1
|
Yao X, Mai X, Tian Y, Liu Y, Jin G, Li Z, Chen S, Dai X, Huang L, Fan Z, Pan G, Pan X, Li X, Yu MC, Sun J, Ou J, Chen H, Xie L. Skeletal muscle-specific Bambi deletion induces hypertrophy and oxidative switching coupling with adipocyte thermogenesis against metabolic disorders. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1352-1368. [PMID: 39821828 DOI: 10.1007/s11427-023-2586-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 04/01/2024] [Indexed: 01/19/2025]
Abstract
Skeletal muscle plays a significant role in both local and systemic energy metabolism. The current investigation aims to explore the role of the Bambi gene in skeletal muscle, focusing on its implications for muscle hypertrophy and systemic metabolism. We hypothesize that skeletal muscle-specific deletion of Bambi induces muscle hypertrophy, improves metabolic performance, and activates thermogenic adipocytes via the reprogramming of progenitor of iWAT, offering potential therapeutic strategies for metabolic syndromes. Leveraging the Chromatin immunoprecipitation (ChIP)-seq and bioinformatics analysis, Bambi gene is shown to be a direct target of HIF2α, which is further confirmed by ChIP-qPCR and promoter luciferase assay. Skeletal muscle-specific Bambi deletion led to significant muscle hypertrophy and improved metabolic parameters, even under high-fat diet conditions. This deletion induced metabolic reprogramming of stromal vascular fractions (SVFs) into thermogenic adipocytes, contributing to systemic metabolic improvements, potentially through the secretory factor. Notably, mice with skeletal muscle-specific Bambi deletion demonstrate resistance to high-fat diet-induced metabolic disorders, highlighting a potential therapeutic pathway for metabolic syndrome management. Thus, skeletal muscle-specific deletion of Bambi triggers muscle growth, enhances metabolic performance, and activates thermogenic adipocytes. These findings suggest Bambi as a novel therapeutic target for metabolic syndromes, providing new insights into the interaction between muscle hypertrophy and systemic metabolic improvement. The study underscores the potential of manipulating muscle physiology to regulate whole-body metabolism, offering a novel perspective on treating metabolic disorders.
Collapse
Affiliation(s)
- Xiangping Yao
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, 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, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Xudong Mai
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, 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, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Ye Tian
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, 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, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Yifan Liu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, 524023, China
| | - Guanghui Jin
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University; Organ Transplantation Research Center of Guangdong Province, Guangdong province engineering laboratory for transplantation medicine; Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China
| | - Ze Li
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China
| | - Shujie Chen
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, 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, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Xiaoshuang Dai
- BGI Institute of Applied Agriculture, BGI-Shenzhen, Shenzhen, 518120, China
| | - Liujing Huang
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, 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, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Zijing Fan
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China
| | - Guihua 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, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, 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, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, 999077, China
| | - Xiangmin Li
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Michael C Yu
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, 14260, USA
| | - Jia Sun
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jingxing Ou
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University; Organ Transplantation Research Center of Guangdong Province, Guangdong province engineering laboratory for transplantation medicine; Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China.
| | - Hong Chen
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Liwei Xie
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, 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, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China.
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China.
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, 524023, China.
- Department of Internal Medicine, Shunde Women and Children's Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, 528300, China.
- College of Life and Health Sciences, Guangdong Industry Polytechnic, Guangzhou, 510300, China.
| |
Collapse
|
2
|
Zhu P, Pfrender EM, Steffeck AWT, Reczek CR, Zhou Y, Thakkar AV, Gupta NR, Kupai A, Willbanks A, Lieber RL, Roy I, Chandel NS, Peek CB. Immunomodulatory role of the stem cell circadian clock in muscle repair. SCIENCE ADVANCES 2025; 11:eadq8538. [PMID: 40043110 PMCID: PMC11881903 DOI: 10.1126/sciadv.adq8538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 01/30/2025] [Indexed: 03/09/2025]
Abstract
Circadian rhythms orchestrate physiological processes such as metabolism, immune function, and tissue regeneration, aligning them with the optimal time of day (TOD). This study identifies an interplay between the circadian clock within muscle stem cells (SCs) and their capacity to modulate the immune microenvironment during muscle regeneration. We reveal that the SC clock triggers TOD-dependent inflammatory gene transcription after injury, particularly genes related to neutrophil activity and chemotaxis. These responses are driven by cytosolic regeneration of the signaling metabolite nicotinamide adenine dinucleotide (oxidized form) (NAD+), as enhancing cytosolic NAD+ regeneration in SCs is sufficient to induce inflammatory responses that influence muscle regeneration. Mononuclear single-cell sequencing of the regenerating muscle niche further implicates the cytokine CCL2 in mediating SC-neutrophil cross-talk in a TOD-dependent manner. Our findings highlight the intersection between SC metabolic shifts and immune responses within the muscle microenvironment, dictated by circadian rhythms, and underscore the potential for targeting circadian and metabolic pathways to enhance tissue regeneration.
Collapse
Affiliation(s)
- Pei Zhu
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Eric M. Pfrender
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Adam W. T. Steffeck
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Colleen R. Reczek
- Department of Medicine, Division of Pulmonary and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yalu Zhou
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Feinberg Cardiovascular and Renal Research Institute, Chicago, IL, USA
| | - Abhishek Vijay Thakkar
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Neha R. Gupta
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ariana Kupai
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Amber Willbanks
- Shirley Ryan AbilityLab (formerly known as Rehabilitation Institute of Chicago), Chicago, IL, USA
| | - Richard L. Lieber
- Shirley Ryan AbilityLab (formerly known as Rehabilitation Institute of Chicago), Chicago, IL, USA
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL, USA
- Hines VA Hospital, Maywood, IL, USA
| | - Ishan Roy
- Shirley Ryan AbilityLab (formerly known as Rehabilitation Institute of Chicago), Chicago, IL, USA
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Navdeep S. Chandel
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Pulmonary and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Clara B. Peek
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
3
|
Maimaiti Y, Abulitifu M, Ajimu Z, Su T, Zhang Z, Yu Z, Xu H. FOXO regulation of TXNIP induces ferroptosis in satellite cells by inhibiting glutathione metabolism, promoting Sarcopenia. Cell Mol Life Sci 2025; 82:81. [PMID: 39982519 PMCID: PMC11845654 DOI: 10.1007/s00018-025-05592-1] [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: 10/29/2024] [Revised: 12/30/2024] [Accepted: 01/12/2025] [Indexed: 02/22/2025]
Abstract
Aging-related sarcopenia represents a significant health concern due to its impact on the quality of life in the elderly. This study elucidates the molecular mechanisms underlying sarcopenia by employing single-cell sequencing and public transcriptome databases to compare young and aged mouse skeletal muscles. Cellular classification and pseudotime analyses differentiated cell types and their interrelationships, revealing a marked reduction in satellite cell numbers and a consistent upregulation of TXNIP (Thioredoxin interacting protein) across various muscle cell populations in aged mice. Further transcriptomic data integration and batch correction from the GEO (Gene Expression Omnibus) database highlighted key differentially expressed genes. The role of TXNIP and its transcriptional regulation by FOXO1 (Forkhead box O1) was confirmed through in vitro experiments, which demonstrated FOXO1's influence on TXNIP expression and its subsequent suppression of glutathione metabolism, leading to satellite cell ferroptosis. Additionally, in vivo studies showed that overexpression of TXNIP in young mice's muscle tissues significantly reduced muscle mass, suggesting its potential role in the initiation of sarcopenia. Our findings suggest that FOXO1-mediated regulation of TXNIP and the disruption of glutathione metabolism are central to the process of sarcopenia, offering new insights into its pathogenesis.
Collapse
Affiliation(s)
- Yasenjiang Maimaiti
- Gerontology Center, People's Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Urumqi, Xinjiang, China
| | - Mukedasi Abulitifu
- Gerontology Center, People's Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Urumqi, Xinjiang, China
| | - Zulifeiya Ajimu
- Gerontology Center, People's Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Urumqi, Xinjiang, China
| | - Ting Su
- Gerontology Center, People's Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Urumqi, Xinjiang, China
| | - Zhanying Zhang
- Gerontology Center, People's Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Urumqi, Xinjiang, China
| | - Zhichao Yu
- Gerontology Center, People's Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Urumqi, Xinjiang, China
| | - Hong Xu
- Gerontology Center, People's Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Urumqi, Xinjiang, China.
| |
Collapse
|
4
|
Sartorelli V, Ciuffoli V. Metabolic regulation in adult and aging skeletal muscle stem cells. Genes Dev 2025; 39:186-208. [PMID: 39662967 PMCID: PMC11789647 DOI: 10.1101/gad.352277.124] [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] [Indexed: 12/13/2024]
Abstract
Adult stem cells maintain homeostasis and enable regeneration of most tissues. Quiescence, proliferation, and differentiation of stem cells and their progenitors are tightly regulated processes governed by dynamic transcriptional, epigenetic, and metabolic programs. Previously thought to merely reflect a cell's energy state, metabolism is now recognized for its critical regulatory functions, controlling not only energy and biomass production but also the cell's transcriptome and epigenome. In this review, we explore how metabolic pathways, metabolites, and transcriptional and epigenetic regulators are functionally interlinked in adult and aging skeletal muscle stem cells.
Collapse
Affiliation(s)
- Vittorio Sartorelli
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Veronica Ciuffoli
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
5
|
Yousefi F, Foster LA, Selim OA, Zhao C. Integrating Physical and Biochemical Cues for Muscle Engineering: Scaffolds and Graft Durability. Bioengineering (Basel) 2024; 11:1245. [PMID: 39768063 PMCID: PMC11673930 DOI: 10.3390/bioengineering11121245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
Muscle stem cells (MuSCs) are essential for skeletal muscle regeneration, influenced by a complex interplay of mechanical, biochemical, and molecular cues. Properties of the extracellular matrix (ECM) such as stiffness and alignment guide stem cell fate through mechanosensitive pathways, where forces like shear stress translate into biochemical signals, affecting cell behavior. Aging introduces senescence which disrupts the MuSC niche, leading to reduced regenerative capacity via epigenetic alterations and metabolic shifts. Transplantation further challenges MuSC viability, often resulting in fibrosis driven by dysregulated fibro-adipogenic progenitors (FAPs). Addressing these issues, scaffold designs integrated with pharmacotherapy emulate ECM environments, providing cues that enhance graft functionality and endurance. These scaffolds facilitate the synergy between mechanotransduction and intracellular signaling, optimizing MuSC proliferation and differentiation. Innovations utilizing human pluripotent stem cell-derived myogenic progenitors and exosome-mediated delivery exploit bioactive properties for targeted repair. Additionally, 3D-printed and electrospun scaffolds with adjustable biomechanical traits tackle scalability in treating volumetric muscle loss. Advanced techniques like single-cell RNA sequencing and high-resolution imaging unravel muscle repair mechanisms, offering precise mapping of cellular interactions. Collectively, this interdisciplinary approach fortifies tissue graft durability and MuSC maintenance, propelling therapeutic strategies for muscle injuries and degenerative diseases.
Collapse
Affiliation(s)
- Farbod Yousefi
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| | - Lauren Ann Foster
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
- Atlanta Veterans Affairs Medical Center, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Omar A. Selim
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| | - Chunfeng Zhao
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| |
Collapse
|
6
|
Guo R, Wu Z, Liu A, Li Q, Han T, Shen C. Hypoxic preconditioning-engineered bone marrow mesenchymal stem cell-derived exosomes promote muscle satellite cell activation and skeletal muscle regeneration via the miR-210-3p/KLF7 mechanism. Int Immunopharmacol 2024; 142:113143. [PMID: 39306891 DOI: 10.1016/j.intimp.2024.113143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 08/12/2024] [Accepted: 09/08/2024] [Indexed: 10/12/2024]
Abstract
Sarcopenia is a gradual and widespread decline in muscle mass and function in skeletal muscle, leading to significant implications for individuals and society. Currently, there is a lack of effective treatment methods for sarcopenia. Muscle satellite cells(SCs) play a crucial role in the occurrence and development of sarcopenia, and their proliferation and differentiation abilities are closely related to the progression of disease. This study evaluated the effects of exosomes derived from hypoxic preconditioning bone marrow mesenchymal stem cells (BMSCs) on the proliferation of SCs and skeletal muscle regeneration. We found that the capacity for the proliferation and differentiation of SCs in elderly rats was notably diminished, leading us to create a sarcopenia model in elderly rats. By separating and extracting exosomes from BMSCs treated with normoxic (N-Exos) and hypoxic (H-Exos) conditions, in vivo and in vitro studies showed that both N-Exos and H-Exos can regulate the proliferation and differentiation of SCs in elderly rats, and promote skeletal muscle regeneration and functional recovery. The beneficial effects of H-Exos were also more significant than those of the N-Exos group. In vitro studies demonstrated that H-Exos could influence the expression of the KLF7 gene and protein in SCs by delivering miR-210-3P. This, in turn, impacted the phosphorylation of the PI3K/AKT signaling pathway and contributed to the function of SCs. H-Exos stimulated SCs and promoted skeletal muscle regeneration during sarcopenia by delivering miR-210-3P to target the KLF7/PI3K/AKT signaling pathway. This may serve as a possible treatment option for sarcopenia.
Collapse
Affiliation(s)
- Ruocheng Guo
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, Hefei 230032, P. R. China; Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, P. R. China
| | - Zuomeng Wu
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, Hefei 230032, P. R. China; Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, P. R. China
| | - Ao Liu
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, Hefei 230032, P. R. China; Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, P. R. China
| | - Qiuwei Li
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, Hefei 230032, P. R. China; Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, P. R. China
| | - Tianyu Han
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, Hefei 230032, P. R. China; Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, P. R. China
| | - Cailiang Shen
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, Hefei 230032, P. R. China; Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, P. R. China.
| |
Collapse
|
7
|
Stevens NE, Loreti M, Ramirez-Sanchez I, Dos Reis FCG, Sacco A, Breen EC, Nogueira L. Cigarette smoke exposure impairs early-stage recovery from lengthening contraction-induced muscle injury in male mice. Physiol Rep 2024; 12:e70064. [PMID: 39328164 PMCID: PMC11427903 DOI: 10.14814/phy2.70064] [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: 05/04/2024] [Revised: 09/05/2024] [Accepted: 09/15/2024] [Indexed: 09/28/2024] Open
Abstract
The use of tobacco cigarettes produces locomotor muscle weakness and fatigue intolerance. Also, smokers and chronic obstructive pulmonary disease patients have a greater incidence of muscle injury and a deficient myogenic response. However, the effects of smoke exposure on the recovery from eccentric exercise-induced muscle injuries are unknown. Mice were exposed daily to cigarette smoke (CS) or room air (Air) for 4 months; the anterior crural muscles from one limb were injured by a lengthening contractions protocol (LCP) and recovered for 7 days. Lung compliance was greater, and body weights were lower, in CS-exposed than in the Air group. In LCP-subjected limbs, CS exposure lowered tibialis anterior myofiber cross-sectional area, decreased the size of centrally nucleated myofibers, and decreased extensor digitorum longus (EDL) mass, but did not affect EDL force from both limbs. CS exposure upregulated the mRNA levels of several myogenic (Pax7, Myf5, nNOS) genes in the EDL. The combination of CS exposure and LCP decreased Myf5 and nNOS mRNA levels and exacerbated pro-inflammatory mRNA levels. These data suggest that smoke exposure leads to an excessive pro-inflammatory response in regenerating muscle that is associated with a lower muscle mass recovery from a type of injury that often occurs during strenuous exercise.
Collapse
Affiliation(s)
- Nicole E Stevens
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Mafalda Loreti
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Israel Ramirez-Sanchez
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, La Jolla, California, USA
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, IPN, Mexico City, Mexico
| | - Felipe C G Dos Reis
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Alessandra Sacco
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Ellen C Breen
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Leonardo Nogueira
- School of Exercise and Nutritional Sciences, College of Health and Human Services, San Diego State University, San Diego, California, USA
| |
Collapse
|
8
|
Duan H, Chen S, Mai X, Fu L, Huang L, Xiao L, Liao M, Chen H, Liu G, Xie L. Low-intensity pulsed ultrasound (LIPUS) promotes skeletal muscle regeneration by regulating PGC-1α/AMPK/GLUT4 pathways in satellite cells/myoblasts. Cell Signal 2024; 117:111097. [PMID: 38355078 DOI: 10.1016/j.cellsig.2024.111097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/19/2024] [Accepted: 02/11/2024] [Indexed: 02/16/2024]
Abstract
Low-Intensity Pulsed Ultrasound (LIPUS) holds therapeutic potential in promoting skeletal muscle regeneration, a biological process mediated by satellite cells and myoblasts. Despite their central roles in regeneration, the detailed mechanistic of LIPUS influence on satellite cells and myoblasts are not fully underexplored. In the current investigation, we administrated LIPUS treatment to injured skeletal muscles and C2C12 myoblasts over five consecutive days. Muscle samples were collected on days 6 and 30 post-injury for an in-depth histological and molecular assessment, both in vivo and in vitro with immunofluorescence analysis. During the acute injury phase, LIPUS treatment significantly augmented the satellite cell population, concurrently enhancing the number and size of newly formed myofibers whilst reducing fibrosis levels. At 30 days post-injury, the LIPUS-treated group demonstrated a more robust satellite cell pool and a higher myofiber count, suggesting that early LIPUS intervention facilitates satellite cell proliferation and differentiation, thereby promoting long-term recovery. Additionally, LIPUS markedly accelerated C2C12 myoblast differentiation, with observed increases in AMPK phosphorylation in myoblasts, leading to elevated expression of Glut4 and PGC-1α, and subsequent glucose uptake and mitochondrial biogenesis. These findings imply that LIPUS-induced modulation of myoblasts may culminate in enhanced cellular energy availability, laying a theoretical groundwork for employing LIPUS in ameliorating skeletal muscle regeneration post-injury. NEW & NOTEWORTHY: Utilizing the cardiotoxin (CTX) muscle injury model, we investigated the influence of LIPUS on satellite cell homeostasis and skeletal muscle regeneration. Our findings indicate that LIPUS promotes satellite cell proliferation and differentiation, thereby facilitating skeletal muscle repair. Additionally, in vitro investigations lend credence to the hypothesis that the regulatory effect of LIPUS on satellite cells may be attributed to its capability to enhance cellular energy metabolism.
Collapse
Affiliation(s)
- Huimin Duan
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510000, 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, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; Department of Anesthesiology, The Seventh Affiliated Hospital, Southern Medical University, Foshan 528244, Guangdong, China
| | - Xudong Mai
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Liping Fu
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Liujing Huang
- Medical Affairs Department, Guangzhou Betrue Technology Co., Ltd, Guangzhou 510700, China
| | - Lanling Xiao
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China
| | - Miaomiao Liao
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Hong Chen
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Gang Liu
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510000, 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, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; Department of Anesthesiology, The Seventh Affiliated Hospital, Southern Medical University, Foshan 528244, Guangdong, China; Department of Internal Medicine, Shunde Women and Children's Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China; Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; College of Life and Health Sciences, Guangdong Industry Polytechnic, Guangzhou, Guangdong 510300, China.
| |
Collapse
|
9
|
Zhu P, Peek CB. Circadian timing of satellite cell function and muscle regeneration. Curr Top Dev Biol 2024; 158:307-339. [PMID: 38670711 DOI: 10.1016/bs.ctdb.2024.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Recent research has highlighted an important role for the molecular circadian machinery in the regulation of tissue-specific function and stress responses. Indeed, disruption of circadian function, which is pervasive in modern society, is linked to accelerated aging, obesity, and type 2 diabetes. Furthermore, evidence supporting the importance of the circadian clock within both the mature muscle tissue and satellite cells to regulate the maintenance of muscle mass and repair capacity in response injury has recently emerged. Here, we review the discovery of circadian clocks within the satellite cell (a.k.a. adult muscle stem cell) and how they act to regulate metabolism, epigenetics, and myogenesis during both healthy and diseased states.
Collapse
Affiliation(s)
- Pei Zhu
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States; Department of Medicine-Endocrinology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.
| | - Clara B Peek
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States; Department of Medicine-Endocrinology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.
| |
Collapse
|
10
|
Chen S, Huang L, Liu B, Duan H, Li Z, Liu Y, Li H, Fu X, Lin J, Xu Y, Liu L, Wan D, Yin Y, Xie L. Dynamic changes in butyrate levels regulate satellite cell homeostasis by preventing spontaneous activation during aging. SCIENCE CHINA. LIFE SCIENCES 2024; 67:745-764. [PMID: 38157106 DOI: 10.1007/s11427-023-2400-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 06/15/2023] [Indexed: 01/03/2024]
Abstract
The gut microbiota plays a pivotal role in systemic metabolic processes and in particular functions, such as developing and preserving the skeletal muscle system. However, the interplay between gut microbiota/metabolites and the regulation of satellite cell (SC) homeostasis, particularly during aging, remains elusive. We propose that gut microbiota and its metabolites modulate SC physiology and homeostasis throughout skeletal muscle development, regeneration, and aging process. Our investigation reveals that microbial dysbiosis manipulated by either antibiotic treatment or fecal microbiota transplantation from aged to adult mice, leads to the activation of SCs or a significant reduction in the total number. Furthermore, employing multi-omics (e.g., RNA-seq, 16S rRNA gene sequencing, and metabolomics) and bioinformatic analysis, we demonstrate that the reduced butyrate levels, alongside the gut microbial dysbiosis, could be the primary factor contributing to the reduction in the number of SCs and subsequent impairments during skeletal muscle aging. Meanwhile, butyrate supplementation can mitigate the antibiotics-induced SC activation irrespective of gut microbiota, potentially by inhibiting the proliferation and differentiation of SCs/myoblasts. The butyrate effect is likely facilitated through the monocarboxylate transporter 1 (Mct1), a lactate transporter enriched on membranes of SCs and myoblasts. As a result, butyrate could serve as an alternative strategy to enhance SC homeostasis and function during skeletal muscle aging. Our findings shed light on the potential application of microbial metabolites in maintaining SC homeostasis and preventing skeletal muscle aging.
Collapse
Affiliation(s)
- Shujie Chen
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, 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, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Liujing Huang
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, 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, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Bingdong Liu
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, 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, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Huimin Duan
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Ze Li
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yifan Liu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, 524023, China
| | - Hu Li
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, China
| | - Xiang Fu
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, China
| | - Jingchao Lin
- Metabo-Profile Biotechnology (Shanghai) Co. Ltd., Shanghai, 201315, China
| | - Yinlan Xu
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China
| | - Li Liu
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Dan Wan
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, 524023, China.
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
| | - Yulong Yin
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, 524023, China.
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
| | - Liwei Xie
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, 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, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China.
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China.
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
11
|
Yin A, Fu W, Elengickal A, Kim J, Liu Y, Bigot A, Mamchaoui K, Call JA, Yin H. Chronic hypoxia impairs skeletal muscle repair via HIF-2α stabilization. J Cachexia Sarcopenia Muscle 2024; 15:631-645. [PMID: 38333911 PMCID: PMC10995261 DOI: 10.1002/jcsm.13436] [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: 08/14/2023] [Revised: 11/28/2023] [Accepted: 01/02/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Chronic hypoxia and skeletal muscle atrophy commonly coexist in patients with COPD and CHF, yet the underlying physio-pathological mechanisms remain elusive. Muscle regeneration, driven by muscle stem cells (MuSCs), holds therapeutic potential for mitigating muscle atrophy. This study endeavours to investigate the influence of chronic hypoxia on muscle regeneration, unravel key molecular mechanisms, and explore potential therapeutic interventions. METHODS Experimental mice were exposed to prolonged normobaric hypoxic air (15% pO2, 1 atm, 2 weeks) to establish a chronic hypoxia model. The impact of chronic hypoxia on body composition, muscle mass, muscle strength, and the expression levels of hypoxia-inducible factors HIF-1α and HIF-2α in MuSC was examined. The influence of chronic hypoxia on muscle regeneration, MuSC proliferation, and the recovery of muscle mass and strength following cardiotoxin-induced injury were assessed. The muscle regeneration capacities under chronic hypoxia were compared between wildtype mice, MuSC-specific HIF-2α knockout mice, and mice treated with HIF-2α inhibitor PT2385, and angiotensin converting enzyme (ACE) inhibitor lisinopril. Transcriptomic analysis was performed to identify hypoxia- and HIF-2α-dependent molecular mechanisms. Statistical significance was determined using analysis of variance (ANOVA) and Mann-Whitney U tests. RESULTS Chronic hypoxia led to limb muscle atrophy (EDL: 17.7%, P < 0.001; Soleus: 11.5% reduction in weight, P < 0.001) and weakness (10.0% reduction in peak-isometric torque, P < 0.001), along with impaired muscle regeneration characterized by diminished myofibre cross-sectional areas, increased fibrosis (P < 0.001), and incomplete strength recovery (92.3% of pre-injury levels, P < 0.05). HIF-2α stabilization in MuSC under chronic hypoxia hindered MuSC proliferation (26.1% reduction of MuSC at 10 dpi, P < 0.01). HIF-2α ablation in MuSC mitigated the adverse effects of chronic hypoxia on muscle regeneration and MuSC proliferation (30.9% increase in MuSC numbers at 10 dpi, P < 0.01), while HIF-1α ablation did not have the same effect. HIF-2α stabilization under chronic hypoxia led to elevated local ACE, a novel direct target of HIF-2α. Notably, pharmacological interventions with PT2385 or lisinopril enhanced muscle regeneration under chronic hypoxia (PT2385: 81.3% increase, P < 0.001; lisinopril: 34.6% increase in MuSC numbers at 10 dpi, P < 0.05), suggesting their therapeutic potential for alleviating chronic hypoxia-associated muscle atrophy. CONCLUSIONS Chronic hypoxia detrimentally affects skeletal muscle regeneration by stabilizing HIF-2α in MuSC and thereby diminishing MuSC proliferation. HIF-2α increases local ACE levels in skeletal muscle, contributing to hypoxia-induced regenerative deficits. Administration of HIF-2α or ACE inhibitors may prove beneficial to ameliorate chronic hypoxia-associated muscle atrophy and weakness by improving muscle regeneration under chronic hypoxia.
Collapse
Affiliation(s)
- Amelia Yin
- Center for Molecular MedicineThe University of GeorgiaAthensGAUSA
- Department of Biochemistry and Molecular BiologyThe University of GeorgiaAthensGAUSA
| | - Wenyan Fu
- Center for Molecular MedicineThe University of GeorgiaAthensGAUSA
- Department of Biochemistry and Molecular BiologyThe University of GeorgiaAthensGAUSA
| | - Anthony Elengickal
- Department of Biochemistry and Molecular BiologyThe University of GeorgiaAthensGAUSA
| | - Joonhee Kim
- Department of Biochemistry and Molecular BiologyThe University of GeorgiaAthensGAUSA
| | - Yang Liu
- Center for Molecular MedicineThe University of GeorgiaAthensGAUSA
- Department of Biochemistry and Molecular BiologyThe University of GeorgiaAthensGAUSA
| | - Anne Bigot
- Sorbonne Université, Inserm, Institut de MyologieCentre de Recherche en MyologieParisFrance
| | - Kamal Mamchaoui
- Sorbonne Université, Inserm, Institut de MyologieCentre de Recherche en MyologieParisFrance
| | - Jarrod A. Call
- Department of Physiology and PharmacologyThe University of GeorgiaAthensGAUSA
| | - Hang Yin
- Center for Molecular MedicineThe University of GeorgiaAthensGAUSA
- Department of Biochemistry and Molecular BiologyThe University of GeorgiaAthensGAUSA
| |
Collapse
|
12
|
Shi J, Lv Q, Miao D, Xiong Z, Wei Z, Wu S, Tan D, Wang K, Zhang X. HIF2α Promotes Cancer Metastasis through TCF7L2-Dependent Fatty Acid Synthesis in ccRCC. RESEARCH (WASHINGTON, D.C.) 2024; 7:0322. [PMID: 38390305 PMCID: PMC10882601 DOI: 10.34133/research.0322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 01/27/2024] [Indexed: 02/24/2024]
Abstract
Recent studies have highlighted the notable involvement of the crosstalk between hypoxia-inducible factor 2 alpha (HIF2α) and Wnt signaling components in tumorigenesis. However, the cellular function and precise regulatory mechanisms of HIF2α and Wnt signaling interactions in clear cell renal cell carcinoma (ccRCC) remain elusive. To analyze the correlation between HIF2α and Wnt signaling, we utilized the Cancer Genome Atlas - Kidney Renal Clear Cell Carcinoma (TCGA-KIRC) public database, HIF2α RNA sequencing data, and conducted luciferase reporter assays. A Wnt-related gene set was employed to identify key regulators of Wnt signaling controlled by HIF2α in ccRCC. Furthermore, we assessed the biological effects of TCF7L2 on ccRCC metastasis and lipid metabolism in both in vivo and in vitro settings. Our outcomes confirm TCF7L2 as a key gene involved in HIF2α-mediated regulation of the canonical Wnt pathway. Functional studies demonstrate that TCF7L2 promotes metastasis in ccRCC. Mechanistic investigations reveal that HIF2α stabilizes TCF7L2 mRNA in a method based on m6A by transcriptionally regulating METTL3. Up-regulation of TCF7L2 enhances cellular fatty acid oxidation, which promotes histone acetylation. This facilitates the transcription of genes connected to epithelial-mesenchymal transition and ultimately enhances metastasis of ccRCC. These outcomes offer a novel understanding into the involvement of lipid metabolism in the signaling pathway regulation, offering valuable implications for targeted treatment in ccRCC.
Collapse
Affiliation(s)
- Jian Shi
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
| | - Qingyang Lv
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
| | - Daojia Miao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
| | - Zhiyong Xiong
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
| | - Zhihao Wei
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
| | - Songming Wu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
| | - Diaoyi Tan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
| | - Keshan Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
- Institute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, P. R. China
| |
Collapse
|
13
|
Kim H, Liu Y, Kim J, Kim Y, Klouda T, Fisch S, Baek SH, Liu T, Dahlberg S, Hu CJ, Tian W, Jiang X, Kosmas K, Christou HA, Korman BD, Vargas SO, Wu JC, Stenmark KR, Perez VDJ, Nicolls MR, Raby BA, Yuan K. Pericytes contribute to pulmonary vascular remodeling via HIF2α signaling. EMBO Rep 2024; 25:616-645. [PMID: 38243138 PMCID: PMC10897382 DOI: 10.1038/s44319-023-00054-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 01/21/2024] Open
Abstract
Vascular remodeling is the process of structural alteration and cell rearrangement of blood vessels in response to injury and is the cause of many of the world's most afflicted cardiovascular conditions, including pulmonary arterial hypertension (PAH). Many studies have focused on the effects of vascular endothelial cells and smooth muscle cells (SMCs) during vascular remodeling, but pericytes, an indispensable cell population residing largely in capillaries, are ignored in this maladaptive process. Here, we report that hypoxia-inducible factor 2α (HIF2α) expression is increased in the lung tissues of PAH patients, and HIF2α overexpressed pericytes result in greater contractility and an impaired endothelial-pericyte interaction. Using single-cell RNAseq and hypoxia-induced pulmonary hypertension (PH) models, we show that HIF2α is a major molecular regulator for the transformation of pericytes into SMC-like cells. Pericyte-selective HIF2α overexpression in mice exacerbates PH and right ventricular hypertrophy. Temporal cellular lineage tracing shows that HIF2α overexpressing reporter NG2+ cells (pericyte-selective) relocate from capillaries to arterioles and co-express SMA. This novel insight into the crucial role of NG2+ pericytes in pulmonary vascular remodeling via HIF2α signaling suggests a potential drug target for PH.
Collapse
Affiliation(s)
- Hyunbum Kim
- Division of Pulmonary Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yu Liu
- Stanford Cardiovascular Institute; Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Jiwon Kim
- Division of Pulmonary Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yunhye Kim
- Division of Pulmonary Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Timothy Klouda
- Division of Pulmonary Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sudeshna Fisch
- Department of Medicine, Brigham and Women Hospital, Boston, MA, USA
| | - Seung Han Baek
- Division of Pulmonary Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tiffany Liu
- Division of Pulmonary Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Suzanne Dahlberg
- Division of Pulmonary Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Cheng-Jun Hu
- Cardiovascular Pulmonary Research Laboratories, Division of Pulmonary Sciences and Critical Care Medicine, Division of Pediatrics-Critical Care, Departments of Medicine and Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Wen Tian
- Division of Pulmonary, Allergy and Critical Care Medicine, Dept of Medicine, Stanford University, Stanford, CA, USA
| | - Xinguo Jiang
- Division of Pulmonary, Allergy and Critical Care Medicine, Dept of Medicine, Stanford University, Stanford, CA, USA
| | - Kosmas Kosmas
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Helen A Christou
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Benjamin D Korman
- Division of Allergy/Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, 14623, USA
| | - Sara O Vargas
- Division of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute; Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Kurt R Stenmark
- Cardiovascular Pulmonary Research Laboratories, Division of Pulmonary Sciences and Critical Care Medicine, Division of Pediatrics-Critical Care, Departments of Medicine and Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Vinicio de Jesus Perez
- Division of Pulmonary, Allergy and Critical Care Medicine, Dept of Medicine, Stanford University, Stanford, CA, USA
| | - Mark R Nicolls
- Division of Pulmonary, Allergy and Critical Care Medicine, Dept of Medicine, Stanford University, Stanford, CA, USA
| | - Benjamin A Raby
- Division of Pulmonary Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ke Yuan
- Division of Pulmonary Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Endo Y, Zhu C, Giunta E, Guo C, Koh DJ, Sinha I. The Role of Hypoxia and Hypoxia Signaling in Skeletal Muscle Physiology. Adv Biol (Weinh) 2024; 8:e2200300. [PMID: 37817370 DOI: 10.1002/adbi.202200300] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 04/06/2023] [Indexed: 10/12/2023]
Abstract
Hypoxia and hypoxia signaling play an integral role in regulating skeletal muscle physiology. Environmental hypoxia and tissue hypoxia in muscles cue for their appropriate physiological response and adaptation, and cause an array of cellular and metabolic changes. In addition, muscle stem cells (satellite cells), exist in a hypoxic state, and this intrinsic hypoxic state correlates with their quiescence and stemness. The mechanisms of hypoxia-mediated regulation of satellite cells and myogenesis are yet to be characterized, and their seemingly contradicting effects reported leave their exact roles somewhat perplexing. This review summarizes the recent findings on the effect of hypoxia and hypoxia signaling on the key aspects of muscle physiology, namely, stem cell maintenance and myogenesis with a particular attention given to distinguish the intrinsic versus local hypoxia in an attempt to better understand their respective regulatory roles and how their relationship affects the overall response. This review further describes their mechanistic links and their possible implications on the relevant pathologies and therapeutics.
Collapse
Affiliation(s)
- Yori Endo
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard University, Boston, MA, 02115, USA
| | - Christina Zhu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard University, Boston, MA, 02115, USA
- Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX, 79430, USA
| | - Elena Giunta
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard University, Boston, MA, 02115, USA
- Ludwig-Maximilians-Universität München, Geschwister-Scholl-Platz 1, 80539, München, Germany
| | - Cynthia Guo
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard University, Boston, MA, 02115, USA
- Warren Alpert Medical School, Brown University, Providence, RI, 02903, USA
| | - Daniel J Koh
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard University, Boston, MA, 02115, USA
| | - Indranil Sinha
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard University, Boston, MA, 02115, USA
| |
Collapse
|
15
|
Endo Y. Editorial: Application of innovative techniques in genetic and cellular therapies. Front Bioeng Biotechnol 2023; 11:1336900. [PMID: 38164404 PMCID: PMC10757950 DOI: 10.3389/fbioe.2023.1336900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 12/08/2023] [Indexed: 01/03/2024] Open
Affiliation(s)
- Yori Endo
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
16
|
Liu S, Liu Z, Wang P, Li W, Zhao S, Liu Y, Chu M. Estrogen-mediated oar-miR-485-5p targets PPP1R13B to regulate myoblast proliferation in sheep. Int J Biol Macromol 2023; 236:123987. [PMID: 36906210 DOI: 10.1016/j.ijbiomac.2023.123987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/03/2023] [Accepted: 02/19/2023] [Indexed: 03/11/2023]
Abstract
Ovaries are important endocrine organs in female animals that secrete various steroid hormones, which are involved in multiple physiological functions. Estrogen, one of the hormones secreted by ovaries, is essential for the overall maintenance of muscle growth and development. However, the molecular mechanisms that affect muscle growth and development in sheep following ovariectomy remain unclear. In this study, we identified 1662 differentially expressed mRNAs (DEGs) and 40 differentially expressed miRNAs (DEMs) in sheep that underwent ovariectomy compared with those that underwent sham surgery. A total of 178 DEG-DEM pairs were negatively correlated. GO and KEGG analysis showed that PPP1R13B was involved in the PI3K-Akt signaling pathway, which was essential for muscle development. Using in vitro experiments, we examined the effect of PPP1R13B on myoblast proliferation and found that overexpression or inhibition of PPP1R13B increased or decreased the expression of myoblast proliferation markers, respectively. PPP1R13B was identified as a functional downstream target of miR-485-5p. Our results suggested that miR-485-5p promoted myoblast proliferation by regulating proliferation factors in myoblasts by targeting PPP1R13B. Notably, exogenous estradiol supplementation to myoblasts regulated the expression of oar-miR-485-5p and PPP1R13B and promoted myoblast proliferation. These results provided new insights into the molecular mechanism by which ovaries influence muscle growth and development in sheep.
Collapse
Affiliation(s)
- Siqi Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Ziyi Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Peng Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Wentao Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Shengguo Zhao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Yufang Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Mingxing Chu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
17
|
Łoboda A, Dulak J. Nuclear Factor Erythroid 2-Related Factor 2 and Its Targets in Skeletal Muscle Repair and Regeneration. Antioxid Redox Signal 2023; 38:619-642. [PMID: 36597355 DOI: 10.1089/ars.2022.0208] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Significance: Skeletal muscles have a robust regenerative capacity in response to acute and chronic injuries. Muscle repair and redox homeostasis are intimately linked; increased generation of reactive oxygen species leads to cellular dysfunction and contributes to muscle wasting and progression of muscle diseases. In exemplary muscle disease, Duchenne muscular dystrophy (DMD), caused by mutations in the DMD gene that encodes the muscle structural protein dystrophin, the regeneration machinery is severely compromised, while oxidative stress contributes to the progression of the disease. The nuclear factor erythroid 2-related factor 2 (NRF2) and its target genes, including heme oxygenase-1 (HO-1), provide protective mechanisms against oxidative insults. Recent Advances: Relevant advances have been evolving in recent years in understanding the mechanisms by which NRF2 regulates processes that contribute to effective muscle regeneration. To this end, pathways related to muscle satellite cell differentiation, oxidative stress, mitochondrial metabolism, inflammation, fibrosis, and angiogenesis have been studied. The regulatory role of NRF2 in skeletal muscle ferroptosis has been also suggested. Animal studies have shown that NRF2 pathway activation can stop or reverse skeletal muscle pathology, especially when endogenous stress defence mechanisms are imbalanced. Critical Issues: Despite the growing recognition of NRF2 as a factor that regulates various aspects of muscle regeneration, the mechanistic impact on muscle pathology in various models of muscle injury remains imprecise. Future Directions: Further studies are necessary to fully uncover the role of NRF2 in muscle regeneration, both in physiological and pathological conditions, and to investigate the possibilities for development of new therapeutic modalities. Antioxid. Redox Signal. 38, 619-642.
Collapse
Affiliation(s)
- Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| |
Collapse
|
18
|
Thome T, Kim K, Dong G, Ryan TE. The Role of Mitochondrial and Redox Alterations in the Skeletal Myopathy Associated with Chronic Kidney Disease. Antioxid Redox Signal 2023; 38:318-337. [PMID: 36245209 PMCID: PMC9986033 DOI: 10.1089/ars.2022.0143] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/28/2022] [Accepted: 10/01/2022] [Indexed: 11/12/2022]
Abstract
Significance: An estimated 700 million people globally suffer from chronic kidney disease (CKD). In addition to increasing cardiovascular disease risk, CKD is a catabolic disease that results in a loss of muscle mass and function, which are strongly associated with mortality and a reduced quality of life. Despite the importance of muscle health and function, there are no treatments available to prevent or attenuate the myopathy associated with CKD. Recent Advances: Recent studies have begun to unravel the changes in mitochondrial and redox homeostasis within skeletal muscle during CKD. Impairments in mitochondrial metabolism, characterized by reduced oxidative phosphorylation, are found in both rodents and patients with CKD. Associated with aberrant mitochondrial function, clinical and preclinical findings have documented signs of oxidative stress, although the molecular source and species are ill-defined. Critical Issues: First, we review the pathobiology of CKD and its associated myopathy, and we review muscle cell bioenergetics and redox biology. Second, we discuss evidence from clinical and preclinical studies that have implicated the involvement of mitochondrial and redox alterations in CKD-associated myopathy and review the underlying mechanisms reported. Third, we discuss gaps in knowledge related to mitochondrial and redox alterations on muscle health and function in CKD. Future Directions: Despite what has been learned, effective treatments to improve muscle health in CKD remain elusive. Further studies are needed to uncover the complex mitochondrial and redox alterations, including post-transcriptional protein alterations, in patients with CKD and how these changes interact with known or unknown catabolic pathways contributing to poor muscle health and function. Antioxid. Redox Signal. 38, 318-337.
Collapse
Affiliation(s)
- Trace Thome
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Kyoungrae Kim
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Gengfu Dong
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
- Center for Exercise Science, University of Florida, Gainesville, Florida, USA
- Myology Institute, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
19
|
Identification of Key Genes and Biological Pathways Associated with Skeletal Muscle Maturation and Hypertrophy in Bos taurus, Ovis aries, and Sus scrofa. Animals (Basel) 2022; 12:ani12243471. [PMID: 36552391 PMCID: PMC9774933 DOI: 10.3390/ani12243471] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
The aim of the current study was to identify the major genes and pathways involved in the process of hypertrophy and skeletal muscle maturation that is common for Bos taurus, Ovis aries, and Sus scrofa species. Gene expression profiles related to Bos taurus, Ovis aries, and Sus scrofa muscle, with accession numbers GSE44030, GSE23563, and GSE38518, respectively, were downloaded from the GEO database. Differentially expressed genes (DEGs) were screened out using the Limma package of R software. Genes with Fold Change > 2 and an adjusted p-value < 0.05 were identified as significantly different between two treatments in each species. Subsequently, gene ontology and pathway enrichment analyses were performed. Moreover, hub genes were detected by creating a protein−protein interaction network (PPI). The results of the analysis in Bos taurus showed that in the period of 280 dpc−3-months old, a total of 1839 genes showed a significant difference. In Ovis aries, however, during the period of 135dpc−2-months old, a total of 486 genes were significantly different. Additionally, in the 91 dpc−adult period, a total of 2949 genes were significantly different in Sus scrofa. The results of the KEGG pathway enrichment analysis and GO function annotation in each species separately revealed that in Bos taurus, DEGs were mainly enriched through skeletal muscle fiber development and skeletal muscle contraction, and the positive regulation of fibroblast proliferation, positive regulation of skeletal muscle fiber development, PPAR signaling pathway, and HIF-1 signaling pathway. In Ovis aries, DEGs were mainly enriched through regulating cell growth, skeletal muscle fiber development, the positive regulation of fibroblast proliferation, skeletal muscle cell differentiation, and the PI3K-Akt signaling, HIF-1 signaling, and Rap1 signaling pathways. In Sus scrofa, DEGs were mainly enriched through regulating striated muscle tissue development, the negative regulation of fibroblast proliferation and myoblast differentiation, and the HIF-1 signaling, AMPK signaling, and PI3K-Akt signaling pathways. Using a Venn diagram, 36 common DEGs were identified between Bos taurus, Ovis aries, and Sus scrofa. A biological pathways analysis of 36 common DEGs in Bos taurus, Ovis aries, and Sus scrofa allowed for the identification of common pathways/biological processes, such as myoblast differentiation, the regulation of muscle cell differentiation, and positive regulation of skeletal muscle fiber development, that orchestrated the development and maturation of skeletal muscle. As a result, hub genes were identified, including PPARGC1A, MYOD1, EPAS1, IGF2, CXCR4, and APOA1, in all examined species. This study provided a better understanding of the relationships between genes and their biological pathways in the skeletal muscle maturation process.
Collapse
|
20
|
Tam WK, Cheung JPY, Koljonen PA, Kwan KYH, Cheung KM, Leung VYL. Slow twitch paraspinal muscle dysregulation in adolescent idiopathic scoliosis exhibiting HIF-2α misexpression. JOR Spine 2022; 5:e1227. [PMID: 36601371 PMCID: PMC9799082 DOI: 10.1002/jsp2.1227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/30/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022] Open
Abstract
Background Adolescent idiopathic scoliosis (AIS) refers to a three-dimensional spinal deformity which has a typical onset during adolescence. In most cases, the cause of the deformity cannot be clearly identified. Unbalanced paraspinal muscle activity in AIS patients was reported and hypoxia was implicated to regulate myogenesis. This study aims to investigate the association between myogenesis/muscle toning and HIF-αs activity in the pathogenesis of AIS. Methods HIF-αs expression was examined by enzyme-linked immunosorbent assay and western blot in paraspinal myoblasts isolated from 18 subjects who underwent deformity correction surgery. QPCR was conducted to measure the gene expression levels of perinatal muscle fiber markers MYH3, MYH8; slow twitch muscle fiber markers MHY7; fast twitch muscle fiber markers MYH4; and myogenic regulatory factors MYF5 and MYOG. Slow and fast twitch muscle fiber composition in concave/convex paraspinal musculature of AIS subjects was evaluated by immunostaining of myosin heavy chain type I (MyHC I) and myosin heavy chain type II (MyHC II). Results Reduced HIF-2α induction under hypoxia was found in paraspinal myoblast culture of 33% AIS subjects. We detected a suppression of perinatal and slow twitch muscle fiber associated genes, but not fast twitch muscle fiber-associated genes and myogenic regulatory factors in HIF-2α misexpressed AIS myoblasts. Distinct reduction of slow twitch muscle fiber was evidenced in convex paraspinal musculature, suggesting an asymmetric expression of slow twitch muscle fiber in HIF-2α misexpressed AIS patients. Conclusions This study indicates an association of abnormal HIF-2α expression in paraspinal myoblasts and a disproportionate slow twitch muscle fiber content in the convexity of the curvature in a subset of AIS subjects, suggesting HIF-2α dysregulation as a possible risk factor for AIS. The role of HIF-2α in paraspinal muscle function during spinal growth and its relevance in AIS prognosis warrants further investigation.
Collapse
Affiliation(s)
- Wai Kit Tam
- Department of Orthopaedics and TraumatologyThe University of Hong KongPokfulamHong Kong SARChina
| | - Jason P. Y. Cheung
- Department of Orthopaedics and TraumatologyThe University of Hong KongPokfulamHong Kong SARChina
- Orthopaedic Medical CenterHKU‐Shenzhen HospitalShenzhenChina
| | - Paul A. Koljonen
- Department of Orthopaedics and TraumatologyThe University of Hong KongPokfulamHong Kong SARChina
| | - Kenny Y. H. Kwan
- Department of Orthopaedics and TraumatologyThe University of Hong KongPokfulamHong Kong SARChina
- Orthopaedic Medical CenterHKU‐Shenzhen HospitalShenzhenChina
| | - Kenneth M.C. Cheung
- Department of Orthopaedics and TraumatologyThe University of Hong KongPokfulamHong Kong SARChina
- Orthopaedic Medical CenterHKU‐Shenzhen HospitalShenzhenChina
| | - Victor Y. L. Leung
- Department of Orthopaedics and TraumatologyThe University of Hong KongPokfulamHong Kong SARChina
- Orthopaedic Medical CenterHKU‐Shenzhen HospitalShenzhenChina
| |
Collapse
|
21
|
Bronisz-Budzyńska I, Kozakowska M, Pietraszek-Gremplewicz K, Madej M, Józkowicz A, Łoboda A, Dulak J. NRF2 Regulates Viability, Proliferation, Resistance to Oxidative Stress, and Differentiation of Murine Myoblasts and Muscle Satellite Cells. Cells 2022; 11:cells11203321. [PMID: 36291188 PMCID: PMC9600498 DOI: 10.3390/cells11203321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/27/2022] Open
Abstract
Increased oxidative stress can slow down the regeneration of skeletal muscle and affect the activity of muscle satellite cells (mSCs). Therefore, we evaluated the role of the NRF2 transcription factor (encoded by the Nfe2l2 gene), the main regulator of the antioxidant response, in muscle cell biology. We used (i) an immortalized murine myoblast cell line (C2C12) with stable overexpression of NRF2 and (ii) primary mSCs isolated from wild-type and Nfe2l2 (transcriptionally)-deficient mice (Nfe2l2tKO). NRF2 promoted myoblast proliferation and viability under oxidative stress conditions and decreased the production of reactive oxygen species. Furthermore, NRF2 overexpression inhibited C2C12 cell differentiation by down-regulating the expression of myogenic regulatory factors (MRFs) and muscle-specific microRNAs. We also showed that NRF2 is indispensable for the viability of mSCs since the lack of its transcriptional activity caused high mortality of cells cultured in vitro under normoxic conditions. Concomitantly, Nfe2l2tKO mSCs grown and differentiated under hypoxic conditions were viable and much more differentiated compared to cells isolated from wild-type mice. Taken together, NRF2 significantly influences the properties of myoblasts and muscle satellite cells. This effect might be modulated by the muscle microenvironment.
Collapse
|
22
|
Zhang L, Li D, Chang C, Sun Y. Myostatin/HIF2α-Mediated Ferroptosis is Involved in Skeletal Muscle Dysfunction in Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2022; 17:2383-2399. [PMID: 36185172 PMCID: PMC9519128 DOI: 10.2147/copd.s377226] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 09/12/2022] [Indexed: 11/23/2022] Open
Abstract
Objective Methods Results Conclusion
Collapse
Affiliation(s)
- Lijiao Zhang
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Danyang Li
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Chun Chang
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Yongchang Sun
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
- Correspondence: Yongchang Sun, Email
| |
Collapse
|
23
|
Divvela SSK, Offei EB, Suerland F, Revuelta García D, Kwiatkowski J, Balakrishnan-Renuka A, Bohne P, Böing M, Morosan-Puopolo G, Mark MD, Brand-Saberi B. Atonal homolog 8/Math6 regulates differentiation and maintenance of skeletal muscle. Front Cell Dev Biol 2022; 10:950414. [PMID: 36060799 PMCID: PMC9438786 DOI: 10.3389/fcell.2022.950414] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/06/2022] [Indexed: 12/16/2022] Open
Abstract
Atonal Homolog 8 (Atoh8) belongs to a large superfamily of transcriptional regulators called basic helix-loop-helix (bHLH) transcription factors. Atoh8 (murine homolog “Math6”) has been shown to be involved in organogenesis during murine embryonic development. We have previously identified the expression of Atoh8 during skeletal myogenesis in chicken where we described its involvement in hypaxial myotome formation suggesting a regulatory role of Atoh8 in skeletal muscle development. Within the current study, we analyzed the effect of the loss of function of Atoh8 in murine primary myoblasts and during differentiation of pluripotent stem cells into myotubes, and the effect of its gain of function in C2C12 cells. Based on the observed results, we conclude that Atoh8 regulates myoblast proliferation via modulating myostatin signaling. Further, our data revealed a reduced muscle mass, strength and fiber size with significant changes to the muscle fiber type suggesting atrophy in skeletal muscle of Atoh8 mutants. We further report that Atoh8 knockout mice suffer from a condition similar to ambient hypoxia which may be the primary cause of the phenotype. Altogether, this study shows the significance of Atoh8 not only in myogenesis but also in the maintenance of skeletal muscle.
Collapse
Affiliation(s)
| | - Eric Bekoe Offei
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
- University of Ghana, School of Veterinary Medicine, Legon, Ghana
| | - Florian Suerland
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - David Revuelta García
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Julia Kwiatkowski
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Ajeesh Balakrishnan-Renuka
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Pauline Bohne
- Department of Behavioral Neuroscience, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Marion Böing
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Gabriela Morosan-Puopolo
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Melanie D. Mark
- Department of Behavioral Neuroscience, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
- *Correspondence: Beate Brand-Saberi,
| |
Collapse
|
24
|
Brun CE, Sincennes MC, Lin AYT, Hall D, Jarassier W, Feige P, Le Grand F, Rudnicki MA. GLI3 regulates muscle stem cell entry into G Alert and self-renewal. Nat Commun 2022; 13:3961. [PMID: 35803939 PMCID: PMC9270324 DOI: 10.1038/s41467-022-31695-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/27/2022] [Indexed: 12/12/2022] Open
Abstract
Satellite cells are required for the growth, maintenance, and regeneration of skeletal muscle. Quiescent satellite cells possess a primary cilium, a structure that regulates the processing of the GLI family of transcription factors. Here we find that GLI3 processing by the primary cilium plays a critical role for satellite cell function. GLI3 is required to maintain satellite cells in a G0 dormant state. Strikingly, satellite cells lacking GLI3 enter the GAlert state in the absence of injury. Furthermore, GLI3 depletion stimulates expansion of the stem cell pool. As a result, satellite cells lacking GLI3 display rapid cell-cycle entry, increased proliferation and augmented self-renewal, and markedly enhanced regenerative capacity. At the molecular level, we establish that the loss of GLI3 induces mTORC1 signaling activation. Therefore, our results provide a mechanism by which GLI3 controls mTORC1 signaling, consequently regulating muscle stem cell activation and fate.
Collapse
Affiliation(s)
- Caroline E Brun
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Marie-Claude Sincennes
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Alexander Y T Lin
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Derek Hall
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - William Jarassier
- Univ Lyon, Univ Lyon 1, CNRS, INSERM, Pathophysiology and Genetics of Neuron and Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Peter Feige
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Fabien Le Grand
- Univ Lyon, Univ Lyon 1, CNRS, INSERM, Pathophysiology and Genetics of Neuron and Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Michael A Rudnicki
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada.
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
- Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
25
|
Patent highlights October–November 2021. Pharm Pat Anal 2022; 11:37-44. [DOI: 10.4155/ppa-2022-0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A snapshot of noteworthy recent developments in the patent literature of relevance to pharmaceutical and medical research and development.
Collapse
|
26
|
Elashry MI, Kinde M, Klymiuk MC, Eldaey A, Wenisch S, Arnhold S. The effect of hypoxia on myogenic differentiation and multipotency of the skeletal muscle-derived stem cells in mice. Stem Cell Res Ther 2022; 13:56. [PMID: 35123554 PMCID: PMC8817503 DOI: 10.1186/s13287-022-02730-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/20/2022] [Indexed: 01/01/2023] Open
Abstract
Abstract
Background
Skeletal muscle-derived stem cells (SC) have become a promising approach for investigating myogenic differentiation and optimizing tissue regeneration. Muscle regeneration is performed by SC, a self-renewal cell population underlying the basal lamina of muscle fibers. Here, we examined the impact of hypoxia condition on the regenerative capacity of SC either in their native microenvironment or via isolation in a monolayer culture using ectopic differentiation inductions. Furthermore, the effect of low oxygen tension on myogenic differentiation protocols of the myoblasts cell line C2C12 was examined.
Methods
Hind limb muscles of wild type mice were processed for both SC/fiber isolation and myoblast extraction using magnetic beads. SC were induced for myogenic, adipogenic and osteogenic commitments under normoxic (21% O2) and hypoxic (3% O2) conditions. SC proliferation and differentiation were evaluated using histological staining, immunohistochemistry, morphometric analysis and RT-qPCR. The data were statistically analyzed using ANOVA.
Results
The data revealed enhanced SC proliferation and motility following differentiation induction after 48 h under hypoxia. Following myogenic induction, the number of undifferentiated cells positive for Pax7 were increased at 72 h under hypoxia. Hypoxia upregulated MyoD and downregulated Myogenin expression at day-7 post-myogenic induction. Hypoxia promoted both SC adipogenesis and osteogenesis under respective induction as shown by using Oil Red O and Alizarin Red S staining. The expression of adipogenic markers; peroxisome proliferator activated receptor gamma (PPARγ) and fatty acid-binding protein 4 (FABP4) were upregulated under hypoxia up to day 14 compared to normoxic condition. Enhanced osteogenic differentiation was detected under hypoxic condition via upregulation of osteocalcin and osteopontin expression up to day 14 as well as, increased calcium deposition at day 21. Hypoxia exposure increases the number of adipocytes and the size of fat vacuoles per adipocyte compared to normoxic culture. Combining the differentiation medium with dexamethasone under hypoxia improves the efficiency of the myogenic differentiation protocol of C2C12 by increasing the length of the myotubes.
Conclusions
Hypoxia exposure increases cell resources for clinical applications and promotes SC multipotency and thus beneficial for tissue regeneration.
Collapse
|
27
|
Mori T, Onodera Y, Itokazu M, Takehara T, Shigi K, Iwawaki N, Akagi M, Teramura T. Depletion of NIMA-related kinase Nek2 induces aberrant self-renewal and apoptosis in stem/progenitor cells of aged muscular tissues. Mech Ageing Dev 2022; 201:111619. [PMID: 34995645 DOI: 10.1016/j.mad.2022.111619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/23/2021] [Accepted: 01/03/2022] [Indexed: 11/25/2022]
Abstract
Frailty of the locomotory organs has become a widespread problem in the geriatric population. The major factor leading to frailty is an age-associated decrease in muscular mass and a reduced number of muscular cells and myofibers. In aged muscular tissues, muscular satellite cells (MuSCs) are reduced due to abnormalities in their self-renewal and the induction of apoptosis. However, the molecular mechanisms connecting aging-associated physiological changes and the reduction of MuSCs are largely unknown. NIMA-related kinase 2 (Nek2), a member of the Nek family of serine/threonine kinases, was found to be downregulated in aged MuSCs/progenitors. Further, Nek2 downregulation was found to inhibit self-renewal and apoptotic cell death by activating the p53-dependent checkpoint. Attenuated NEK2 expression was also observed in the muscular tissues of elderly donors, and its function was confirmed to be conserved in humans. Overall, this study proposes a novel mechanism for inducing muscular atrophy to understand aging-associated muscular diseases.
Collapse
Affiliation(s)
| | - Yuta Onodera
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Japan
| | - Maki Itokazu
- Department of Rehabilitation Medicine, Kindai University Faculty of Medicine, Japan
| | - Toshiyuki Takehara
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Japan
| | - Kanae Shigi
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Japan
| | - Natsumi Iwawaki
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Japan
| | - Masao Akagi
- Department of Orthopedic Surgery, Kindai University Faculty of Medicine, Japan
| | - Takeshi Teramura
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Japan.
| |
Collapse
|
28
|
Control of satellite cell function in muscle regeneration and its disruption in ageing. Nat Rev Mol Cell Biol 2021; 23:204-226. [PMID: 34663964 DOI: 10.1038/s41580-021-00421-2] [Citation(s) in RCA: 231] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2021] [Indexed: 12/19/2022]
Abstract
Skeletal muscle contains a designated population of adult stem cells, called satellite cells, which are generally quiescent. In homeostasis, satellite cells proliferate only sporadically and usually by asymmetric cell division to replace myofibres damaged by daily activity and maintain the stem cell pool. However, satellite cells can also be robustly activated upon tissue injury, after which they undergo symmetric divisions to generate new stem cells and numerous proliferating myoblasts that later differentiate to muscle cells (myocytes) to rebuild the muscle fibre, thereby supporting skeletal muscle regeneration. Recent discoveries show that satellite cells have a great degree of population heterogeneity, and that their cell fate choices during the regeneration process are dictated by both intrinsic and extrinsic mechanisms. Extrinsic cues come largely from communication with the numerous distinct stromal cell types in their niche, creating a dynamically interactive microenvironment. This Review discusses the role and regulation of satellite cells in skeletal muscle homeostasis and regeneration. In particular, we highlight the cell-intrinsic control of quiescence versus activation, the importance of satellite cell-niche communication, and deregulation of these mechanisms associated with ageing. The increasing understanding of how satellite cells are regulated will help to advance muscle regeneration and rejuvenation therapies.
Collapse
|
29
|
Archacka K, Grabowska I, Mierzejewski B, Graffstein J, Górzyńska A, Krawczyk M, Różycka AM, Kalaszczyńska I, Muras G, Stremińska W, Jańczyk-Ilach K, Walczak P, Janowski M, Ciemerych MA, Brzoska E. Hypoxia preconditioned bone marrow-derived mesenchymal stromal/stem cells enhance myoblast fusion and skeletal muscle regeneration. Stem Cell Res Ther 2021; 12:448. [PMID: 34372911 PMCID: PMC8351116 DOI: 10.1186/s13287-021-02530-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/08/2021] [Indexed: 12/19/2022] Open
Abstract
Background The skeletal muscle reconstruction occurs thanks to unipotent stem cells, i.e., satellite cells. The satellite cells remain quiescent and localized between myofiber sarcolemma and basal lamina. They are activated in response to muscle injury, proliferate, differentiate into myoblasts, and recreate myofibers. The stem and progenitor cells support skeletal muscle regeneration, which could be disturbed by extensive damage, sarcopenia, cachexia, or genetic diseases like dystrophy. Many lines of evidence showed that the level of oxygen regulates the course of cell proliferation and differentiation. Methods In the present study, we analyzed hypoxia impact on human and pig bone marrow-derived mesenchymal stromal cell (MSC) and mouse myoblast proliferation, differentiation, and fusion. Moreover, the influence of the transplantation of human bone marrow-derived MSCs cultured under hypoxic conditions on skeletal muscle regeneration was studied. Results We showed that bone marrow-derived MSCs increased VEGF expression and improved myogenesis under hypoxic conditions in vitro. Transplantation of hypoxia preconditioned bone marrow-derived MSCs into injured muscles resulted in the improved cell engraftment and formation of new vessels. Conclusions We suggested that SDF-1 and VEGF secreted by hypoxia preconditioned bone marrow-derived MSCs played an essential role in cell engraftment and angiogenesis. Importantly, hypoxia preconditioned bone marrow-derived MSCs more efficiently engrafted injured muscles; however, they did not undergo myogenic differentiation. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02530-3.
Collapse
Affiliation(s)
- Karolina Archacka
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Iwona Grabowska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Bartosz Mierzejewski
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Joanna Graffstein
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Alicja Górzyńska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Marta Krawczyk
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Anna M Różycka
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Ilona Kalaszczyńska
- Department of Histology and Embryology, Medical University of Warsaw, 02-004, Warsaw, Poland.,Laboratory for Cell Research and Application, Medical University of Warsaw, 02-097, Warsaw, Poland
| | - Gabriela Muras
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Władysława Stremińska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Katarzyna Jańczyk-Ilach
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Piotr Walczak
- Department of Pathophysiology, Faculty of Medical Sciences, University of Warmia and Mazury, Warszawska 30 St, 10-082, Olsztyn, Poland.,Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, the Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Mirosław Janowski
- Center for Advanced Imaging Research, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD, 21201, USA.,NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawinskiego 5 St, 02-106, Warsaw, Poland
| | - Maria A Ciemerych
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Edyta Brzoska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland.
| |
Collapse
|
30
|
Pircher T, Wackerhage H, Aszodi A, Kammerlander C, Böcker W, Saller MM. Hypoxic Signaling in Skeletal Muscle Maintenance and Regeneration: A Systematic Review. Front Physiol 2021; 12:684899. [PMID: 34248671 PMCID: PMC8260947 DOI: 10.3389/fphys.2021.684899] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/26/2021] [Indexed: 12/26/2022] Open
Abstract
In skeletal muscle tissue, oxygen (O2) plays a pivotal role in both metabolism and the regulation of several intercellular pathways, which can modify proliferation, differentiation and survival of cells within the myogenic lineage. The concentration of oxygen in muscle tissue is reduced during embryogenesis and pathological conditions. Myogenic progenitor cells, namely satellite cells, are necessary for muscular regeneration in adults and are localized in a hypoxic microenvironment under the basal lamina, suggesting that the O2 level could affect their function. This review presents the effects of reduced oxygen levels (hypoxia) on satellite cell survival, myoblast regeneration and differentiation in vertebrates. Further investigations and understanding of the pathways involved in adult muscle regeneration during hypoxic conditions are maybe clinically relevant to seek for novel drug treatments for patients with severe muscle damage. We especially outlined the effect of hypoxia-inducible factor 1-alpha (HIF1A), the most studied transcriptional regulator of cellular and developmental response to hypoxia, whose investigation has recently been awarded with the Nobel price.
Collapse
Affiliation(s)
- Tamara Pircher
- Experimental Surgery and Regenerative Medicine, Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Munich, Germany
| | - Henning Wackerhage
- Faculty of Sport and Health Sciences, Technical University of Munich, Munich, Germany
| | - Attila Aszodi
- Experimental Surgery and Regenerative Medicine, Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Munich, Germany
| | - Christian Kammerlander
- Experimental Surgery and Regenerative Medicine, Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Munich, Germany
| | - Wolfgang Böcker
- Experimental Surgery and Regenerative Medicine, Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Munich, Germany
| | - Maximilian Michael Saller
- Experimental Surgery and Regenerative Medicine, Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Munich, Germany
| |
Collapse
|
31
|
Ding H, Chen S, Pan X, Dai X, Pan G, Li Z, Mai X, Tian Y, Zhang S, Liu B, Cao G, Yao Z, Yao X, Gao L, Yang L, Chen X, Sun J, Chen H, Han M, Yin Y, Xu G, Li H, Wu W, Chen Z, Lin J, Xiang L, Hu J, Lu Y, Zhu X, Xie L. Transferrin receptor 1 ablation in satellite cells impedes skeletal muscle regeneration through activation of ferroptosis. J Cachexia Sarcopenia Muscle 2021; 12:746-768. [PMID: 33955709 PMCID: PMC8200440 DOI: 10.1002/jcsm.12700] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/18/2021] [Accepted: 03/16/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Satellite cells (SCs) are critical to skeletal muscle regeneration. Inactivation of SCs is linked to skeletal muscle loss. Transferrin receptor 1 (Tfr1) is associated with muscular dysfunction as muscle-specific deletion of Tfr1 results in growth retardation, metabolic disorder, and lethality, shedding light on the importance of Tfr1 in muscle physiology. However, its physiological function regarding skeletal muscle ageing and regeneration remains unexplored. METHODS RNA sequencing is applied to skeletal muscles of different ages to identify Tfr1 associated to skeletal muscle ageing. Mice with conditional SC ablation of Tfr1 were generated. Between Tfr1SC/WT and Tfr1SC/KO (n = 6-8 mice per group), cardiotoxin was intramuscularly injected, and transverse abdominal muscle was dissected, weighted, and cryosectioned, followed by immunostaining, haematoxylin and eosin staining, and Masson staining. These phenotypical analyses were followed with functional analysis such as flow cytometry, tread mill, Prussian blue staining, and transmission electron microscopy to identify pathological pathways that contribute to regeneration defects. RESULTS By comparing gene expression between young (2 weeks old, n = 3) and aged (80 weeks old, n = 3) mice among four types of muscles, we identified that Tfr1 expression is declined in muscles of aged mice (~80% reduction, P < 0.005), so as to its protein level in SCs of aged mice. From in vivo and ex vivo experiments, Tfr1 deletion in SCs results in an irreversible depletion of SCs (~60% reduction, P < 0.005) and cell-autonomous defect in SC proliferation and differentiation, leading to skeletal muscle regeneration impairment, followed by labile iron accumulation, lipogenesis, and decreased Gpx4 and Nrf2 protein levels leading to reactive oxygen species scavenger defects. These abnormal phenomena including iron accumulation, activation of unsaturated fatty acid biosynthesis, and lipid peroxidation are orchestrated with the occurrence of ferroptosis in skeletal muscle. Ferroptosis further exacerbates SC proliferation and skeletal muscle regeneration. Ferrostatin-1, a ferroptosis inhibitor, could not rescue ferroptosis. However, intramuscular administration of lentivirus-expressing Tfr1 could partially reduce labile iron accumulation, decrease lipogenesis, and promote skeletal muscle regeneration. Most importantly, declined Tfr1 but increased Slc39a14 protein level on cellular membrane contributes to labile iron accumulation in skeletal muscle of aged rodents (~80 weeks old), leading to activation of ferroptosis in aged skeletal muscle. This is inhibited by ferrostatin-1 to improve running time (P = 0.0257) and distance (P = 0.0248). CONCLUSIONS Satellite cell-specific deletion of Tfr1 impairs skeletal muscle regeneration with activation of ferroptosis. This phenomenon is recapitulated in skeletal muscle of aged rodents and human sarcopenia. Our study provides mechanistic information for developing novel therapeutic strategies against muscular ageing and diseases.
Collapse
Affiliation(s)
- Hongrong Ding
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern ChinaInstitute of Microbiology, Guangdong Academy of SciencesGuangzhouChina
- Guangdong Provincial Key Laboratory of Molecular Diagnosis, The Marine Biomedical Research InstituteGuangdong Medical UniversityZhanjiangChina
| | - Shujie Chen
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern ChinaInstitute of Microbiology, Guangdong Academy of SciencesGuangzhouChina
- Department of Endocrinology and MetabolismZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Xiaohan Pan
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern ChinaInstitute of Microbiology, Guangdong Academy of SciencesGuangzhouChina
| | - Xiaoshuang Dai
- BGI Institute of Applied AgricultureBGI‐ShenzhenShenzhenChina
| | - Guihua Pan
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern ChinaInstitute of Microbiology, Guangdong Academy of SciencesGuangzhouChina
| | - Ze Li
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern ChinaInstitute of Microbiology, Guangdong Academy of SciencesGuangzhouChina
- Department of Endocrinology and MetabolismZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Xudong Mai
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern ChinaInstitute of Microbiology, Guangdong Academy of SciencesGuangzhouChina
- Department of Endocrinology and MetabolismZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Ye Tian
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern ChinaInstitute of Microbiology, Guangdong Academy of SciencesGuangzhouChina
- Department of Endocrinology and MetabolismZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Susu Zhang
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern ChinaInstitute of Microbiology, Guangdong Academy of SciencesGuangzhouChina
- Department of Endocrinology and MetabolismZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Bingdong Liu
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern ChinaInstitute of Microbiology, Guangdong Academy of SciencesGuangzhouChina
| | - Guangchao Cao
- The Biomedical Translational Research Institute, Faculty of Medical ScienceJinan UniversityGuangzhouChina
| | - Zhicheng Yao
- The Third Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
| | - Xiangping Yao
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern ChinaInstitute of Microbiology, Guangdong Academy of SciencesGuangzhouChina
- Department of Endocrinology and MetabolismZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Liang Gao
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern ChinaInstitute of Microbiology, Guangdong Academy of SciencesGuangzhouChina
| | - Li Yang
- The Third Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
| | - Xiaoyan Chen
- The Third Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
| | - Jia Sun
- Department of Endocrinology and MetabolismZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Hong Chen
- Department of Endocrinology and MetabolismZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Mulan Han
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern ChinaInstitute of Microbiology, Guangdong Academy of SciencesGuangzhouChina
| | - Yulong Yin
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern ChinaInstitute of Microbiology, Guangdong Academy of SciencesGuangzhouChina
- China Institute of Subtropical AgricultureChinese Academy of SciencesChangshaHunanChina
| | - Guohuan Xu
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern ChinaInstitute of Microbiology, Guangdong Academy of SciencesGuangzhouChina
| | - Huijun Li
- College of Public HealthXinxiang Medical UniversityXinxiangChina
| | - Weidong Wu
- College of Public HealthXinxiang Medical UniversityXinxiangChina
| | - Zheng Chen
- HIT Center for Life Sciences, School of Life Science and TechnologyHarbin Institute of TechnologyHarbinChina
| | - Jingchao Lin
- Metabo‐Profile Biotechnology (Shanghai) Co. Ltd.ShanghaiChina
| | - Liping Xiang
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Endocrinology and MetabolismZhongshan Hospital, Fudan UniversityShanghaiChina
| | - Jun Hu
- Department of OrthopedicsThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
| | - Yan Lu
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Endocrinology and MetabolismZhongshan Hospital, Fudan UniversityShanghaiChina
| | - Xiao Zhu
- Guangdong Provincial Key Laboratory of Molecular Diagnosis, The Marine Biomedical Research InstituteGuangdong Medical UniversityZhanjiangChina
| | - Liwei Xie
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern ChinaInstitute of Microbiology, Guangdong Academy of SciencesGuangzhouChina
- Department of Endocrinology and MetabolismZhujiang Hospital, Southern Medical UniversityGuangzhouChina
- College of Public HealthXinxiang Medical UniversityXinxiangChina
| |
Collapse
|
32
|
Ban HS, Uto Y, Nakamura H. Hypoxia-inducible factor (HIF) inhibitors: a patent survey (2016-2020). Expert Opin Ther Pat 2021; 31:387-397. [PMID: 33455469 DOI: 10.1080/13543776.2021.1874345] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Introduction: Hypoxia-inducible factor (HIF) is a master regulator of oxygen homeostasis. The increased expression of genes targeted by HIF is associated with many human diseases, including ischemic cardiovascular disease, stroke, chronic lung disease, and cancer.Areas covered: This patent survey summarizes the information about patented HIF inhibitors over the last 5 years.Expert opinion: HIF inhibitors have shown promise for the treatment of hypoxic pulmonary hypertension, a circadian rhythm disorder, calcific aortic valve disease, cerebrovascular accident, and heterotopic ossification. In addition, HIF-2α inhibitors can be used for the treatment or prevention of iron overload disorders, Crohn's disease, ulcerative colitis, and thyroid eye disease, or to improve muscle generation and repair. PT2385 completed phase I clinical trials for the treatment of clear cell renal cell carcinoma. It exerted a higher synergistic inhibitory effect on tumor growth in combination with anti-PD-1 antibody, in comparison with each treatment alone, indicating that effective immunotherapy for solid tumors counteracts of the immunosuppression induced by hypoxia. Therefore, considering the effects of hypoxia on cancer cells, stromal cells, and effector immune cells, it is important to develop inhibitors of molecular pathways activated by hypoxia for successful treatments.
Collapse
Affiliation(s)
- Hyun Seung Ban
- Biomedical Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Yoshikazu Uto
- ASCA Company, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Hiroyuki Nakamura
- cLaboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| |
Collapse
|
33
|
Xiong Z, Xiong W, Xiao W, Yuan C, Shi J, Huang Y, Wang C, Meng X, Chen Z, Yang H, Chen K, Zhang X. NNT-induced tumor cell "slimming" reverses the pro-carcinogenesis effect of HIF2a in tumors. Clin Transl Med 2021; 11:e264. [PMID: 33463050 PMCID: PMC7803359 DOI: 10.1002/ctm2.264] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND HIF2a and lipid accumulation play key roles in the progression of clear cell renal cell carcinoma (ccRCC). Tumor cell "slimming" is a new concept in which tumor cells with abnormal lipids efficiently consume lipids to inhibit tumor progression without producing additional ATP. However, their respective regulatory mechanisms are still unclear. The purpose of this study is uncovering the links between these three key elements of ccRCC to elucidate new mechanisms of ccRCC metabolic abnormalities and providing a basis for new drug development for ccRCC. METHODS Bioinformatics screening and analyses were performed in ccRCC according to TCGA-KIRC database. qRT-PCR, luciferase reporter assay, western blot, chromatin immunoprecipitation (ChIP) assays, and other biological methods were used to explore and verify related pathways. Various cell line models and animal models were used to perform related functional experiments. RESULTS Screening based on sequencing data after HIF2a knockdown and three independent mitochondrial metabolism-related gene sets showed that nicotinamide nucleotide transhydrogenase (NNT) was a mediator between HIF2a and tumor cells "slimming." Further research showed that NNT had significant prognostic predictive value and was downregulated in ccRCC. It is regulated by HIF2a and can significantly activate lipid browning-mediated tumor cell "slimming." Mechanistic investigations indicated that HIF2a enhanced the expression of miR-455-5p via binding to HIF2a-related response elements in the miR-455-5p promoter, which suppresses NNT expression by binding to its 3' untranslated region. CONCLUSIONS Our study revealed a novel mechanism by which HIF2a decreased NNT level through a microRNA that suppressed tumor cell "slimming," resulting in the progression of ccRCC. This mechanism provides a fresh perspective of lipid accumulation in ccRCC and may help target novel strategies for the treatment of tumors with abnormal lipid metabolism.
Collapse
Affiliation(s)
- Zhiyong Xiong
- Department of UrologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of UrologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Wei Xiong
- Department of NephrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Wen Xiao
- Department of UrologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of UrologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Changfei Yuan
- Department of UrologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of UrologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jian Shi
- Department of UrologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of UrologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yu Huang
- Department of UrologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of UrologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Cheng Wang
- Department of UrologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of UrologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiangui Meng
- Department of UrologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of UrologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Zhixian Chen
- Department of UrologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of UrologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hongmei Yang
- Department of Pathogenic BiologySchool of Basic MedicineHuazhong University of Science and TechnologyWuhanChina
| | - Ke Chen
- Department of UrologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of UrologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaoping Zhang
- Department of UrologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of UrologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
34
|
Cirillo F, Resmini G, Angelino E, Ferrara M, Tarantino A, Piccoli M, Rota P, Ghiroldi A, Monasky MM, Ciconte G, Pappone C, Graziani A, Anastasia L. HIF-1α Directly Controls WNT7A Expression During Myogenesis. Front Cell Dev Biol 2020; 8:593508. [PMID: 33262987 PMCID: PMC7686515 DOI: 10.3389/fcell.2020.593508] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/20/2020] [Indexed: 11/13/2022] Open
Abstract
Herein we unveil that Hypoxia-inducible factor-1α (HIF-1α) directly regulates WNT7A expression during myogenesis. In fact, chromatin immunoprecipitation (ChiP) and site-directed mutagenesis experiments revealed two distinct hypoxia response elements (HREs) that are specific HIF-1α binding sites on the WNT7A promoter. Remarkably, a pharmacological activation of HIF-1α induced WNT7A expression and enhanced muscle differentiation. On the other hand, silencing of WNT7A using CRISPR/Cas9 genome editing blocked the effects of HIF-1α activation on myogenesis. Finally, treatment with prolyl hydroxylases (PHDs) inhibitors improved muscle regeneration in vitro and in vivo in a cardiotoxin (CTX)-induced muscle injury mouse model, paving the way for further studies to test its efficacy on acute and chronic muscular pathologies.
Collapse
Affiliation(s)
- Federica Cirillo
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Giulia Resmini
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Elia Angelino
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Michele Ferrara
- Division of Genetics and Cell Biology, Chromatin Dynamics Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Adriana Tarantino
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, San Donato Milanese, Italy.,Arrhythmology Department, IRCCS Policlinico San Donato, Milan, Italy
| | - Marco Piccoli
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Paola Rota
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, San Donato Milanese, Italy.,Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Andrea Ghiroldi
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | | | - Giuseppe Ciconte
- Arrhythmology Department, IRCCS Policlinico San Donato, Milan, Italy
| | - Carlo Pappone
- Arrhythmology Department, IRCCS Policlinico San Donato, Milan, Italy.,Vita-Salute San Raffaele University, Faculty of Medicine, Milan, Italy
| | - Andrea Graziani
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Luigi Anastasia
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, San Donato Milanese, Italy.,Vita-Salute San Raffaele University, Faculty of Medicine, Milan, Italy
| |
Collapse
|
35
|
Endo Y, Baldino K, Li B, Zhang Y, Sakthivel D, MacArthur M, Panayi AC, Kip P, Spencer DJ, Jasuja R, Bagchi D, Bhasin S, Nuutila K, Neppl RL, Wagers AJ, Sinha I. Loss of ARNT in skeletal muscle limits muscle regeneration in aging. FASEB J 2020; 34:16086-16104. [PMID: 33064329 PMCID: PMC7756517 DOI: 10.1096/fj.202000761rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 09/23/2020] [Accepted: 09/28/2020] [Indexed: 12/11/2022]
Abstract
The ability of skeletal muscle to regenerate declines significantly with aging. The expression of aryl hydrocarbon receptor nuclear translocator (ARNT), a critical component of the hypoxia signaling pathway, was less abundant in skeletal muscle of old (23-25 months old) mice. This loss of ARNT was associated with decreased levels of Notch1 intracellular domain (N1ICD) and impaired regenerative response to injury in comparison to young (2-3 months old) mice. Knockdown of ARNT in a primary muscle cell line impaired differentiation in vitro. Skeletal muscle-specific ARNT deletion in young mice resulted in decreased levels of whole muscle N1ICD and limited muscle regeneration. Administration of a systemic hypoxia pathway activator (ML228), which simulates the actions of ARNT, rescued skeletal muscle regeneration in both old and ARNT-deleted mice. These results suggest that the loss of ARNT in skeletal muscle is partially responsible for diminished myogenic potential in aging and activation of hypoxia signaling holds promise for rescuing regenerative activity in old muscle.
Collapse
Affiliation(s)
- Yori Endo
- Division of Plastic SurgeryBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Kodi Baldino
- Division of Plastic SurgeryBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Bin Li
- Division of Plastic SurgeryBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
- Department of Plastic and Aesthetic SurgeryNanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Yuteng Zhang
- Division of Plastic SurgeryBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
- Department of Plastic and Aesthetic SurgeryNanfang HospitalSouthern Medical UniversityGuangzhouChina
| | | | - Michael MacArthur
- Department of Genetics and Complex DiseasesHarvard School of Public HealthBostonMAUSA
- Division of Vascular and Endovascular SurgeryBrigham and Women's HospitalBostonMAUSA
| | - Adriana C. Panayi
- Division of Plastic SurgeryBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Peter Kip
- Division of Vascular and Endovascular SurgeryBrigham and Women's HospitalBostonMAUSA
| | | | - Ravi Jasuja
- Division of EndocrinologyBrigham and Women's HospitalBostonMAUSA
| | - Debalina Bagchi
- Department of Orthopedic SurgeryBrigham and Women's Hospital, Harvard Medical SchoolBostonMAUSA
| | - Shalender Bhasin
- Division of EndocrinologyBrigham and Women's HospitalBostonMAUSA
| | - Kristo Nuutila
- Division of Plastic SurgeryBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Ronald L. Neppl
- Department of Orthopedic SurgeryBrigham and Women's Hospital, Harvard Medical SchoolBostonMAUSA
| | - Amy J. Wagers
- Joslin Diabetes CenterBostonMAUSA
- Harvard Department of Stem Cell and Regenerative BiologyHarvard Stem Cell InstituteCambridgeMAUSA
- Paul F. Glenn Center for the Biology of AgingHarvard Medical SchoolBostonMAUSA
| | - Indranil Sinha
- Division of Plastic SurgeryBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
- Harvard Department of Stem Cell and Regenerative BiologyHarvard Stem Cell InstituteCambridgeMAUSA
| |
Collapse
|
36
|
Buchanan SM, Price FD, Castiglioni A, Gee AW, Schneider J, Matyas MN, Hayhurst M, Tabebordbar M, Wagers AJ, Rubin LL. Pro-myogenic small molecules revealed by a chemical screen on primary muscle stem cells. Skelet Muscle 2020; 10:28. [PMID: 33036659 PMCID: PMC7547525 DOI: 10.1186/s13395-020-00248-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/14/2020] [Indexed: 11/10/2022] Open
Abstract
Satellite cells are the canonical muscle stem cells that regenerate damaged skeletal muscle. Loss of function of these cells has been linked to reduced muscle repair capacity and compromised muscle health in acute muscle injury and congenital neuromuscular diseases. To identify new pathways that can prevent loss of skeletal muscle function or enhance regenerative potential, we established an imaging-based screen capable of identifying small molecules that promote the expansion of freshly isolated satellite cells. We found several classes of receptor tyrosine kinase (RTK) inhibitors that increased freshly isolated satellite cell numbers in vitro. Further exploration of one of these compounds, the RTK inhibitor CEP-701 (also known as lestaurtinib), revealed potent activity on mouse satellite cells both in vitro and in vivo. This expansion potential was not seen upon exposure of proliferating committed myoblasts or non-myogenic fibroblasts to CEP-701. When delivered subcutaneously to acutely injured animals, CEP-701 increased both the total number of satellite cells and the rate of muscle repair, as revealed by an increased cross-sectional area of regenerating fibers. Moreover, freshly isolated satellite cells expanded ex vivo in the presence of CEP-701 displayed enhanced muscle engraftment potential upon in vivo transplantation. We provide compelling evidence that certain RTKs, and in particular RET, regulate satellite cell expansion during muscle regeneration. This study demonstrates the power of small molecule screens of even rare adult stem cell populations for identifying stem cell-targeting compounds with therapeutic potential.
Collapse
Affiliation(s)
- Sean M Buchanan
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Feodor D Price
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Alessandra Castiglioni
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA.,Cancer Immunology Department, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Amanda Wagner Gee
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Joel Schneider
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Mark N Matyas
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Monica Hayhurst
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Mohammadsharif Tabebordbar
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Amy J Wagers
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Lee L Rubin
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA.
| |
Collapse
|
37
|
Overfeeding and Substrate Availability, But Not Age or BMI, Alter Human Satellite Cell Function. Nutrients 2020; 12:nu12082215. [PMID: 32722351 PMCID: PMC7468931 DOI: 10.3390/nu12082215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/09/2020] [Accepted: 07/21/2020] [Indexed: 12/25/2022] Open
Abstract
Satellite cells (SC) aid skeletal muscle growth and regeneration. SC-mediated skeletal muscle repair can both be influenced by and exacerbate several diseases linked to a fatty diet, obesity, and aging. The purpose of this study was to evaluate the effects of different lifestyle factors on SC function, including body mass index (BMI), age, and high-fat overfeeding. For this study, SCs were isolated from the vastus lateralis of sedentary young (18–30 years) and sedentary older (60–80 years) men with varying BMIs (18–32 kg/m2), as well as young sedentary men before and after four weeks of overfeeding (OVF) (55% fat/ + 1000 kcal, n = 4). The isolated SCs were then treated in vitro with a control (5 mM glucose, 10% fetal bovine serum (FBS)) or a high substrate growth media (HSM) (10% FBS, 25 mM glucose, and 400 μM 2:1 oleate–palmitate). Cells were assessed on their ability to proliferate, differentiate, and fuel substrate oxidation after differentiation. The effect of HSM was measured as the percentage difference between SCs exposed to HSM compared to control media. In vitro SC function was not affected by donor age. OVF reduced SC proliferation rates (–19% p < 0.05) but did not influence differentiation. Cellular proliferation in response to HSM was correlated to the donor’s body mass index (BMI) (r2 = 0.6121, p < 0.01). When exposed to HSM, SCs from normal weight (BMI 18–25 kg/m2) participants exhibited reduced proliferation and fusion rates with increased fatty-acid oxidation (p < 0.05), while SCs from participants with higher BMIs (BMI 25–32 kg/m2) demonstrated enhanced proliferation in HSM. HSM reduced proliferation and fusion (p < 0.05) in SCs isolated from subjects before OVF, whereas HSM exposure accelerated proliferation and fusion in SCs collected following OVF. These results indicated that diet has a greater influence on SC function than age and BMI. Though age and BMI do not influence in vitro SC function when grown in controlled conditions, both factors influenced the response of SCs to substrate challenges, indicating age and BMI may mediate responses to diet.
Collapse
|
38
|
Prolyl hydroxylase domain 2 reduction enhances skeletal muscle tissue regeneration after soft tissue trauma in mice. PLoS One 2020; 15:e0233261. [PMID: 32413092 PMCID: PMC7228053 DOI: 10.1371/journal.pone.0233261] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/01/2020] [Indexed: 12/13/2022] Open
Abstract
The transcription factor Hypoxia-inducible factor 1 (HIF-1) plays a pivotal role in tissue regeneration. HIF-1 is negatively controlled by O2-dependent prolyl hydroxylases with a predominant role of prolyl hydroxylase 2 isoform (Phd2). Transgenic mice, hypomorphic for this isoform, accumulate more HIF-1 under normoxic conditions. Using these mice, we investigated the influence of Phd2 and HIF-1 on the regenerative capability of skeletal muscle tissue after myotrauma. Phd2-hypomorphic and wild type mice (on C57Bl/6 background) were grouped with regeneration times from 6 to 168 hours after closed mechanic muscle trauma to the hind limb. Tissue samples were analysed by immuno-staining and real-time PCR. Bone marrow derived macrophages of wild type and Phd2-hypomorphic mice were isolated and analysed via flow cytometry and quantitative real-time PCR. Phd2 reduction led to a higher regenerative capability due to enhanced activation of myogenic factors accompanied by induction of genes responsible for glucose and lactate metabolism in Phd2-hypomorphic mice. Macrophage infiltration into the trauma areas in hypomorphic mice started earlier and was more pronounced compared to wild type mice. Phd2-hypomorphic mice also showed higher numbers of macrophages in areas with sustained trauma 72 hours after myotrauma application. In conclusion, we postulate that the HIF-1 pathway is activated secondary to a Phd2 reduction which may lead to i) higher activation of myogenic factors, ii) increased number of positive stem cell proliferation markers, and iii) accelerated macrophage recruitment to areas of trauma, resulting in faster muscle tissue regeneration after myotrauma. With the current development of prolyl hydroxylase domain inhibitors, our findings point towards a potential clinical benefit after myotrauma.
Collapse
|
39
|
Valle-Tenney R, Rebolledo D, Acuña MJ, Brandan E. HIF-hypoxia signaling in skeletal muscle physiology and fibrosis. J Cell Commun Signal 2020; 14:147-158. [PMID: 32088838 DOI: 10.1007/s12079-020-00553-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 02/11/2020] [Indexed: 02/06/2023] Open
Abstract
Hypoxia refers to the decrease in oxygen tension in the tissues, and the central effector of the hypoxic response is the transcription factor Hypoxia-Inducible Factor α (HIF1-α). Transient hypoxia in acute events, such as exercising or regeneration after damage, play an important role in skeletal muscle physiology and homeostasis. However, sustained activation of hypoxic signaling is a feature of skeletal muscle injury and disease, which can be a consequence of chronic damage but can also increase the severity of the pathology and worsen its outcome. Here, we review evidence that supports the idea that hypoxia and HIF-1α can contribute to the establishment of fibrosis in skeletal muscle through its crosstalk with other profibrotic factors, such as Transforming growth factor β (TGF-β), the induction of profibrotic cytokines expression, as is the case of Connective Tissue Growth Factor (CTGF/CCN2), or being the target of the Renin-angiotensin system (RAS).
Collapse
Affiliation(s)
- Roger Valle-Tenney
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile.,Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniela Rebolledo
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.,Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O Higgins, Santiago, Chile
| | - María José Acuña
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile.,Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O Higgins, Santiago, Chile
| | - Enrique Brandan
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile. .,Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Fundación Ciencia & Vida, Santiago, Chile. .,Department Cell and Molecular Biology, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
40
|
Nichenko AS, Southern WM, Tehrani KF, Qualls AE, Flemington AB, Mercer GH, Yin A, Mortensen LJ, Yin H, Call JA. Mitochondrial-specific autophagy linked to mitochondrial dysfunction following traumatic freeze injury in mice. Am J Physiol Cell Physiol 2019; 318:C242-C252. [PMID: 31721614 DOI: 10.1152/ajpcell.00123.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The objective of this study was to interrogate the link between mitochondrial dysfunction and mitochondrial-specific autophagy in skeletal muscle. C57BL/6J mice were used to establish a time course of mitochondrial function and autophagy induction after fatigue (n = 12), eccentric contraction-induced injury (n = 20), or traumatic freeze injury (FI, n = 28); only FI resulted in a combination of mitochondrial dysfunction, i.e., decreased mitochondrial respiration, and autophagy induction. Moving forward, we tested the hypothesis that mitochondrial-specific autophagy is important for the timely recovery of mitochondrial function after FI. Following FI, there is a significant increase in several mitochondrial-specific autophagy-related protein contents including dynamin-related protein 1 (Drp1), BCL1 interacting protein (BNIP3), Pink1, and Parkin (~2-fold, P < 0.02). Also, mitochondrial-enriched fractions from FI muscles showed microtubule-associated protein light chain B1 (LC3)II colocalization suggesting autophagosome assembly around the damaged mitochondrial. Unc-51 like autophagy activating kinase (Ulk1) is considered necessary for mitochondrial-specific autophagy and herein we utilized a mouse model with Ulk1 deficiency in adult skeletal muscle (myogenin-Cre). While Ulk1 knockouts had contractile weakness compared with littermate controls (-27%, P < 0.02), the recovery of mitochondrial function was not different, and this may be due in part to a partial rescue of Ulk1 protein content within the regenerating muscle tissue of knockouts from differentiated satellite cells in which Ulk1 was not genetically altered via myogenin-Cre. Lastly, autophagy flux was significantly less in injured versus uninjured muscles (-26%, P < 0.02) despite the increase in autophagy-related protein content. This suggests autophagy flux is not upregulated to match increases in autophagy machinery after injury and represents a potential bottleneck in the clearance of damaged mitochondria by autophagy.
Collapse
Affiliation(s)
- Anna S Nichenko
- Department of Kinesiology, University of Georgia, Athens, Georgia.,Regenerative Bioscience Center, University of Georgia, Athens, Georgia
| | - W Michael Southern
- Department of Kinesiology, University of Georgia, Athens, Georgia.,Regenerative Bioscience Center, University of Georgia, Athens, Georgia
| | | | - Anita E Qualls
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia
| | | | - Grant H Mercer
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia
| | - Amelia Yin
- Center for Molecular Medicine, University of Georgia, Athens, Georgia.,Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia
| | - Luke J Mortensen
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia
| | - Hang Yin
- Center for Molecular Medicine, University of Georgia, Athens, Georgia.,Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia
| | - Jarrod A Call
- Department of Kinesiology, University of Georgia, Athens, Georgia.,Regenerative Bioscience Center, University of Georgia, Athens, Georgia
| |
Collapse
|
41
|
Cui W, Liu CX, Wang J, Zhang YC, Shen Q, Feng ZH, Wu J, Li JX. An oleanolic acid derivative reduces denervation-induced muscle atrophy via activation of CNTF-mediated JAK2/STAT3 signaling pathway. Eur J Pharmacol 2019; 861:172612. [PMID: 31421088 DOI: 10.1016/j.ejphar.2019.172612] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 08/10/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023]
Abstract
Denervation caused by sciatic nerve injury has brought great harm to the patients, especially denervation-induced muscle atrophy. The body stress produces a large number of Schwann cells when the sciatic nerve is injured, and the cells secrete some cytokines including ciliary neurotrophic factor (CNTF) that not only play a role in promoting the repair of sciatic nerve, but also maintain the normal physiological function of the muscles surrounding the damaged nerves. CNTF upregulates janus kinase 2 (JAK2) and signal transducers and activators of transcription 3 (STAT3) signals in myoblasts, and consequently accelerates the proliferation and differentiation of myoblasts. This effect on myoblasts is the most effective way to relieve muscle atrophy. Therefore, increasing CNTF is a promising direction to improve muscle atrophy. In the present study, an oleanolic acid derivative, HA-19, increased the proliferation of Schwann cells, and elevated CNTF production of the cells. HA-19 up-regulated the phosphorylation of JAK2 and STAT3 not only by directly acting on myoblasts, but also by increasing the secretion of CNTF of Schwann cells; and consequently, promoted the proliferation and differentiation of myoblasts. In denervation-induced muscle atrophy mice model, treatment with HA-19 significantly increased the weights of tibialis anterior (TA), gastrocnemius (Gastroc.), extensor digitorum longus (EDL), soleus and quadriceps (Quad.) under atrophied state. And, very interestingly, these muscles under normal condition were also strengthened by HA-19. Our finding demonstrated that HA-19 has a great potential as a lead compound for the drug discovery of anti-denervation-induced muscle atrophy.
Collapse
Affiliation(s)
- Wei Cui
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Chen-Xi Liu
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Jie Wang
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yu-Chao Zhang
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Qi Shen
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Zhen-Hua Feng
- The Center of Diagnosis and Treatment for Joint Disease, Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, 210008, China
| | - Jing Wu
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China.
| | - Jian-Xin Li
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China.
| |
Collapse
|
42
|
Lampert MA, Gustafsson ÅB. Mitochondria and autophagy in adult stem cells: proliferate or differentiate. J Muscle Res Cell Motil 2019; 41:355-362. [PMID: 31313217 DOI: 10.1007/s10974-019-09542-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 07/10/2019] [Indexed: 12/13/2022]
Abstract
Adult stem cells are undifferentiated cells that are found in many different tissues after development. They are responsible for regenerating and repairing tissues after injury, as well as replacing cells when needed. Adult stem cells maintain a delicate balance between self-renewal to prevent depletion of the stem cell pool and differentiation to continually replenish downstream lineages. The important role of mitochondria in generating energy, calcium storage and regulating cell death is well established. However, new research has linked mitochondria to stem cell maintenance and fate. In addition, efficient mitochondrial quality control is critical for stem cell homeostasis to ensure their long-term survival in tissues. In this review, we discuss the latest evidence linking mitochondrial function, remodeling and turnover via autophagy to regulation of adult stem cell self-renewal and differentiation.
Collapse
Affiliation(s)
- Mark A Lampert
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive #0751, La Jolla, CA, 92093-0751, USA
| | - Åsa B Gustafsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive #0751, La Jolla, CA, 92093-0751, USA.
| |
Collapse
|
43
|
Cui W, Liu CX, Zhang YC, Shen Q, Feng ZH, Wang J, Lu SF, Wu J, Li JX. A novel oleanolic acid derivative HA-19 ameliorates muscle atrophy via promoting protein synthesis and preventing protein degradation. Toxicol Appl Pharmacol 2019; 378:114625. [PMID: 31201822 DOI: 10.1016/j.taap.2019.114625] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/18/2019] [Accepted: 06/11/2019] [Indexed: 12/13/2022]
Abstract
Muscle atrophy refers to a decrease in the size of muscles in the body, occurs in certain muscles with inactivity in many diseases and lacks effective therapies up to date. Natural products still play an important role in drug discovery. In the present study, derivatives of a natural product, oleanolic acid, were screened with myoblast differentiation and myotube atrophy assays, respectively. Results revealed that one of the derivatives, HA-19 showed the most potent anti-muscle atrophy activity, and was used for further studies. We demonstrated that HA-19 led to the increase of the protein synthesis by activating mechanistic target of rapamycin complex 1 (mTORC1)/p70 S6K pathways, and also enhanced myoblast proliferation and terminal differentiation via up-regulating of the myogenic transcription factors Pax7, MyoD and Myogenin. The interesting thing was that HA-19 also suppressed protein degradation to prevent myotube atrophy by down-regulating negative growth factors, FoxO1, MuRF1 and Atrogin-1. The results were also supported by puromycin labelling and protein ubiquitination assays. These data revealed that HA-19 possessed a "dual effect" on inhibition of muscle atrophy. In disuse-induced muscle atrophy mice model, HA-19 treatment significantly increased the weights of bilateral tibialis anterior (TA), gastrocnemius (Gastroc.), quadriceps (Quad.), suggesting the effectiveness of HA-19 to remit disuse-induced muscle atrophy. Our finding demonstrated that HA-19 has a great potential as an inhibitor or lead compound for the anti-muscle atrophy drug discovery.
Collapse
Affiliation(s)
- Wei Cui
- State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Chen-Xi Liu
- State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yu-Chao Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Qi Shen
- State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Zhen-Hua Feng
- The Center of Diagnosis and Treatment for Joint Disease, Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing 210008, China
| | - Jie Wang
- State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Sheng-Feng Lu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jing Wu
- State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Jian-Xin Li
- State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
44
|
Pan X, Liu B, Chen S, Ding H, Yao X, Cheng Y, Xu D, Yin Y, Dai X, Sun J, Xu G, Pan J, Xiao L, Xie L. Nr4a1 as a myogenic factor is upregulated in satellite cells/myoblast under proliferation and differentiation state. Biochem Biophys Res Commun 2019; 513:573-581. [DOI: 10.1016/j.bbrc.2019.04.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 04/03/2019] [Indexed: 01/07/2023]
|
45
|
Chen F, Zhou J, Li Y, Zhao Y, Yuan J, Cao Y, Wang L, Zhang Z, Zhang B, Wang CC, Cheung TH, Wu Z, Wong CCL, Sun H, Wang H. YY1 regulates skeletal muscle regeneration through controlling metabolic reprogramming of satellite cells. EMBO J 2019; 38:embj.201899727. [PMID: 30979776 DOI: 10.15252/embj.201899727] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 12/17/2018] [Accepted: 12/19/2018] [Indexed: 02/04/2023] Open
Abstract
Skeletal muscle satellite cells (SCs) are adult muscle stem cells responsible for muscle regeneration after acute or chronic injuries. The lineage progression of quiescent SC toward activation, proliferation, and differentiation during the regeneration is orchestrated by cascades of transcription factors (TFs). Here, we elucidate the function of TF Yin Yang1 (YY1) in muscle regeneration. Muscle-specific deletion of YY1 in embryonic muscle progenitors leads to severe deformity of diaphragm muscle formation, thus neonatal death. Inducible deletion of YY1 in SC almost completely blocks the acute damage-induced muscle repair and exacerbates the chronic injury-induced dystrophic phenotype. Examination of SC revealed that YY1 loss results in cell-autonomous defect in activation and proliferation. Mechanistic search revealed that YY1 binds and represses mitochondrial gene expression. Simultaneously, it also stabilizes Hif1α protein and activates Hif1α-mediated glycolytic genes to facilitate a metabolic reprogramming toward glycolysis which is needed for SC proliferation. Altogether, our findings have identified YY1 as a key regulator of SC metabolic reprogramming through its dual roles in modulating both mitochondrial and glycolytic pathways.
Collapse
Affiliation(s)
- Fengyuan Chen
- Department of Orthopedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Jiajian Zhou
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Yuying Li
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Yu Zhao
- Department of Orthopedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Jie Yuan
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Yang Cao
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Lijun Wang
- Department of Orthopedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Zongkang Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Baoting Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Chi Chiu Wang
- Department of Obstetrics and Gynecology, Li Ka Shing Institute of Health Sciences, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Tom H Cheung
- The State Key Lab in Molecular Neuroscience, Division of Life Science, Center for Stem Cell Research and Center for Systems Biology and Human Diseases, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Zhenguo Wu
- The State Key Lab in Molecular Neuroscience, Division of Life Science, Center for Stem Cell Research and Center for Systems Biology and Human Diseases, The Hong Kong University of Science and Technology, Hong Kong, China
| | | | - Hao Sun
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Huating Wang
- Department of Orthopedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
46
|
Hori S, Hiramuki Y, Nishimura D, Sato F, Sehara-Fujisawa A. PDH‐mediated metabolic flow is critical for skeletal muscle stem cell differentiation and myotube formation during regeneration in mice. FASEB J 2019; 33:8094-8109. [DOI: 10.1096/fj.201802479r] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- Shimpei Hori
- Department of Growth RegulationInstitute for Frontier Life and Medical SciencesKyoto University Kyoto Japan
| | - Yosuke Hiramuki
- Department of Growth RegulationInstitute for Frontier Life and Medical SciencesKyoto University Kyoto Japan
- Human Biology DivisionFred Hutchinson Cancer Research Center Seattle Washington USA
| | - Daigo Nishimura
- Department of Growth RegulationInstitute for Frontier Life and Medical SciencesKyoto University Kyoto Japan
| | - Fuminori Sato
- Department of Growth RegulationInstitute for Frontier Life and Medical SciencesKyoto University Kyoto Japan
| | - Atsuko Sehara-Fujisawa
- Department of Growth RegulationInstitute for Frontier Life and Medical SciencesKyoto University Kyoto Japan
| |
Collapse
|
47
|
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
|