, Canada
- Institute of Biomedical and Biomaterial Engineering, University of Toronto, 1 King's College circle,, Toronto, M5S 1A8, ON, Canada
- Department of Physiology and Neuroscience, The Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Philippe P Monnier
- Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard St.,, Toronto, M5T 2O8, ON, Canada.
- Institute of Biomedical and Biomaterial Engineering, University of Toronto, 1 King's College circle,, Toronto, M5S 1A8, ON, Canada.
- Department of Ophthalmology and Vision Sciences, Faculty of Medicine, University of Toronto, 340 College St.,, ON, Toronto, M5T 3A9, Canada.
| |
Collapse
|
4
|
Du G, Zhang P, Guo J, Pang X, Kan G, Zeng B, Chen X, Liang J, Zhan Y. MF-Net: Automated Muscle Fiber Segmentation From Immunofluorescence Images Using a Local-Global Feature Fusion Network. J Digit Imaging 2023; 36:2411-2426. [PMID: 37714969 PMCID: PMC10584774 DOI: 10.1007/s10278-023-00890-1] [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: 06/06/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 09/17/2023] Open
Abstract
Histological assessment of skeletal muscle slices is very important for the accurate evaluation of weightless muscle atrophy. The accurate identification and segmentation of muscle fiber boundary is an important prerequisite for the evaluation of skeletal muscle fiber atrophy. However, there are many challenges to segment muscle fiber from immunofluorescence images, including the presence of low contrast in fiber boundaries in immunofluorescence images and the influence of background noise. Due to the limitations of traditional convolutional neural network-based segmentation methods in capturing global information, they cannot achieve ideal segmentation results. In this paper, we propose a muscle fiber segmentation network (MF-Net) method for effective segmentation of macaque muscle fibers in immunofluorescence images. The network adopts a dual encoder branch composed of convolutional neural networks and transformer to effectively capture local and global feature information in the immunofluorescence image, highlight foreground features, and suppress irrelevant background noise. In addition, a low-level feature decoder module is proposed to capture more global context information by combining different image scales to supplement the missing detail pixels. In this study, a comprehensive experiment was carried out on the immunofluorescence datasets of six macaques' weightlessness models and compared with the state-of-the-art deep learning model. It is proved from five segmentation indices that the proposed automatic segmentation method can be accurately and effectively applied to muscle fiber segmentation in shank immunofluorescence images.
Collapse
Affiliation(s)
| | - Peng Zhang
- China Astronaut Research and Training Center, Beijing, 100094, People's Republic of China
| | - Jianzhong Guo
- Institute of Applied Acoustics, School of Physics and Information Technology, Shaanxi Normal University, Xi'an, 710062, China
| | - Xiangsheng Pang
- China Astronaut Research and Training Center, Beijing, 100094, People's Republic of China
| | - Guanghan Kan
- China Astronaut Research and Training Center, Beijing, 100094, People's Republic of China
| | - Bin Zeng
- China Astronaut Research and Training Center, Beijing, 100094, People's Republic of China
| | - Xiaoping Chen
- China Astronaut Research and Training Center, Beijing, 100094, People's Republic of China.
| | - Jimin Liang
- School of Electronic Engineering, Xidian University, Xi'an, Shaanxi, 710071, China.
| | - Yonghua Zhan
- School of Life Science and Technology, Xidian University & Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an, Shaanxi, 710126, China.
| |
Collapse
|
5
|
Zhang H, Wang F, Pang X, Zhou Y, Li S, Li W, Zhang P, Chen X. Decreased expression of H19/miR-675 ameliorates muscle atrophy by regulating the IGF1R/Akt/FoxO signaling pathway. Mol Med 2023; 29:78. [PMID: 37344807 DOI: 10.1186/s10020-023-00683-w] [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: 01/19/2023] [Accepted: 06/10/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUND Long non-coding RNA (lncRNA) H19 is one of the most highly expressed and conserved transcripts in mammalian development, and its functions have been fully discussed in many contexts including tumorigenesis and skeletal muscle development. However, its exact role in muscle atrophy remains largely unknown. This study investigated the effect of lncRNA H19 on muscle atrophy and the potential underlying mechanism. METHODS Hindlimb suspension (HS) of C57BL/6 mice and starvation of C2C12 cells with PBS were conducted to induce atrophy. Real-time PCR and Western blotting were used to measure the expression of RNAs and proteins. LncRNA H19 and its encoded miR-675 were overexpressed or inhibited in different models of muscle atrophy. Immunofluorescence was carried out to examine the cross-sectional area (CSA) and minimal Feret's diameter (MFD) of myofibers and myotube diameter. RESULTS The expression levels of lncRNA H19 and miR-675 were significantly reduced in both the soleus and gastrocnemius muscles in response to HS. Overexpression of lncRNA H19 led to an increase in Atrogin-1 mRNA expression, and this effect was reversed by inhibiting miR-675. The overexpression of miR-675 aggravated both HS- and starving-induced muscle atrophy by inhibiting the IGF1R/Akt signaling pathway and promoting FoxO/Atrogin-1 expression. Conversely, miR-675 inhibition had the opposite effects. CONCLUSION The lncRNA H19/miR-675 axis can induce muscle atrophy, and its downregulation in mice with HS-induced muscle atrophy may act as a protective mechanism against this condition.
Collapse
Affiliation(s)
- He Zhang
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
- Department of Physical Education, Central South University, Changsha, Hunan, China
| | - Fei Wang
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| | - Xiangsheng Pang
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Yue Zhou
- School of Sport Science, Beijing Sport University, Beijing, China
| | - Shiming Li
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Wenjiong Li
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| | - Peng Zhang
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China.
| | - Xiaoping Chen
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China.
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China.
| |
Collapse
|
6
|
Silvestri L, Pettinato M, Furiosi V, Bavuso Volpe L, Nai A, Pagani A. Managing the Dual Nature of Iron to Preserve Health. Int J Mol Sci 2023; 24:ijms24043995. [PMID: 36835406 PMCID: PMC9961779 DOI: 10.3390/ijms24043995] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/09/2023] [Accepted: 02/15/2023] [Indexed: 02/18/2023] Open
Abstract
Because of its peculiar redox properties, iron is an essential element in living organisms, being involved in crucial biochemical processes such as oxygen transport, energy production, DNA metabolism, and many others. However, its propensity to accept or donate electrons makes it potentially highly toxic when present in excess and inadequately buffered, as it can generate reactive oxygen species. For this reason, several mechanisms evolved to prevent both iron overload and iron deficiency. At the cellular level, iron regulatory proteins, sensors of intracellular iron levels, and post-transcriptional modifications regulate the expression and translation of genes encoding proteins that modulate the uptake, storage, utilization, and export of iron. At the systemic level, the liver controls body iron levels by producing hepcidin, a peptide hormone that reduces the amount of iron entering the bloodstream by blocking the function of ferroportin, the sole iron exporter in mammals. The regulation of hepcidin occurs through the integration of multiple signals, primarily iron, inflammation and infection, and erythropoiesis. These signals modulate hepcidin levels by accessory proteins such as the hemochromatosis proteins hemojuvelin, HFE, and transferrin receptor 2, the serine protease TMPRSS6, the proinflammatory cytokine IL6, and the erythroid regulator Erythroferrone. The deregulation of the hepcidin/ferroportin axis is the central pathogenic mechanism of diseases characterized by iron overload, such as hemochromatosis and iron-loading anemias, or by iron deficiency, such as IRIDA and anemia of inflammation. Understanding the basic mechanisms involved in the regulation of hepcidin will help in identifying new therapeutic targets to treat these disorders.
Collapse
Affiliation(s)
- Laura Silvestri
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milan, Italy
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Correspondence: ; Tel.: +39-0226436889; Fax: +39-0226434723
| | - Mariateresa Pettinato
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Valeria Furiosi
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Letizia Bavuso Volpe
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Antonella Nai
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milan, Italy
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Alessia Pagani
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milan, Italy
| |
Collapse
|
7
|
Duan G, Li J, Duan Y, Zheng C, Guo Q, Li F, Zheng J, Yu J, Zhang P, Wan M, Long C. Mitochondrial Iron Metabolism: The Crucial Actors in Diseases. Molecules 2022; 28:29. [PMID: 36615225 PMCID: PMC9822237 DOI: 10.3390/molecules28010029] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Iron is a trace element necessary for cell growth, development, and cellular homeostasis, but insufficient or excessive level of iron is toxic. Intracellularly, sufficient amounts of iron are required for mitochondria (the center of iron utilization) to maintain their normal physiologic function. Iron deficiency impairs mitochondrial metabolism and respiratory activity, while mitochondrial iron overload promotes ROS production during mitochondrial electron transport, thus promoting potential disease development. This review provides an overview of iron homeostasis, mitochondrial iron metabolism, and how mitochondrial iron imbalances-induced mitochondrial dysfunction contribute to diseases.
Collapse
Affiliation(s)
- Geyan Duan
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianjun Li
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yehui Duan
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Changbing Zheng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Qiuping Guo
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fengna Li
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Zheng
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiayi Yu
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peiwen Zhang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Mengliao Wan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Cimin Long
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
8
|
Tan B, Zeng J, Meng F, Wang S, Xiao L, Zhao X, Hong L, Zheng E, Wu Z, Li Z, Gu T. Comprehensive analysis of pre-mRNA alternative splicing regulated by m6A methylation in pig oxidative and glycolytic skeletal muscles. BMC Genomics 2022; 23:804. [PMID: 36474138 PMCID: PMC9724443 DOI: 10.1186/s12864-022-09043-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Different types of skeletal myofibers exhibit distinct physiological and metabolic properties that are associated with meat quality traits in livestock. Alternative splicing (AS) of pre-mRNA can generate multiple transcripts from an individual gene by differential selection of splice sites. N6-methyladenosine (m6A) is the most abundant modification in mRNAs, but its regulation for AS in different muscles remains unknown. RESULTS: We characterized AS events and m6A methylation pattern in pig oxidative and glycolytic muscles. A tota1 of 1294 differential AS events were identified, and differentially spliced genes were significantly enriched in processes related to different phenotypes between oxidative and glycolytic muscles. We constructed the regulatory network between splicing factors and corresponding differential AS events and identified NOVA1 and KHDRBS2 as key splicing factors. AS event was enriched in m6A-modified genes, and the methylation level was positively correlated with the number of AS events in genes. The dynamic change in m6A enrichment was associated with 115 differentially skipping exon (SE-DAS) events within 92 genes involving in various processes, including muscle contraction and myofibril assembly. We obtained 23.4% SE-DAS events (27/115) regulated by METTL3-meditaed m6A and experimentally validated the aberrant splicing of ZNF280D, PHE4DIP, and NEB. The inhibition of m6A methyltransferase METTL3 could induce the conversion of oxidative fiber to glycolytic fiber in PSCs. CONCLUSION Our study suggested that m6A modification could contribute to significant difference in phenotypes between oxidative and glycolytic muscles by mediating the regulation of AS. These findings would provide novel insights into mechanisms underlying muscle fiber conversion.
Collapse
Affiliation(s)
- Baohua Tan
- grid.20561.300000 0000 9546 5767National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| | - Jiekang Zeng
- grid.20561.300000 0000 9546 5767National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| | - Fanming Meng
- grid.135769.f0000 0001 0561 6611State Key Laboratory of Livestock and Poultry Breeding, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, 510640 Guangzhou, Guangdong People’s Republic of China
| | - Shanshan Wang
- grid.20561.300000 0000 9546 5767National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| | - Liyao Xiao
- grid.20561.300000 0000 9546 5767National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| | - Xinming Zhao
- grid.20561.300000 0000 9546 5767National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| | - Linjun Hong
- grid.20561.300000 0000 9546 5767National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| | - Enqin Zheng
- grid.20561.300000 0000 9546 5767National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| | - Zhenfang Wu
- grid.20561.300000 0000 9546 5767National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, 510642 Guangzhou, China
| | - Zicong Li
- grid.20561.300000 0000 9546 5767National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| | - Ting Gu
- grid.20561.300000 0000 9546 5767National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China ,grid.20561.300000 0000 9546 5767Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642 Guangzhou, China
| |
Collapse
|
9
|
Chen J, Li Z, Zhang Y, Zhang X, Zhang S, Liu Z, Yuan H, Pang X, Liu Y, Tao W, Chen X, Zhang P, Chen GQ. Mechanism of reduced muscle atrophy via ketone body (D)-3-hydroxybutyrate. Cell Biosci 2022; 12:94. [PMID: 35725651 PMCID: PMC9208164 DOI: 10.1186/s13578-022-00826-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/03/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Muscle atrophy is an increasingly global health problem affecting millions, there is a lack of clinical drugs or effective therapy. Excessive loss of muscle mass is the typical characteristic of muscle atrophy, manifesting as muscle weakness accompanied by impaired metabolism of protein and nucleotide. (D)-3-hydroxybutyrate (3HB), one of the main components of the ketone body, has been reported to be effective for the obvious hemodynamic effects in atrophic cardiomyocytes and exerts beneficial metabolic reprogramming effects in healthy muscle. This study aims to exploit how the 3HB exerts therapeutic effects for treating muscle atrophy induced by hindlimb unloaded mice. RESULTS Anabolism/catabolism balance of muscle protein was maintained with 3HB via the Akt/FoxO3a and the mTOR/4E-BP1 pathways; protein homeostasis of 3HB regulation includes pathways of ubiquitin-proteasomal, autophagic-lysosomal, responses of unfolded-proteins, heat shock and anti-oxidation. Metabolomic analysis revealed the effect of 3HB decreased purine degradation and reduced the uric acid in atrophied muscles; enhanced utilization from glutamine to glutamate also provides evidence for the promotion of 3HB during the synthesis of proteins and nucleotides. CONCLUSIONS 3HB significantly inhibits the loss of muscle weights, myofiber sizes and myofiber diameters in hindlimb unloaded mouse model; it facilitates positive balance of proteins and nucleotides with enhanced accumulation of glutamate and decreased uric acid in wasting muscles, revealing effectiveness for treating muscle atrophy.
Collapse
Affiliation(s)
- Jin Chen
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Zihua Li
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yudian Zhang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xu Zhang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Shujie Zhang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Zonghan Liu
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Huimei Yuan
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Xiangsheng Pang
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Yaxuan Liu
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Wuchen Tao
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Xiaoping Chen
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, 100094, China.
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.
| | - Peng Zhang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.
| | - Guo-Qiang Chen
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, China.
- MOE Key Lab of Industrial Biocatalysis, Dept of Chemical Engineering, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
10
|
Transforming Growth Factor-Beta in Skeletal Muscle Wasting. Int J Mol Sci 2022; 23:ijms23031167. [PMID: 35163088 PMCID: PMC8835446 DOI: 10.3390/ijms23031167] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/11/2022] [Accepted: 01/19/2022] [Indexed: 12/21/2022] Open
Abstract
Transforming growth factor-beta (TGF-β) is part of a family of molecules that is present in many body tissues and performs many different functions. Evidence has been obtained from mice and human cancer patients with bony metastases and non-metastatic disease, as well as pediatric burn patients, that inflammation leads to bone resorption and release of TGF-β from the bone matrix with paracrine effects on muscle protein balance, possibly mediated by the generation of reactive oxygen species. Whether immobilization, which confounds the etiology of bone resorption in burn injury, also leads to the release of TGF-β from bone contributing to muscle wasting in other conditions is unclear. The use of anti-resorptive therapy in both metastatic cancer patients and pediatric burn patients has been successful in the prevention of muscle wasting, thereby creating an additional therapeutic niche for this class of drugs. The liberation of TGF-β may be one way in which bone helps to control muscle mass, but further investigation will be necessary to assess whether the rate of bone resorption is the determining factor for the release of TGF-β. Moreover, whether different resorptive conditions, such as immobilization and hyperparathyroidism, also involve TGF-β release in the pathogenesis of muscle wasting needs to be investigated.
Collapse
|
11
|
Yin L, Li N, Jia W, Wang N, Liang M, Yang X, Du G. Skeletal muscle atrophy: From mechanisms to treatments. Pharmacol Res 2021; 172:105807. [PMID: 34389456 DOI: 10.1016/j.phrs.2021.105807] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/03/2021] [Accepted: 08/07/2021] [Indexed: 02/07/2023]
Abstract
Skeletal muscle is a crucial tissue for movement, gestural assistance, metabolic homeostasis, and thermogenesis. It makes up approximately 40% of the total body weight and 50% of total protein. However, several pathological abnormalities (e.g., chronic diseases, cancer, long-term infection, aging) can induce an imbalance in skeletal muscle protein synthesis and degradation, which triggers muscle wasting and even leads to atrophy. Skeletal muscle atrophy is characterized by weakening, shrinking, and decreasing muscle mass and fiber cross-sectional area at the histological level. It manifests as a reduction in force production, easy fatigue and decreased exercise capability, along with a lower quality of life. Mechanistically, there are several pathophysiological processes involved in skeletal muscle atrophy, including oxidative stress and inflammation, which then activate signal transduction, such as the ubiquitin proteasome system, autophagy lysosome system, and mTOR. Considering the great economic and social burden that muscle atrophy can inflict, effective prevention and treatment strategies are essential but still limited. Exercise is widely acknowledged as the most effective therapy for skeletal muscle atrophy; unfortunately, it is not applicable for all patients. Several active substances for skeletal muscle atrophy have been discovered and evaluated in clinical trials, however, they have not been marketed to date. Knowledge is being gained on the underlying mechanisms, highlighting more promising treatment strategies in the future. In this paper, the mechanisms and treatment strategies for skeletal muscle atrophy are briefly reviewed.
Collapse
Affiliation(s)
- Lin Yin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Na Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Weihua Jia
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Nuoqi Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Meidai Liang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Xiuying Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China.
| | - Guanhua Du
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China.
| |
Collapse
|
12
|
Chen H, Qian Z, Zhang S, Tang J, Fang L, Jiang F, Ge D, Chang J, Cao J, Yang L, Cao X. Silencing COX-2 blocks PDK1/TRAF4-induced AKT activation to inhibit fibrogenesis during skeletal muscle atrophy. Redox Biol 2021; 38:101774. [PMID: 33152664 PMCID: PMC7645269 DOI: 10.1016/j.redox.2020.101774] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 02/08/2023] Open
Abstract
Skeletal muscle atrophy with high prevalence can induce weakness and fatigability and place huge burden on both health and quality of life. During skeletal muscle degeneration, excessive fibroblasts and extracellular matrix (ECM) accumulated to replace and impair the resident muscle fiber and led to loss of muscle mass. Cyclooxygenase-2 (COX-2), the rate-limiting enzyme in synthesis of prostaglandin, has been identified as a positive regulator in pathophysiological process like inflammation and oxidative stress. In our study, we found injured muscles of human subjects and mouse model overexpressed COX-2 compared to the non-damaged region and COX-2 was also upregulated in fibroblasts following TGF-β stimulation. Then we detected the effect of selective COX-2 inhibitor celecoxib on fibrogenesis. Celecoxib mediated anti-fibrotic effect by inhibiting fibroblast differentiation, proliferation and migration as well as inactivating TGF-β-dependent signaling pathway, non-canonical TGF-β pathways and suppressing generation of reactive oxygen species (ROS) and oxidative stress. In vivo pharmacological inhibition of COX-2 by celecoxib decreased tissue fibrosis and increased skeletal muscle fiber preservation reflected by less ECM formation and myofibroblast accumulation with decreased p-ERK1/2, p-Smad2/3, TGF-βR1, VEGF, NOX2 and NOX4 expression. Expression profiling further found that celecoxib could suppress PDK1 expression. The interaction between COX-2 and PDK1/AKT signaling remained unclear, here we found that COX-2 could bind to PDK1/AKT to form compound. Knockdown of COX-2 in fibroblasts by pharmacological inactivation or by siRNA restrained PDK1 expression and AKT phosphorylation induced by TGF-β treatment. Besides, si-COX-2 prevented TGF-β-induced K63-ubiquitination of AKT by blocking the interaction between AKT and E3 ubiquitin ligase TRAF4. In summary, we found blocking COX-2 inhibited fibrogenesis after muscle atrophy induced by injury and suppressed AKT signaling pathway by inhibiting upstream PDK1 expression and preventing the recruitment of TRAF4 to AKT, indicating that COX-2/PDK1/AKT signaling pathway promised to be target for treating muscle atrophy in the future.
Collapse
Affiliation(s)
- Hongtao Chen
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhanyang Qian
- Department of Orthopedics, Zhongda Hospital of Southeast University, Nanjing, Jiangsu, China
| | - Sheng Zhang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jian Tang
- Department of Plastic and Burn Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Le Fang
- Department of Critical Care Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fan Jiang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dawei Ge
- Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jie Chang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiang Cao
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Yang
- Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Xiaojian Cao
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
13
|
Young GH, Tang SC, Wu VC, Wang KC, Nong JY, Huang PY, Hu CJ, Chiou HY, Jeng JS, Hsu CY. The functional role of hemojuvelin in acute ischemic stroke. J Cereb Blood Flow Metab 2020; 40:1316-1327. [PMID: 31307288 PMCID: PMC7238368 DOI: 10.1177/0271678x19861448] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Our study aimed to establish the role of hemojuvelin (HJV) in acute ischemic stroke (AIS). We performed immunohistochemistry for HJV expression in human brain tissues from 10 AIS and 2 non-stroke autopsy subjects. Plasma HJV was measured in 112 AIS patients within 48 h after stroke. The results showed significantly increased HJV expression in brain tissues from AIS patients compare to non-stroke subjects. After adjusting for clinical variables, plasma levels of HJV within 48 h after stroke were an independent predictor of poor functional outcome three months post-stroke (OR:1.78, 95% CI: 1.03-3.07; P = 0.038). In basic part, Western blotting showed that HJV expression in mice brains was apparent at 3 h after middle cerebral artery occlusion (MCAO), and increased significantly at 72 h. In cultured cortical neurons, expression of HJV protein increased remarkably 24 h after oxygen glucose deprivation (OGD), and small interfering RNAs (siHJV) transfected OGD neurons had a lower apoptotic rate. Importantly, 72 h post-MCAO, HJV knockout mice had significantly smaller infarcts and less expression of cleaved caspase-3 protein compared with wild-type mice. In summary, HJV participates in the mechanisms of post-stroke neuronal injury, and that plasma HJV levels can be a potential early outcome indicator for AIS patients.
Collapse
Affiliation(s)
| | - Sung-Chun Tang
- Stroke Center and Department of Neurology, National Taiwan University Hospital, Taipei
| | - Vin-Cent Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei
| | - Kuo-Chuan Wang
- Department of Surgery, National Taiwan University Hospital, Taipei
| | - Jing-Yi Nong
- Department of Internal Medicine, National Taiwan University Hospital, Taipei
| | - Po-Yuan Huang
- Stroke Center and Department of Neurology, National Taiwan University Hospital, Taipei
| | - Chaur-Jong Hu
- Department of Neurology, Taipei Medical University-Shuang Ho Hospital, Taipei
| | - Hung-Yi Chiou
- School of Public Health, Taipei Medical University, Taipei
| | - Jiann-Shing Jeng
- Stroke Center and Department of Neurology, National Taiwan University Hospital, Taipei
| | - Chung Y Hsu
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung
| |
Collapse
|
14
|
Wu SH, Lu IC, Tai MH, Chai CY, Kwan AL, Huang SH. Erythropoietin Alleviates Burn-induced Muscle Wasting. Int J Med Sci 2020; 17:33-44. [PMID: 31929736 PMCID: PMC6945565 DOI: 10.7150/ijms.38590] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 11/05/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Burn injury induces long-term skeletal muscle pathology. We hypothesized EPO could attenuate burn-induced muscle fiber atrophy. Methods: Rats were allocated into four groups: a sham burn group, an untreated burn group subjected to third degree hind paw burn, and two burn groups treated with weekly or daily EPO for four weeks. Gastrocnemius muscle was analyzed at four weeks post-burn. Results: EPO attenuated the reduction of mean myofiber cross-sectional area post-burn and the level of the protective effect was no significant difference between two EPO-treated groups (p=0.784). Furthermore, EPO decreased the expression of atrophy-related ubiquitin ligase, atrogin-1, which was up-regulated in response to burn. Compared to untreated burn rats, those receiving weekly or daily EPO groups had less cell apoptosis by TUNEL assay. EPO decreased the expression of cleaved caspase 3 (key factor in the caspase-dependent pathway) and apoptosis-inducing factor (implicated in the caspase-independent pathway) after burn. Furthermore, EPO alleviated connective tissue overproduction following burn via transforming growth factor beta 1-Smad2/3 pathway. Daily EPO group caused significant erythrocytosis compared with untreated burn group but not weekly EPO group. Conclusion: EPO therapy attenuated skeletal muscle apoptosis and fibrosis at four weeks post-burn. Weekly EPO may be a safe and effective option in muscle wasting post-burn.
Collapse
Affiliation(s)
- Sheng-Hua Wu
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Anesthesiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Anesthesiology, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Anesthesiology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
| | - I-Cheng Lu
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Anesthesiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Anesthesiology, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Hong Tai
- Center for Neuroscience, National Sun Yat-Sen University, Kaohsiung, Taiwan.,Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Chee-Yin Chai
- Departments of Pathology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Aij-Lie Kwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shu-Hung Huang
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Plastic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
15
|
Anker MS, Anker SD, Coats AJ, von Haehling S. The Journal of Cachexia, Sarcopenia and Muscle stays the front-runner in geriatrics and gerontology. J Cachexia Sarcopenia Muscle 2019; 10:1151-1164. [PMID: 31821753 PMCID: PMC6903443 DOI: 10.1002/jcsm.12518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Markus S. Anker
- Division of Cardiology and Metabolism, Department of CardiologyCharité Universitätsmedizin BerlinBerlinGermany
- Berlin Institute of Health Center for Regenerative Therapies (BCRT)BerlinGermany
- German Centre for Cardiovascular Research (DZHK) partner site BerlinBerlinGermany
- Department of CardiologyCharité Campus Benjamin FranklinBerlinGermany
| | - Stefan D. Anker
- Division of Cardiology and Metabolism, Department of CardiologyCharité Universitätsmedizin BerlinBerlinGermany
- Berlin Institute of Health Center for Regenerative Therapies (BCRT)BerlinGermany
- German Centre for Cardiovascular Research (DZHK) partner site BerlinBerlinGermany
- Department of Cardiology (CVK)Charité Universitätsmedizin BerlinBerlinGermany
- Charité Universitätsmedizin BerlinBerlinGermany
| | | | - Stephan von Haehling
- Department of Cardiology and Pneumology, Heart Center GöttingenUniversity of Göttingen Medical Center, Georg‐August‐UniversityGöttingenGermany
- German Center for Cardiovascular Medicine (DZHK), partner site GöttingenGöttingenGermany
| |
Collapse
|
16
|
Biferali B, Proietti D, Mozzetta C, Madaro L. Fibro-Adipogenic Progenitors Cross-Talk in Skeletal Muscle: The Social Network. Front Physiol 2019; 10:1074. [PMID: 31496956 PMCID: PMC6713247 DOI: 10.3389/fphys.2019.01074] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 08/05/2019] [Indexed: 01/09/2023] Open
Abstract
Skeletal muscle is composed of a large and heterogeneous assortment of cell populations that interact with each other to maintain muscle homeostasis and orchestrate regeneration. Although satellite cells (SCs) – which are muscle stem cells – are the protagonists of functional muscle repair following damage, several other cells such as inflammatory, vascular, and mesenchymal cells coordinate muscle regeneration in a finely tuned process. Fibro–adipogenic progenitors (FAPs) are a muscle interstitial mesenchymal cell population, which supports SCs differentiation during tissue regeneration. During the first days following muscle injury FAPs undergo massive expansion, which is followed by their macrophage-mediated clearance and the re-establishment of their steady-state pool. It is during this critical time window that FAPs, together with the other cellular components of the muscle stem cell niche, establish a dynamic network of interactions that culminate in muscle repair. A number of different molecules have been recently identified as important mediators of this cross-talk, and its alteration has been associated with different muscle pathologies. In this review, we will focus on the soluble factors that regulate FAPs activity, highlighting their roles in orchestrating the inter-cellular interactions between FAPs and the other cell populations that participate in muscle regeneration.
Collapse
Affiliation(s)
- Beatrice Biferali
- Department of Biology and Biotechnology "C. Darwin," Sapienza University of Rome, Rome, Italy.,Institute of Molecular Biology and Pathology (IBPM), CNR National Research Council of Italy, c/o Department of Biology and Biotechnology "C. Darwin," Sapienza University of Rome, Rome, Italy
| | - Daisy Proietti
- IRCCS Santa Lucia Foundation, Rome, Italy.,DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Chiara Mozzetta
- Institute of Molecular Biology and Pathology (IBPM), CNR National Research Council of Italy, c/o Department of Biology and Biotechnology "C. Darwin," Sapienza University of Rome, Rome, Italy
| | | |
Collapse
|
17
|
Zhang P, He J, Wang F, Gong J, Wang L, Wu Q, Li W, Liu H, Wang J, Zhang K, Li M, Huang X, Pu C, Li Y, Jiang F, Wang F, Min J, Chen X. Hemojuvelin is a novel suppressor for Duchenne muscular dystrophy and age-related muscle wasting. J Cachexia Sarcopenia Muscle 2019; 10:557-573. [PMID: 30884219 PMCID: PMC6596404 DOI: 10.1002/jcsm.12414] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 01/27/2019] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Muscle wasting occurs in response to various physiological and pathological conditions, including ageing and Duchenne muscular dystrophy (DMD). Transforming growth factor-β1 (TGF-β1) contributes to muscle pathogenesis in elderly people and DMD patients; inhibition of TGF-β1 signalling is a promising therapeutic strategy for muscle-wasting disorders. Hemojuvelin (HJV or Hjv as the murine homologue) is a membrane-bound protein that is highly expressed in skeletal muscle, heart, and liver. In hepatic cells, Hjv acts as a coreceptor for bone morphogenetic protein, a TGF-β subfamily member. The aim of this study was to investigate whether Hjv plays an essential role in muscle physiological and pathophysiological processes by acting as a coreceptor for TGF-β1 signalling. METHODS Conventional and conditional Hjv knockout mice as well as mdx and aged mice transfected with Hjv overexpression vector were used to study the role of Hjv in muscle physiology and pathophysiology. qRT-PCR, western blotting, and immunohistochemistry examinations were conducted to evaluate gene, protein, and structural changes in vivo and in vitro. Exercise endurance was determined using treadmill running test, and muscle force was detected by an isometric transducer. RNA interference, immunoprecipitation, and dual-luciferase reporter assays were utilized to explore the mechanism by which Hjv regulates TGF-β1 signalling in skeletal muscle. RESULTS Conventional and conditional Hjv knockout mice displayed muscle atrophy, fibrosis, reduced running endurance, and muscle force. HJV was significantly down-regulated in the muscles of DMD patients (n = 3, mean age: 11.7 ± 5.7 years) and mdx mice as well as in those of aged humans (n = 10, 20% women, mean age: 75.1 ± 9.5 years) and mice. Overexpression of Hjv rescued dystrophic and age-related muscle wasting. Unlike its function in hepatic cells, the bone morphogenetic protein downstream phosphorylated p-Smad1/5/8 signalling pathway was unchanged, but TGF-β1, TGF-β receptor II (TβRII), and p-Smad2/3 expression were increased in Hjv-deficient muscles. Mechanistically, loss of Hjv promoted activation of Smad3 signalling induced by TGF-β1, whereas Hjv overexpression inhibited TGF-β1/Smad3 signalling by directly interacting with TβRII on the muscle membrane. CONCLUSIONS Our findings identify an unrecognized role of HJV in skeletal muscle by regulating TGF-β1/Smad3 signalling as a coreceptor for TβRII. Unlike the TGF-β1/Smad3 pathway, HJV could be a reliable drug target as its expression is not widespread. Novel therapeutic strategies could potentially be devised to interfere only with the muscle function of HJV to treat DMD and age-related muscle wasting.
Collapse
Affiliation(s)
- Peng Zhang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Jian He
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Fei Wang
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| | - Jing Gong
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Lu Wang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Qian Wu
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenjiong Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Hongju Liu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Jing Wang
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| | - Kunshan Zhang
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Mao Li
- Department of Neurology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xusheng Huang
- Department of Neurology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Chuanqiang Pu
- Department of Neurology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Ying Li
- No. 454 Hospital of People's Liberation Army, Nanjing, China
| | - Fengjie Jiang
- No. 454 Hospital of People's Liberation Army, Nanjing, China
| | - Fudi Wang
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoping Chen
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China.,National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| |
Collapse
|