1
|
Soro-Arnáiz I, Fitzgerald G, Cherkaoui S, Zhang J, Gilardoni P, Ghosh A, Bar-Nur O, Masschelein E, Maechler P, Zamboni N, Poms M, Cremonesi A, Garcia-Cañaveras JC, De Bock K, Morscher RJ. GLUD1 determines murine muscle stem cell fate by controlling mitochondrial glutamate levels. Dev Cell 2024; 59:2850-2865.e8. [PMID: 39121856 DOI: 10.1016/j.devcel.2024.07.015] [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: 04/04/2024] [Accepted: 07/16/2024] [Indexed: 08/12/2024]
Abstract
Muscle stem cells (MuSCs) enable muscle growth and regeneration after exercise or injury, but how metabolism controls their regenerative potential is poorly understood. We describe that primary metabolic changes can determine murine MuSC fate decisions. We found that glutamine anaplerosis into the tricarboxylic acid (TCA) cycle decreases during MuSC differentiation and coincides with decreased expression of the mitochondrial glutamate deaminase GLUD1. Deletion of Glud1 in proliferating MuSCs resulted in precocious differentiation and fusion, combined with loss of self-renewal in vitro and in vivo. Mechanistically, deleting Glud1 caused mitochondrial glutamate accumulation and inhibited the malate-aspartate shuttle (MAS). The resulting defect in transporting NADH-reducing equivalents into the mitochondria induced compartment-specific NAD+/NADH ratio shifts. MAS activity restoration or directly altering NAD+/NADH ratios normalized myogenesis. In conclusion, GLUD1 prevents deleterious mitochondrial glutamate accumulation and inactivation of the MAS in proliferating MuSCs. It thereby acts as a compartment-specific metabolic brake on MuSC differentiation.
Collapse
Affiliation(s)
- Inés Soro-Arnáiz
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zurich), Schwerzenbach, 8603 Zurich, Switzerland
| | - Gillian Fitzgerald
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zurich), Schwerzenbach, 8603 Zurich, Switzerland; Pediatric Cancer Metabolism Laboratory, Children's Research Center, University of Zurich, 8032 Zurich, Switzerland; Division of Oncology, University Children's Hospital Zurich and Children's Research Center, University of Zurich, 8032 Zurich, Switzerland
| | - Sarah Cherkaoui
- Pediatric Cancer Metabolism Laboratory, Children's Research Center, University of Zurich, 8032 Zurich, Switzerland; Division of Oncology, University Children's Hospital Zurich and Children's Research Center, University of Zurich, 8032 Zurich, Switzerland
| | - Jing Zhang
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zurich), Schwerzenbach, 8603 Zurich, Switzerland
| | - Paola Gilardoni
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zurich), Schwerzenbach, 8603 Zurich, Switzerland
| | - Adhideb Ghosh
- Laboratory of Regenerative and Movement Biology, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach, 8603 Zurich, Switzerland; Functional Genomics Center Zurich, ETH Zurich and University of Zurich, 8032 Zurich, Switzerland
| | - Ori Bar-Nur
- Laboratory of Regenerative and Movement Biology, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach, 8603 Zurich, Switzerland
| | - Evi Masschelein
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zurich), Schwerzenbach, 8603 Zurich, Switzerland
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, 1211 Geneva, Switzerland
| | - Nicola Zamboni
- Institute of Molecular Systems Biology, ETH Zurich, 8049 Zurich, Switzerland
| | - Martin Poms
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, University of Zurich, 8032 Zurich, Switzerland
| | - Alessio Cremonesi
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, University of Zurich, 8032 Zurich, Switzerland
| | | | - Katrien De Bock
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zurich), Schwerzenbach, 8603 Zurich, Switzerland.
| | - Raphael Johannes Morscher
- Pediatric Cancer Metabolism Laboratory, Children's Research Center, University of Zurich, 8032 Zurich, Switzerland; Division of Oncology, University Children's Hospital Zurich and Children's Research Center, University of Zurich, 8032 Zurich, Switzerland; Division of Human Genetics, Medical University Innsbruck, 6020 Innsbruck, Austria.
| |
Collapse
|
2
|
Assis V, Andrade RVD, de Sousa Neto IV, Barin FR, Ramos GV, Franco OL, Nobrega O, Mesquita-Ferrari RA, Malavazzi TCDS, Dos Santos Rosa T, de Luca Corrêa H, Petriz B, Durigan JLQ, de Cassia Marqueti R. Adaptive responses of skeletal muscle to calcaneal tendon partial injury in rats: insights into remodeling and plasticity. Mol Biol Rep 2024; 51:1078. [PMID: 39432127 DOI: 10.1007/s11033-024-09992-7] [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: 07/23/2024] [Accepted: 10/06/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND Skeletal muscle is a highly adaptive tissue, capable of responding to different physiological and functional demands, even in situations that may cause instability. OBJECTIVES To evaluate how partial calcaneal tendon (CT) injuries affect the remodeling and plasticity of the gastrocnemius muscle over time. METHODS AND RESULTS The study was carried out with Wistar rats randomly divided into five groups. The control group comprised animals not subjected to partial CT damage. The remaining four groups were subjected to partial CT damage and were further categorized based on the time of euthanasia: 3, 14, 28, and 55 days after injury. The gastrocnemius muscle was collected and used for gene expression analysis, zymography, flow cytometry, and morphology. The calcaneal tendon was analyzed only to verify the presence of the partial injury. RESULTS The impact of partial CT injury on the gastrocnemius homeostasis, particularly on gene expression, was more pronounced in the 3-day group compared to the other groups, especially the control group. Cytokine profile and morphologic alterations occurred in the 55 days group when compared to the other groups. CONCLUSIONS The data reported here suggest that partial injury can negatively affect intracellular signaling and degradation pathways, disturbing the muscular extracellular matrix regulatory mechanisms and communication with the tendon. However, skeletal muscle seems to mitigate these harmful effects in comparison with lesions that affect muscle and tendon.
Collapse
Affiliation(s)
- Victoria Assis
- Graduate Program in Rehabilitation Sciences, Laboratory of Molecular Analysis, Faculdade de Ceilândia, Universidade de Brasília, Campus Universitário, Centro Metropolitano 1, Conjunto A, Brasília, 72220-900, Brazil.
| | | | - Ivo Vieira de Sousa Neto
- School of Physical Education and Sport of Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - Gracielle Vieira Ramos
- IPE/HOME - Institute for Research and Teaching of the Orthopaedic Hospital and Specialty Medicine - HOME / FIFA Medical Centre of Excellence, Physiotherapy Department, Universidade Paulista, Brasília, Brazil
- Institute of Health Sciences, Universidade Paulista, São Paulo, Brazil
| | - Octávio Luiz Franco
- Graduate Program in Genomics Science and Biotechnology, Universidade Católica de Brasília, Brasília, Brazil
- Graduate Program of Sciences and Technology of Health, Universidade de Brasília, Brasília, Brazil
- S-Inova Biotech, Graduate Program in Biotechnology, Universidade Católica Dom Bosco, Campo Grande, Brazil
| | - Otavio Nobrega
- Faculty of Health Sciences, Universidade de Brasília, Brasília, Brazil
- Graduate Program of Medical Sciences, Universidade de Brasília, Brasília, Brazil
| | | | | | - Thiago Dos Santos Rosa
- Graduate Program in Genomics Science and Biotechnology, Universidade Católica de Brasília, Brasília, Brazil
- Graduate Program of Physical Education, Universidade Católica de Brasília, Brasilia, Brazil
| | - Hugo de Luca Corrêa
- Graduate Program of Physical Education, Universidade Católica de Brasília, Brasilia, Brazil
| | - Bernando Petriz
- Graduate Program in Genomics Science and Biotechnology, Universidade Católica de Brasília, Brasília, Brazil
| | - João Luiz Quaglioti Durigan
- Graduate Program in Rehabilitation Sciences, Laboratory of Molecular Analysis, Faculdade de Ceilândia, Universidade de Brasília, Campus Universitário, Centro Metropolitano 1, Conjunto A, Brasília, 72220-900, Brazil
| | - Rita de Cassia Marqueti
- Graduate Program in Rehabilitation Sciences, Laboratory of Molecular Analysis, Faculdade de Ceilândia, Universidade de Brasília, Campus Universitário, Centro Metropolitano 1, Conjunto A, Brasília, 72220-900, Brazil.
| |
Collapse
|
3
|
Xue S, Benvie AM, Blum JE, Kolba NJ, Cosgrove BD, Thalacker-Mercer A, Berry DC. Suppressing PDGFRβ Signaling Enhances Myocyte Fusion to Promote Skeletal Muscle Regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618247. [PMID: 39464006 PMCID: PMC11507758 DOI: 10.1101/2024.10.15.618247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Muscle cell fusion is critical for forming and maintaining multinucleated myotubes during skeletal muscle development and regeneration. However, the molecular mechanisms directing cell-cell fusion are not fully understood. Here, we identify platelet-derived growth factor receptor beta (PDGFRβ) signaling as a key modulator of myocyte fusion in adult muscle cells. Our findings demonstrate that genetic deletion of Pdgfrβ enhances muscle regeneration and increases myofiber size, whereas PDGFRβ activation impairs muscle repair. Inhibition of PDGFRβ activity promotes myonuclear accretion in both mouse and human myotubes, whereas PDGFRβ activation stalls myotube development by preventing cell spreading to limit fusion potential. Transcriptomics analysis show that PDGFRβ signaling cooperates with TGFβ signaling to direct myocyte size and fusion. Mechanistically, PDGFRβ signaling requires STAT1 activation, and blocking STAT1 phosphorylation enhances myofiber repair and size during regeneration. Collectively, PDGFRβ signaling acts as a regenerative checkpoint and represents a potential clinical target to rapidly boost skeletal muscle repair.
Collapse
Affiliation(s)
- Siwen Xue
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
| | - Abigail M Benvie
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
| | - Jamie E Blum
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
- Current address: Department of Chemical Engineering; Stanford University; Stanford, CA
| | - Nikolai J Kolba
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
| | | | - Anna Thalacker-Mercer
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
- Department of Cell, Development and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Daniel C Berry
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
- Corresponding author
| |
Collapse
|
4
|
Feng F, Luo K, Yuan X, Lan T, Wang S, Xu X, Lu Z. Aerobic Exercise Protects against Cardiotoxin-Induced Skeletal Muscle Injury in a DDAH1-Dependent Manner. Antioxidants (Basel) 2024; 13:1069. [PMID: 39334728 PMCID: PMC11428882 DOI: 10.3390/antiox13091069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/23/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
Dimethylarginine dimethylaminohydrolase 1 (DDAH1) is a critical enzyme that regulates nitric oxide (NO) signaling through the degradation of asymmetric dimethylarginine (ADMA). Previous studies have revealed a link between the beneficial effects of aerobic exercise and the upregulation of DDAH1 in bones and hearts. We previously reported that skeletal muscle DDAH1 plays a protective role in cardiotoxin (CTX)-induced skeletal muscle injury and regeneration. To determine the effects of aerobic exercise on CTX-induced skeletal muscle injury and the role of DDAH1 in this process, wild-type (WT) mice and skeletal muscle-specific Ddah1-knockout (Ddah1MKO) mice were subjected to swimming training for 8 weeks and then injected with CTX. In WT mice, swimming training for 8 weeks significantly promoted skeletal muscle regeneration and attenuated inflammation, oxidative stress, and apoptosis in the gastrocnemius (GA) muscle after CTX injection. These phenomena were associated with increases in the protein expression of PAX7, myogenin, MEF2A, eNOS, SOD2, and peroxiredoxin 5 and decreases in iNOS expression in GA muscles. Swimming training also decreased serum ADMA levels and increased serum nitrate/nitrite (NOx) levels and skeletal muscle DDAH1 expression. Interestingly, swimming training in Ddah1MKO mice had no obvious effect on CTX-induced skeletal muscle injury or regeneration and did not repress the CTX-induced inflammatory response, superoxide generation, or apoptosis. In summary, our data suggest that DDAH1 is important for the protective effect of aerobic exercise on skeletal muscle injury and regeneration.
Collapse
Affiliation(s)
- Fei Feng
- Sport and Health Science Department, Nanjing Sport Institute, Nanjing 210000, China
| | - Kai Luo
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinyi Yuan
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ting Lan
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Siyu Wang
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xin Xu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Zhongbing Lu
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
5
|
He Y, Lin T, Liang R, Xiang Q, Tang T, Ge N, Yue J. Interleukin 25 promotes muscle regeneration in sarcopenia by regulating macrophage-mediated Sonic Hedgehog signaling. Int Immunopharmacol 2024; 139:112662. [PMID: 39038385 DOI: 10.1016/j.intimp.2024.112662] [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: 04/09/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/24/2024]
Abstract
OBJECTIVE Sarcopenia manifests as a chronic, low-level inflammation along with multiple inflammatory cells infiltration. Interleukin (IL)-25 can regulate the function of macrophages. However, the specific role and mechanisms through which IL-25 functions in sarcopenia are still not fully understood and require further investigation. METHODS Aged mice were utilized as sarcopenia models and examined the expression of inflammatory factors. To investigate the effects of IL-25 on sarcopenia, the model mice received IL-25 treatment and underwent in vivo adoptive transfer of IL-25-induced macrophages. Meanwhile, RAW264.7 cells, bone marrow-derived macrophages, satellite cells and C2C12 cells were used in vitro. Shh insufficiency was induced through intramuscular administration of SHH-shRNA adenoviruses. Then, various assays including scratch wound, cell counting kit-8 and Transwell assays, as well as histological staining and molecular biological methods, were conducted. RESULTS Aged mice exhibited an accelerated decline in muscle strength and mass, along with an increased muscle lipid droplets and macrophage infiltration, and decreased IL-25 levels compared to the young group. IL-25 therapy and the transfer of IL-25-preconditioned macrophages could improve these conditions by promoting M2 macrophage polarization in vivo as well as in vitro. M2 macrophage conditioned medium enhanced satellite cell proliferation and migration, as well as the vitality, migration, and differentiation of C2C12 cells in vitro. Furthermore, IL-25 enhanced Shh expression in macrophages in vitro, and activated the Shh signaling pathway in muscle tissue of aged mice, which could be suppressed by either the inhibitor cyclopamine or Shh knockdown. Mechanistic studies showed that Shh insufficiency suppressed the activation of Akt/mTOR signaling pathway in muscle tissue of aged mice. CONCLUSION IL-25 promotes the secretion of Shh by M2 macrophages and activates the Shh/Akt/mTOR signaling pathway, which improves symptoms and function in sarcopenia mice. This suggests that IL-25 has potential as a therapeutic agent for treating sarcopenia.
Collapse
Affiliation(s)
- Yan He
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Geriatrics, The Second People's Hospital of Yibin, Yibin, Sichuan, China
| | - Taiping Lin
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rui Liang
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiao Xiang
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tianjiao Tang
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ning Ge
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Jirong Yue
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
6
|
Majchrzak K, Hentschel E, Hönzke K, Geithe C, von Maltzahn J. We need to talk-how muscle stem cells communicate. Front Cell Dev Biol 2024; 12:1378548. [PMID: 39050890 PMCID: PMC11266305 DOI: 10.3389/fcell.2024.1378548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/18/2024] [Indexed: 07/27/2024] Open
Abstract
Skeletal muscle is one of the tissues with the highest ability to regenerate, a finely controlled process which is critically depending on muscle stem cells. Muscle stem cell functionality depends on intrinsic signaling pathways and interaction with their immediate niche. Upon injury quiescent muscle stem cells get activated, proliferate and fuse to form new myofibers, a process involving the interaction of multiple cell types in regenerating skeletal muscle. Receptors in muscle stem cells receive the respective signals through direct cell-cell interaction, signaling via secreted factors or cell-matrix interactions thereby regulating responses of muscle stem cells to external stimuli. Here, we discuss how muscle stem cells interact with their immediate niche focusing on how this controls their quiescence, activation and self-renewal and how these processes are altered in age and disease.
Collapse
Affiliation(s)
- Karolina Majchrzak
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
| | - Erik Hentschel
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
| | - Katja Hönzke
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Christiane Geithe
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
| | - Julia von Maltzahn
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
- Faculty for Environment and Natural Sciences, Brandenburg University of Technology Cottbus—Senftenberg, Senftenberg, Germany
| |
Collapse
|
7
|
Luo J, Yang Z, Li X, Xiao C, Yuan H, Yang X, Zhou B, Zheng Y, Zhang J, Yang X. High Muscle Expression of IGF2BP1 Gene Promotes Proliferation and Differentiation of Chicken Primary Myoblasts: Results of Transcriptome Analysis. Animals (Basel) 2024; 14:2024. [PMID: 39061491 PMCID: PMC11274093 DOI: 10.3390/ani14142024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/06/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
Muscle development is a multifaceted process influenced by numerous genes and regulatory networks. Currently, the regulatory network of chicken muscle development remains incompletely elucidated, and its molecular genetic mechanisms require further investigation. The Longsheng-Feng chicken, one of the elite local breeds in Guangxi, serves as an excellent resource for the selection and breeding of high-quality broiler chickens. In this study, we conducted transcriptome sequencing of the pectoral muscles of Longsheng-Feng chickens and AA broiler chickens with different growth rates. Through comprehensive bioinformatics analysis, we identified differentially expressed genes that affect muscle growth and showed that IGF2BP1 is a key participant in chicken muscle development. Subsequently, we employed QRT-PCR, EdU staining, and flow cytometry to further investigate the role of IGF2BP1 in the proliferation and differentiation of chicken myogenic cells. We identified 1143 differentially expressed genes, among which IGF2BP1 is intimately related to the muscle development process and is highly expressed in muscle tissues. Overexpression of IGF2BP1 significantly promotes the proliferation and differentiation of chicken primary myoblasts, while knockdown of IGF2BP1 significantly inhibits these processes. In summary, these results provide valuable preliminary insights into the regulatory roles of IGF2BP1 in chicken growth and development.
Collapse
Affiliation(s)
- Jintang Luo
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (J.L.); (Z.Y.); (X.L.); (C.X.); (H.Y.); (X.Y.); (B.Z.); (Y.Z.); (J.Z.)
| | - Zhuliang Yang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (J.L.); (Z.Y.); (X.L.); (C.X.); (H.Y.); (X.Y.); (B.Z.); (Y.Z.); (J.Z.)
| | - Xianchao Li
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (J.L.); (Z.Y.); (X.L.); (C.X.); (H.Y.); (X.Y.); (B.Z.); (Y.Z.); (J.Z.)
| | - Cong Xiao
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (J.L.); (Z.Y.); (X.L.); (C.X.); (H.Y.); (X.Y.); (B.Z.); (Y.Z.); (J.Z.)
| | - Hong Yuan
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (J.L.); (Z.Y.); (X.L.); (C.X.); (H.Y.); (X.Y.); (B.Z.); (Y.Z.); (J.Z.)
| | - Xueqin Yang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (J.L.); (Z.Y.); (X.L.); (C.X.); (H.Y.); (X.Y.); (B.Z.); (Y.Z.); (J.Z.)
| | - Biyan Zhou
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (J.L.); (Z.Y.); (X.L.); (C.X.); (H.Y.); (X.Y.); (B.Z.); (Y.Z.); (J.Z.)
| | - Yan Zheng
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (J.L.); (Z.Y.); (X.L.); (C.X.); (H.Y.); (X.Y.); (B.Z.); (Y.Z.); (J.Z.)
| | - Jiayi Zhang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (J.L.); (Z.Y.); (X.L.); (C.X.); (H.Y.); (X.Y.); (B.Z.); (Y.Z.); (J.Z.)
| | - Xiurong Yang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (J.L.); (Z.Y.); (X.L.); (C.X.); (H.Y.); (X.Y.); (B.Z.); (Y.Z.); (J.Z.)
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Nanning 530004, China
| |
Collapse
|
8
|
Du Q, Dickinson A, Nakuleswaran P, Maghami S, Alagoda S, Hook AL, Ghaemmaghami AM. Targeting Macrophage Polarization for Reinstating Homeostasis following Tissue Damage. Int J Mol Sci 2024; 25:7278. [PMID: 39000385 PMCID: PMC11242417 DOI: 10.3390/ijms25137278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Tissue regeneration and remodeling involve many complex stages. Macrophages are critical in maintaining micro-environmental homeostasis by regulating inflammation and orchestrating wound healing. They display high plasticity in response to various stimuli, showing a spectrum of functional phenotypes that vary from M1 (pro-inflammatory) to M2 (anti-inflammatory) macrophages. While transient inflammation is an essential trigger for tissue healing following an injury, sustained inflammation (e.g., in foreign body response to implants, diabetes or inflammatory diseases) can hinder tissue healing and cause tissue damage. Modulating macrophage polarization has emerged as an effective strategy for enhancing immune-mediated tissue regeneration and promoting better integration of implantable materials in the host. This article provides an overview of macrophages' functional properties followed by discussing different strategies for modulating macrophage polarization. Advances in the use of synthetic and natural biomaterials to fabricate immune-modulatory materials are highlighted. This reveals that the development and clinical application of more effective immunomodulatory systems targeting macrophage polarization under pathological conditions will be driven by a detailed understanding of the factors that regulate macrophage polarization and biological function in order to optimize existing methods and generate novel strategies to control cell phenotype.
Collapse
Affiliation(s)
- Qiran Du
- Immuno-Bioengineering Group, School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Anna Dickinson
- Medical School, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (A.D.); (P.N.); (S.A.)
| | - Pruthvi Nakuleswaran
- Medical School, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (A.D.); (P.N.); (S.A.)
| | - Susan Maghami
- Hull York Medical School, University of York, York YO10 5DD, UK;
| | - Savindu Alagoda
- Medical School, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (A.D.); (P.N.); (S.A.)
| | - Andrew L. Hook
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Amir M. Ghaemmaghami
- Immuno-Bioengineering Group, School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK;
| |
Collapse
|
9
|
Feng L, Chen Z, Bian H. Skeletal muscle: molecular structure, myogenesis, biological functions, and diseases. MedComm (Beijing) 2024; 5:e649. [PMID: 38988494 PMCID: PMC11234433 DOI: 10.1002/mco2.649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 07/12/2024] Open
Abstract
Skeletal muscle is an important motor organ with multinucleated myofibers as its smallest cellular units. Myofibers are formed after undergoing cell differentiation, cell-cell fusion, myonuclei migration, and myofibril crosslinking among other processes and undergo morphological and functional changes or lesions after being stimulated by internal or external factors. The above processes are collectively referred to as myogenesis. After myofibers mature, the function and behavior of skeletal muscle are closely related to the voluntary movement of the body. In this review, we systematically and comprehensively discuss the physiological and pathological processes associated with skeletal muscles from five perspectives: molecule basis, myogenesis, biological function, adaptive changes, and myopathy. In the molecular structure and myogenesis sections, we gave a brief overview, focusing on skeletal muscle-specific fusogens and nuclei-related behaviors including cell-cell fusion and myonuclei localization. Subsequently, we discussed the three biological functions of skeletal muscle (muscle contraction, thermogenesis, and myokines secretion) and its response to stimulation (atrophy, hypertrophy, and regeneration), and finally settled on myopathy. In general, the integration of these contents provides a holistic perspective, which helps to further elucidate the structure, characteristics, and functions of skeletal muscle.
Collapse
Affiliation(s)
- Lan‐Ting Feng
- Department of Cell Biology & National Translational Science Center for Molecular MedicineNational Key Laboratory of New Drug Discovery and Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Zhi‐Nan Chen
- Department of Cell Biology & National Translational Science Center for Molecular MedicineNational Key Laboratory of New Drug Discovery and Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Huijie Bian
- Department of Cell Biology & National Translational Science Center for Molecular MedicineNational Key Laboratory of New Drug Discovery and Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| |
Collapse
|
10
|
Malavazzi TCDS, Andreo L, Martinelli A, Rodrigues MFSD, Horliana ACRT, Bussadori SK, Fernandes KPS, Nunes FD, Mesquita-Ferrari RA. Preventive and therapeutic vascular photobiomodulation decreases the inflammatory markers and enhances the muscle repair process in an animal model. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 256:112921. [PMID: 38714002 DOI: 10.1016/j.jphotobiol.2024.112921] [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: 03/14/2024] [Accepted: 04/22/2024] [Indexed: 05/09/2024]
Abstract
Photobiomodulation therapy (PBM) has shown positive effects when applied locally to modulate the inflammatory process and facilitate muscle repair. However, the available literature on the mechanisms of action of vascular photobiomodulation (VPBM), a non-invasive method of vascular irradiation, specifically in the context of local muscle repair, is limited. Thus, this study aimed to assess the impact of vascular photobiomodulation (VPBM) using a low-level laser (LLL) on the inflammatory response and the process of skeletal muscle repair whether administered prior to or following cryoinjury-induced acute muscle damage in the tibialis anterior (TA) muscles. Wistar rats (n = 85) were organized into the following experimental groups: (1) Control (n = 5); (2) Non-Injury + VPBM (n = 20); (3) Injured (n = 20); (4) Pre-VPBM + Injury (n = 20); (5) Injury + Post-VPBM (n = 20). VPBM was administered over the vein/artery at the base of the animals' tails (wavelength: 780 nm; power: 40 mW; application area: 0.04 cm2; energy density: 80 J/cm2). Euthanasia of the animals was carried out at 1, 2, 5, and 7 days after inducing the injuries. Tibialis anterior (TA) muscles were collected for both qualitative and quantitative histological analysis using H&E staining and for assessing protein expression of TNF-α, MCP-1, IL-1β, and IL-6 via ELISA. Blood samples were collected and analyzed using an automatic hematological analyzer and a leukocyte differential counter. Data were subjected to statistical analysis (ANOVA/Tukey). The results revealed that applying VPBM prior to injury led to an increase in circulating neutrophils (granulocytes) after 1 day and a subsequent increase in monocytes after 2 and 5 days, compared to the Non-Injury + VPBM and Injured groups. Notably, an increase in erythrocytes and hemoglobin concentration was observed in the Non-Injury + VPBM group on days 1 and 2 in comparison to the Injured group. In terms of histological aspects, only the Prior VPBM + Injured group exhibited a reduction in the number of inflammatory cells after 1, 5, and 7 days, along with an increase in blood vessels at 5 days. Both the Prior VPBM + Injured and Injured + VPBM after groups displayed a decrease in myonecrosis at 1, 2, and 7 days, an increase in newly-formed and immature fibers after 5 and 7 days, and neovascularization after 1, 2, and 7 days. Regarding protein expression, there was an increase in MCP-1 after 1 and 5 days, TNF-α, IL-6, and IL-1β after 1, 2, and 5 days in the Injured + VPBM after group when compared to the other experimental groups. The Prior VPBM + Injured group exhibited increased MCP-1 production after 2 days, in comparison to the Non-Injury + VPBM and Control groups. Notably, on day 7, the Injured group continued to show elevated MCP-1 protein expression when compared to the VPBM groups. In conclusion, VPBM effectively modulated hematological parameters, circulating leukocytes, the protein expression of the chemokine MCP-1, and the proinflammatory cytokines TNF-α and IL-1β, ultimately influencing the inflammatory process. This modulation resulted in a reduction of myonecrosis, restoration of tissue architecture, increased formation of newly and immature muscle fibers, and enhanced neovascularization, with more pronounced effects when VPBM was applied prior to the muscle injury.
Collapse
Affiliation(s)
- Tainá Caroline Dos Santos Malavazzi
- Postgraduate Program in Medicine-Biophotonics, Nove de Julho University (UNINOVE), 235/249 Vergueiro Street, Liberdade, São Paulo, SP 01504-001, Brazil
| | - Lucas Andreo
- Postgraduate Program in Medicine-Biophotonics, Nove de Julho University (UNINOVE), 235/249 Vergueiro Street, Liberdade, São Paulo, SP 01504-001, Brazil
| | - Andreia Martinelli
- Postgraduate Program in Rehabilitation Sciences, Nove de Julho University (UNINOVE), 235/249 Vergueiro Street, Liberdade, Sao Paulo, SP 01504-001, Brazil
| | | | | | - Sandra Kalil Bussadori
- Postgraduate Program in Medicine-Biophotonics, Nove de Julho University (UNINOVE), 235/249 Vergueiro Street, Liberdade, São Paulo, SP 01504-001, Brazil; Postgraduate Program in Rehabilitation Sciences, Nove de Julho University (UNINOVE), 235/249 Vergueiro Street, Liberdade, Sao Paulo, SP 01504-001, Brazil
| | - Kristianne Porta Santos Fernandes
- Postgraduate Program in Medicine-Biophotonics, Nove de Julho University (UNINOVE), 235/249 Vergueiro Street, Liberdade, São Paulo, SP 01504-001, Brazil
| | - Fabio Daumas Nunes
- Department of Stomatology, Discipline of Oral and Maxillofacial Pathology, School of Dentistry, University of São Paulo (FOUSP), Sao Paulo, SP 05508-000, Brazil
| | - Raquel Agnelli Mesquita-Ferrari
- Postgraduate Program in Medicine-Biophotonics, Nove de Julho University (UNINOVE), 235/249 Vergueiro Street, Liberdade, São Paulo, SP 01504-001, Brazil; Postgraduate Program in Rehabilitation Sciences, Nove de Julho University (UNINOVE), 235/249 Vergueiro Street, Liberdade, Sao Paulo, SP 01504-001, Brazil.
| |
Collapse
|
11
|
Cha M, Bak H, Bai SJ, Lee BH, Jang JH. Quadriceps recovery and pain relief in knee osteoarthritis rats by cog polydioxanone filament insertion. Regen Biomater 2024; 11:rbae077. [PMID: 38974667 PMCID: PMC11226885 DOI: 10.1093/rb/rbae077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/03/2024] [Accepted: 06/15/2024] [Indexed: 07/09/2024] Open
Abstract
Quadriceps muscles play a pivotal role in knee osteoarthritis (OA) progression and symptom manifestation, particularly pain. This research investigates the therapeutic effectiveness of muscle enhancement and support therapy (MEST), a recently developed device intended for intramuscular insertion of cog polydioxanone filaments, in quadriceps restoration to alleviate OA pain. Knee OA was induced in Sprague Dawley rats via monoiodoacetate injections. MEST or sham treatment was performed in OA or Naive rat quadriceps. Pain was assessed using paw withdrawal threshold and weight bearing. Quadriceps injury and recovery via MEST were evaluated using biomarkers, tissue morphology, muscle mass, contractile force and hindlimb torque. Satellite cell and macrophage activation, along with their activators, were also assessed. Data were compared at 1- and 3-weeks post-MEST treatment (M-W1 and M-W3). MEST treatment in OA rats caused muscle injury, indicated by elevated serum aspartate transferase and creatinine kinase levels, and local β-actin changes at M-W1. This injury triggered pro-inflammatory macrophage and satellite cell activation, accompanied by heightened interleukin-6 and insulin-like growth factor-1 levels. However, by M-W3, these processes gradually shifted toward inflammation resolution and muscle restoration. This was seen in anti-inflammatory macrophage phenotypes, sustained satellite cell activation and injury markers regressing to baseline. Quadriceps recovery in mass and strength from atrophy correlated with substantial OA pain reduction at M-W3. This study suggests that MEST-induced minor muscle injury triggers macrophage and satellite cell activation, leading to recovery of atrophied quadriceps and pain relief in OA rats.
Collapse
Affiliation(s)
- Myeounghoon Cha
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Heyji Bak
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Sun Joon Bai
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Bae Hwan Lee
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Brain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jun Ho Jang
- R&D Center, OV MEDI Co., Ltd, Gunpo 15847, Republic of Korea
| |
Collapse
|
12
|
Espino-Gonzalez E, Dalbram E, Mounier R, Gondin J, Farup J, Jessen N, Treebak JT. Impaired skeletal muscle regeneration in diabetes: From cellular and molecular mechanisms to novel treatments. Cell Metab 2024; 36:1204-1236. [PMID: 38490209 DOI: 10.1016/j.cmet.2024.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 01/10/2024] [Accepted: 02/22/2024] [Indexed: 03/17/2024]
Abstract
Diabetes represents a major public health concern with a considerable impact on human life and healthcare expenditures. It is now well established that diabetes is characterized by a severe skeletal muscle pathology that limits functional capacity and quality of life. Increasing evidence indicates that diabetes is also one of the most prevalent disorders characterized by impaired skeletal muscle regeneration, yet underlying mechanisms and therapeutic treatments remain poorly established. In this review, we describe the cellular and molecular alterations currently known to occur during skeletal muscle regeneration in people with diabetes and animal models of diabetes, including its associated comorbidities, e.g., obesity, hyperinsulinemia, and insulin resistance. We describe the role of myogenic and non-myogenic cell types on muscle regeneration in conditions with or without diabetes. Therapies for skeletal muscle regeneration and gaps in our knowledge are also discussed, while proposing future directions for the field.
Collapse
Affiliation(s)
- Ever Espino-Gonzalez
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Emilie Dalbram
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Rémi Mounier
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Julien Gondin
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Jean Farup
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Niels Jessen
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark; Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark.
| |
Collapse
|
13
|
Haase M, Comlekoglu T, Petrucciani A, Peirce SM, Blemker SS. Agent-based model demonstrates the impact of nonlinear, complex interactions between cytokinces on muscle regeneration. eLife 2024; 13:RP91924. [PMID: 38828844 PMCID: PMC11147512 DOI: 10.7554/elife.91924] [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: 06/05/2024] Open
Abstract
Muscle regeneration is a complex process due to dynamic and multiscale biochemical and cellular interactions, making it difficult to identify microenvironmental conditions that are beneficial to muscle recovery from injury using experimental approaches alone. To understand the degree to which individual cellular behaviors impact endogenous mechanisms of muscle recovery, we developed an agent-based model (ABM) using the Cellular-Potts framework to simulate the dynamic microenvironment of a cross-section of murine skeletal muscle tissue. We referenced more than 100 published studies to define over 100 parameters and rules that dictate the behavior of muscle fibers, satellite stem cells (SSCs), fibroblasts, neutrophils, macrophages, microvessels, and lymphatic vessels, as well as their interactions with each other and the microenvironment. We utilized parameter density estimation to calibrate the model to temporal biological datasets describing cross-sectional area (CSA) recovery, SSC, and fibroblast cell counts at multiple timepoints following injury. The calibrated model was validated by comparison of other model outputs (macrophage, neutrophil, and capillaries counts) to experimental observations. Predictions for eight model perturbations that varied cell or cytokine input conditions were compared to published experimental studies to validate model predictive capabilities. We used Latin hypercube sampling and partial rank correlation coefficient to identify in silico perturbations of cytokine diffusion coefficients and decay rates to enhance CSA recovery. This analysis suggests that combined alterations of specific cytokine decay and diffusion parameters result in greater fibroblast and SSC proliferation compared to individual perturbations with a 13% increase in CSA recovery compared to unaltered regeneration at 28 days. These results enable guided development of therapeutic strategies that similarly alter muscle physiology (i.e. converting extracellular matrix [ECM]-bound cytokines into freely diffusible forms as studied in cancer therapeutics or delivery of exogenous cytokines) during regeneration to enhance muscle recovery after injury.
Collapse
Affiliation(s)
- Megan Haase
- University of VirginiaCharlottesvilleUnited States
| | | | | | | | | |
Collapse
|
14
|
Rodríguez C, Timóteo-Ferreira F, Minchiotti G, Brunelli S, Guardiola O. Cellular interactions and microenvironment dynamics in skeletal muscle regeneration and disease. Front Cell Dev Biol 2024; 12:1385399. [PMID: 38840849 PMCID: PMC11150574 DOI: 10.3389/fcell.2024.1385399] [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: 02/12/2024] [Accepted: 04/30/2024] [Indexed: 06/07/2024] Open
Abstract
Skeletal muscle regeneration relies on the intricate interplay of various cell populations within the muscle niche-an environment crucial for regulating the behavior of muscle stem cells (MuSCs) and ensuring postnatal tissue maintenance and regeneration. This review delves into the dynamic interactions among key players of this process, including MuSCs, macrophages (MPs), fibro-adipogenic progenitors (FAPs), endothelial cells (ECs), and pericytes (PCs), each assuming pivotal roles in orchestrating homeostasis and regeneration. Dysfunctions in these interactions can lead not only to pathological conditions but also exacerbate muscular dystrophies. The exploration of cellular and molecular crosstalk among these populations in both physiological and dystrophic conditions provides insights into the multifaceted communication networks governing muscle regeneration. Furthermore, this review discusses emerging strategies to modulate the muscle-regenerating niche, presenting a comprehensive overview of current understanding and innovative approaches.
Collapse
Affiliation(s)
- Cristina Rodríguez
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics “A. Buzzati-Traverso”, CNR, Naples, Italy
| | | | - Gabriella Minchiotti
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics “A. Buzzati-Traverso”, CNR, Naples, Italy
| | - Silvia Brunelli
- School of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | - Ombretta Guardiola
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics “A. Buzzati-Traverso”, CNR, Naples, Italy
| |
Collapse
|
15
|
Mou K, Chan SMH, Vlahos R. Musculoskeletal crosstalk in chronic obstructive pulmonary disease and comorbidities: Emerging roles and therapeutic potentials. Pharmacol Ther 2024; 257:108635. [PMID: 38508342 DOI: 10.1016/j.pharmthera.2024.108635] [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: 11/06/2023] [Revised: 02/13/2024] [Accepted: 03/11/2024] [Indexed: 03/22/2024]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a multifaceted respiratory disorder characterized by progressive airflow limitation and systemic implications. It has become increasingly apparent that COPD exerts its influence far beyond the respiratory system, extending its impact to various organ systems. Among these, the musculoskeletal system emerges as a central player in both the pathogenesis and management of COPD and its associated comorbidities. Muscle dysfunction and osteoporosis are prevalent musculoskeletal disorders in COPD patients, leading to a substantial decline in exercise capacity and overall health. These manifestations are influenced by systemic inflammation, oxidative stress, and hormonal imbalances, all hallmarks of COPD. Recent research has uncovered an intricate interplay between COPD and musculoskeletal comorbidities, suggesting that muscle and bone tissues may cross-communicate through the release of signalling molecules, known as "myokines" and "osteokines". We explored this dynamic relationship, with a particular focus on the role of the immune system in mediating the cross-communication between muscle and bone in COPD. Moreover, we delved into existing and emerging therapeutic strategies for managing musculoskeletal disorders in COPD. It underscores the development of personalized treatment approaches that target both the respiratory and musculoskeletal aspects of COPD, offering the promise of improved well-being and quality of life for individuals grappling with this complex condition. This comprehensive review underscores the significance of recognizing the profound impact of COPD on the musculoskeletal system and its comorbidities. By unravelling the intricate connections between these systems and exploring innovative treatment avenues, we can aspire to enhance the overall care and outcomes for COPD patients, ultimately offering hope for improved health and well-being.
Collapse
Affiliation(s)
- Kevin Mou
- Centre for Respiratory Science and Health, School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Stanley M H Chan
- Centre for Respiratory Science and Health, School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Ross Vlahos
- Centre for Respiratory Science and Health, School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia.
| |
Collapse
|
16
|
Lessard L, Saugues A, Gondin J, Mounier R, Kneppers A. Measurement of Myonuclear Accretion In Vitro and In Vivo. Methods Mol Biol 2024. [PMID: 38647863 DOI: 10.1007/7651_2024_540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Adult skeletal muscle stem cells (MuSC) are the regenerative precursors of myofibers and also have an important role in myofiber growth, adaptation, and maintenance by fusing to the myofibers-a process referred to as "myonuclear accretion." Due to a focus on MuSC function during regeneration, myofibers remain a largely overlooked component of the MuSC niche influencing MuSC fate. Here, we describe a method to directly measure the rate of myonuclear accretion in vitro and in vivo using ethynyl-2'-deoxyuridine (EdU)-based tracing of MuSC progeny. This method supports the dissection of MuSC intrinsic and myofiber-derived factors influencing myonuclear accretion as an alternative fate of MuSCs supporting myofiber homeostasis and plasticity.
Collapse
Affiliation(s)
- Lola Lessard
- Institut NeuroMyoGène, Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR5261, INSERM U1315, Lyon, France
| | - Audrey Saugues
- Institut NeuroMyoGène, Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR5261, INSERM U1315, Lyon, France
| | - Julien Gondin
- Institut NeuroMyoGène, Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR5261, INSERM U1315, Lyon, France
| | - Rémi Mounier
- Institut NeuroMyoGène, Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR5261, INSERM U1315, Lyon, France
| | - Anita Kneppers
- Institut NeuroMyoGène, Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR5261, INSERM U1315, Lyon, France.
| |
Collapse
|
17
|
Nolt GL, Keeble AR, Wen Y, Strong AC, Thomas NT, Valentino TR, Brightwell CR, Murach KA, Patrizia S, Weinstabl H, Gollner A, McCarthy JJ, Fry CS, Franti M, Filareto A, Peterson CA, Dungan CM. Inhibition of p53-MDM2 binding reduces senescent cell abundance and improves the adaptive responses of skeletal muscle from aged mice. GeroScience 2024; 46:2153-2176. [PMID: 37872294 PMCID: PMC10828311 DOI: 10.1007/s11357-023-00976-2] [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: 07/24/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023] Open
Abstract
Skeletal muscle adaptation to external stimuli, such as regeneration following injury and hypertrophy in response to resistance exercise, are blunted with advanced age. The accumulation of senescent cells, along with defects in myogenic progenitor cell (MPC) proliferation, have been strongly linked as contributing factors to age-associated impairment in muscle adaptation. p53 plays an integral role in all these processes, as upregulation of p53 causes apoptosis in senescent cells and prevents mitotic catastrophe in MPCs from old mice. The goal of this study was to determine if a novel pharmaceutical agent (BI01), which functions by upregulating p53 through inhibition of binding to MDM2, the primary p53 regulatory protein, improves muscle regeneration and hypertrophy in old mice. BI01 effectively reduced the number of senescent cells in vitro but had no effect on MPC survival or proliferation at a comparable dose. Following repeated oral gavage with 2 mg/kg of BI01 (OS) or vehicle (OV), old mice (24 months) underwent unilateral BaCl2 injury in the tibialis anterior (TA) muscle, with PBS injections serving as controls. After 7 days, satellite cell number was higher in the TA of OS compared to OV mice, as was the expression of genes involved in ATP production. By 35 days, old mice treated with BI01 displayed reduced senescent cell burden, enhanced regeneration (higher muscle mass and fiber cross-sectional area) and restoration of muscle function relative to OV mice. To examine the impact of 2 mg/kg BI01 on muscle hypertrophy, the plantaris muscle was subjected to 28 days of mechanical overload (MOV) in OS and OV mice. In response to MOV, OS mice had larger plantaris muscles and muscle fibers than OV mice, particularly type 2b + x fibers, associated with reduced senescent cells. Together our data show that BI01 is an effective senolytic agent that may also augment muscle metabolism to enhance muscle regeneration and hypertrophy in old mice.
Collapse
Affiliation(s)
- Georgia L Nolt
- Department of Physiology, University of Kentucky, Lexington, KY, USA
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Alexander R Keeble
- Department of Physiology, University of Kentucky, Lexington, KY, USA
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Yuan Wen
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Physical Therapy, University of Kentucky, Lexington, KY, USA
| | - Aubrey C Strong
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Nicholas T Thomas
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, KY, USA
| | - Taylor R Valentino
- Department of Physiology, University of Kentucky, Lexington, KY, USA
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Camille R Brightwell
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, KY, USA
| | - Kevin A Murach
- Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Sini Patrizia
- Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT, 06877, USA
| | - Harald Weinstabl
- Boehringer Ingelheim RCV, Boehringer Ingelheim Pharmaceuticals Inc., Vienna, Austria
| | - Andreas Gollner
- Boehringer Ingelheim RCV, Boehringer Ingelheim Pharmaceuticals Inc., Vienna, Austria
| | - John J McCarthy
- Department of Physiology, University of Kentucky, Lexington, KY, USA
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Christopher S Fry
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, KY, USA
| | - Michael Franti
- Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT, 06877, USA
| | - Antonio Filareto
- Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT, 06877, USA.
| | - Charlotte A Peterson
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Physical Therapy, University of Kentucky, Lexington, KY, USA
| | - Cory M Dungan
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA.
- Department of Physical Therapy, University of Kentucky, Lexington, KY, USA.
- Department of Health, Human Performance, and Recreation, Baylor University, One Bear Place #97313, Waco, TX, 76706, USA.
| |
Collapse
|
18
|
Day KS, Rempel L, Rossi FMV, Theret M. Origins and functions of eosinophils in two non-mucosal tissues. Front Immunol 2024; 15:1368142. [PMID: 38585275 PMCID: PMC10995313 DOI: 10.3389/fimmu.2024.1368142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/26/2024] [Indexed: 04/09/2024] Open
Abstract
Eosinophils are a type of granulocyte named after the presence of their eosin-stained granules. Traditionally, eosinophils have been best known to play prominent roles in anti-parasitic responses and mediating allergic reactions. Knowledge of their behaviour has expanded with time, and they are now recognized to play integral parts in the homeostasis of gastrointestinal, respiratory, skeletal muscle, adipose, and connective tissue systems. As such, they are implicated in a myriad of pathologies, and have been the target of several medical therapies. This review focuses on the lifespan of eosinophils, from their origins in the bone marrow, to their tissue-resident role. In particular, we wish to highlight the functions of eosinophils in non-mucosal tissues with skeletal muscle and the adipose tissues as examples, and to discuss the current understanding of their participation in diseased states in these tissues.
Collapse
Affiliation(s)
- Katie S. Day
- Department of Medical Genetics, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Lucas Rempel
- Department of Medical Genetics, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Fabio M. V. Rossi
- Department of Medical Genetics, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Marine Theret
- Department of Medical Genetics, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
19
|
Haase M, Comlekoglu T, Petrucciani A, Peirce SM, Blemker SS. Agent-based model demonstrates the impact of nonlinear, complex interactions between cytokines on muscle regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.14.553247. [PMID: 37645968 PMCID: PMC10462020 DOI: 10.1101/2023.08.14.553247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Muscle regeneration is a complex process due to dynamic and multiscale biochemical and cellular interactions, making it difficult to identify microenvironmental conditions that are beneficial to muscle recovery from injury using experimental approaches alone. To understand the degree to which individual cellular behaviors impact endogenous mechanisms of muscle recovery, we developed an agent-based model (ABM) using the Cellular Potts framework to simulate the dynamic microenvironment of a cross-section of murine skeletal muscle tissue. We referenced more than 100 published studies to define over 100 parameters and rules that dictate the behavior of muscle fibers, satellite stem cells (SSC), fibroblasts, neutrophils, macrophages, microvessels, and lymphatic vessels, as well as their interactions with each other and the microenvironment. We utilized parameter density estimation to calibrate the model to temporal biological datasets describing cross-sectional area (CSA) recovery, SSC, and fibroblast cell counts at multiple time points following injury. The calibrated model was validated by comparison of other model outputs (macrophage, neutrophil, and capillaries counts) to experimental observations. Predictions for eight model perturbations that varied cell or cytokine input conditions were compared to published experimental studies to validate model predictive capabilities. We used Latin hypercube sampling and partial rank correlation coefficient to identify in silico perturbations of cytokine diffusion coefficients and decay rates to enhance CSA recovery. This analysis suggests that combined alterations of specific cytokine decay and diffusion parameters result in greater fibroblast and SSC proliferation compared to individual perturbations with a 13% increase in CSA recovery compared to unaltered regeneration at 28 days. These results enable guided development of therapeutic strategies that similarly alter muscle physiology (i.e. converting ECM-bound cytokines into freely diffusible forms as studied in cancer therapeutics or delivery of exogenous cytokines) during regeneration to enhance muscle recovery after injury.
Collapse
|
20
|
Geara P, Dilworth FJ. Epigenetic integration of signaling from the regenerative environment. Curr Top Dev Biol 2024; 158:341-374. [PMID: 38670712 DOI: 10.1016/bs.ctdb.2024.02.003] [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
Skeletal muscle has an extraordinary capacity to regenerate itself after injury due to the presence of tissue-resident muscle stem cells. While these muscle stem cells are the primary contributor to the regenerated myofibers, the process occurs in a regenerative microenvironment where multiple different cell types act in a coordinated manner to clear the damaged myofibers and restore tissue homeostasis. In this regenerative environment, immune cells play a well-characterized role in initiating repair by establishing an inflammatory state that permits the removal of dead cells and necrotic muscle tissue at the injury site. More recently, it has come to be appreciated that the immune cells also play a crucial role in communicating with the stem cells within the regenerative environment to help coordinate the timing of repair events through the secretion of cytokines, chemokines, and growth factors. Evidence also suggests that stem cells can help modulate the extent of the inflammatory response by signaling to the immune cells, demonstrating a cross-talk between the different cells in the regenerative environment. Here, we review the current knowledge on the innate immune response to sterile muscle injury and provide insight into the epigenetic mechanisms used by the cells in the regenerative niche to integrate the cellular cross-talk required for efficient muscle repair.
Collapse
Affiliation(s)
- Perla Geara
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI, United States
| | - F Jeffrey Dilworth
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI, United States.
| |
Collapse
|
21
|
Hsu WL, Lin YC, Lin MJ, Wang YW, Lee SJ. Macrophages enhance regeneration of lateral line neuromast derived from interneuromast cells through TGF-β in zebrafish. Dev Growth Differ 2024; 66:133-144. [PMID: 38281811 DOI: 10.1111/dgd.12911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 01/30/2024]
Abstract
Macrophages play a pivotal role in the response to injury, contributing significantly to the repair and regrowth of damaged tissues. The external lateral line system in aquatic organisms offers a practical model for studying regeneration, featuring interneuromast cells connecting sensory neuromasts. Under normal conditions, these cells remain dormant, but their transformation into neuromasts occurs when overcoming inhibitory signals from Schwann cells and posterior lateral line nerves. The mechanism enabling interneuromast cells to evade inhibition by Schwann cells remains unclear. Previous observations suggest that macrophages physically interact with neuromasts, nerves, and Schwann cells during regeneration. This interaction leads to the regeneration of neuromasts in a subset of zebrafish with ablated neuromasts. To explore whether macrophages achieve this effect through secreted cytokines, we conducted experiments involving tail amputation in zebrafish larvae and tested the impact of cytokine inhibitors on neuromast regeneration. Most injured larvae remarkably regenerated a neuromast within 4 days post-amputation. Intriguingly, removal of macrophages and inhibition of the anti-inflammatory cytokine transforming growth factor-beta (TGF-β) significantly delayed neuromast regeneration. Conversely, inhibition of the pro-inflammatory cytokines interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α) had minor effects on the regeneration process. This study provides insights into how macrophages activate interneuromast cells, elucidating the pathways underlying neuromast regeneration.
Collapse
Affiliation(s)
- Wei-Lin Hsu
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yu-Chi Lin
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Meng-Ju Lin
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yi-Wen Wang
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Shyh-Jye Lee
- Department of Life Science, National Taiwan University, Taipei, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
- Center for Biotechnology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
22
|
de Brito Sousa K, Dos Santos Malavazzi TC, Rodrigues MFSD, Silva T, Andreo L, Deana AM, Nunes FD, Bussadori SK, Mesquita-Ferrari RA, Fernandes KPS. Effects of amber LED on inflammatory and regulatory monocytes and lymphocytes. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 251:112848. [PMID: 38266361 DOI: 10.1016/j.jphotobiol.2024.112848] [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: 09/12/2023] [Revised: 12/28/2023] [Accepted: 01/16/2024] [Indexed: 01/26/2024]
Abstract
The primary objective of the present study was to assess the impact of amber LED photobiomodulation (PBM) on human monocytes and lymphocytes that were polarized into proinflammatory and regulatory/reparative phenotypes. Human leukocytes were polarized with LPS or LPS + IL-4 for 2 h and irradiated after 2 and 6 h with amber LED (590 nm). Cell absorbance spectrum and gene and protein expression of IL-1β, IL-6, IL-10, IL-17, TNF-α and IFNγ determined after 24 h. The results showed that irradiation did not significantly alter absorbance of non-polarized monocytes, whereas irradiated polarized monocytes presented reduction in absorbance in 625-850 nm region. Irradiated monocytes polarized with LPS + IL-4 presented reduction in absorbance in 600-725 nm region compared to non-irradiated group. Irradiated non-polarized lymphocytes presented absorbance peaks between 650 and 820 nm not seen in non-irradiated group. No difference was found in absorbance pattern of polarized lymphocytes after irradiation. Irradiation led to reduction in protein synthesis of IL-6 and TNFα in monocytes polarized to proinflammatory phenotype and increase in production of IL-17 in lymphocytes. Irradiation reduced production of IL-10 in monocytes and lymphocytes polarized to immunoregulatory phenotype. In conclusion, amber LED modulates light absorbance and expression of important cytokines in inflammatory/repair processes in monocytes and lymphocytes.
Collapse
Affiliation(s)
- Kaline de Brito Sousa
- Postgraduate Program in Medicine-Biophotonics, Nove de Julho University (UNINOVE), 235/249 Vergueiro Street, Liberdade, São Paulo, SP 01504-001, Brazil
| | - Tainá Caroline Dos Santos Malavazzi
- Postgraduate Program in Medicine-Biophotonics, Nove de Julho University (UNINOVE), 235/249 Vergueiro Street, Liberdade, São Paulo, SP 01504-001, Brazil
| | | | - Tamiris Silva
- Postgraduate Program in Medicine-Biophotonics, Nove de Julho University (UNINOVE), 235/249 Vergueiro Street, Liberdade, São Paulo, SP 01504-001, Brazil
| | - Lucas Andreo
- Postgraduate Program in Medicine-Biophotonics, Nove de Julho University (UNINOVE), 235/249 Vergueiro Street, Liberdade, São Paulo, SP 01504-001, Brazil
| | - Alessandro Melo Deana
- Postgraduate Program in Medicine-Biophotonics, Nove de Julho University (UNINOVE), 235/249 Vergueiro Street, Liberdade, São Paulo, SP 01504-001, Brazil
| | - Fabio Daumas Nunes
- Department of Stomatology, Discipline of Oral and Maxillofacial Pathology, School of Dentistry, University of São Paulo (FOUSP), São Paulo, SP 05508-000 Brazil
| | - Sandra Kalil Bussadori
- Postgraduate Program in Medicine-Biophotonics, Nove de Julho University (UNINOVE), 235/249 Vergueiro Street, Liberdade, São Paulo, SP 01504-001, Brazil; Postgraduate Program in Rehabilitation Sciences, Nove de Julho University (UNINOVE), 235/249 Vergueiro Street, Liberdade, Sao Paulo, SP 01504-001, Brazil
| | - Raquel Agnelli Mesquita-Ferrari
- Postgraduate Program in Medicine-Biophotonics, Nove de Julho University (UNINOVE), 235/249 Vergueiro Street, Liberdade, São Paulo, SP 01504-001, Brazil; Postgraduate Program in Rehabilitation Sciences, Nove de Julho University (UNINOVE), 235/249 Vergueiro Street, Liberdade, Sao Paulo, SP 01504-001, Brazil
| | - Kristianne Porta Santos Fernandes
- Postgraduate Program in Medicine-Biophotonics, Nove de Julho University (UNINOVE), 235/249 Vergueiro Street, Liberdade, São Paulo, SP 01504-001, Brazil.
| |
Collapse
|
23
|
Toita R, Kitamura M, Tsuchiya A, Kang J, Kasahara S. Releasable, Immune-Instructive, Bioinspired Multilayer Coating Resists Implant-Induced Fibrosis while Accelerating Tissue Repair. Adv Healthc Mater 2024; 13:e2302611. [PMID: 38095751 PMCID: PMC11468989 DOI: 10.1002/adhm.202302611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Indexed: 12/21/2023]
Abstract
Implantable biomaterials trigger foreign body reactions (FBRs), which reduces the functional life of medical devices and prevents effective tissue regeneration. Although existing therapeutic approaches can circumvent collagen-rich fibrotic encapsulation secondary to FBRs, they disrupt native tissue repair. Herein, a new surface engineering strategy in which an apoptotic-mimetic, immunomodulatory, phosphatidylserine liposome (PSL) is released from an implant coating to induce the formation of a macrophage phenotype that mitigates FBRs and improves tissue healing is described. PSL-multilayers constructed on implant surfaces via the layer-by-layer method release PSLs over a 1-month period. In rat muscles, poly(etheretherketone) (PEEK), a nondegradable polymer implant model, induces FBRs with dense fibrotic scarring under an aberrant cellular profile that recruits high levels of inflammatory infiltrates, foreign body giant cells (FBGCs), scar-forming myofibroblasts, and inflammatory M1-like macrophages but negligible amounts of anti-inflammatory M2-like phenotypes. However, the PSL-multilayer coating markedly diminishes these detrimental signatures by shifting the macrophage phenotype. Unlike other therapeutics, PSL-multilayered coatings also stimulate muscle regeneration. This study demonstrates that PSL-multilayered coatings are effective in eliminating FBRs and promoting regeneration, hence offering potent and broad applications for implantable biomaterials.
Collapse
Affiliation(s)
- Riki Toita
- Biomedical Research InstituteNational Institute of Advanced Industrial Science and Technology (AIST)1‐8‐31 MidorigaokaIkedaOsaka563‐8577Japan
- AIST‐Osaka University Advanced Photonics and Biosensing Open Innovation LaboratoryAIST2‐1 YamadaokaSuitaOsaka565‐0871Japan
| | - Masahiro Kitamura
- Niterra Co., Ltd.2808 IwasakiKomakiAichi485–8510Japan
- NGK Spark Plug‐AIST Healthcare Materials Cooperative Research Laboratory2266–98 AnagahoraShimoshidami, Moriyama‐kuNagoyaAichi463–8560Japan
| | - Akira Tsuchiya
- Department of BiomaterialsFaculty of Dental ScienceKyushu University3‐1‐1 MaidashiHigashi‐kuFukuoka812–8582Japan
| | - Jeong‐Hun Kang
- Division of Biopharmaceutics and PharmacokineticsNational Cerebral and Cardiovascular Center Research Institute6‐1 Shinmachi, KishibeSuitaOsaka564–8565Japan
| | | |
Collapse
|
24
|
Lindquist KA, Shein SA, Hovhannisyan AH, Mecklenburg J, Zou Y, Lai Z, Tumanov AV, Akopian AN. Associations of tissue damage induced inflammatory plasticity in masseter muscle with the resolution of chronic myalgia. Sci Rep 2023; 13:22057. [PMID: 38086903 PMCID: PMC10716154 DOI: 10.1038/s41598-023-49280-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 12/06/2023] [Indexed: 12/18/2023] Open
Abstract
Gene plasticity during myogenous temporomandibular disorder (TMDM) development is largely unknown. TMDM could be modeled by intramuscular inflammation or tissue damage. To model inflammation induced TMDM we injected complete Freund's adjuvant (CFA) into masseter muscle (MM). To model tissue damage induced TMDM we injected extracellular matrix degrading collagenase type 2 (Col). CFA and Col produced distinct myalgia development trajectories. We performed bulk RNA-seq of MM to generate gene plasticity time course. CFA initiated TMDM (1d post-injection) was mainly linked to chemo-tacticity of monocytes and neutrophils. At CFA-induced hypersensitivity post-resolution (5d post-injection), tissue repair processes were pronounced, while inflammation was absent. Col (0.2U) produced acute hypersensitivity linked to tissue repair without inflammatory processes. Col (10U) generated prolonged hypersensitivity with inflammatory processes dominating initiation phase (1d). Pre-resolution phase (6d) was accompanied with acceleration of expressions for tissue repair and pro-inflammatory genes. Flow cytometry showed that immune processes in MM was associated with accumulations of macrophages, natural killer, dendritic and T-cells, further confirming our RNA-seq findings. Altogether, CFA and Col treatments induced different immune processes in MM. Importantly, TMDM resolution was preceded with muscle cell and extracellular matrix repairs, an elevation in immune system gene expressions and distinct immune cell accumulations in MM.
Collapse
Affiliation(s)
- Karen A Lindquist
- Integrated Biomedical Sciences (IBMS) Program, The School of Medicine, The University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Sergey A Shein
- Departments of Microbiology, Immunology & Molecular Genetics, The School of Medicine, UTHSCSA, San Antonio, TX, 78229, USA
| | - Anahit H Hovhannisyan
- Departments of Endodontics, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Jennifer Mecklenburg
- Departments of Endodontics, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Yi Zou
- Departments of Molecular Medicine, The School of Medicine, UTHSCSA, San Antonio, TX, USA
| | - Zhao Lai
- Departments of Molecular Medicine, The School of Medicine, UTHSCSA, San Antonio, TX, USA
- Greehey Children's Cancer Research Institute, UTHSCSA, San Antonio, TX, 78229, USA
| | - Alexei V Tumanov
- Integrated Biomedical Sciences (IBMS) Program, The School of Medicine, The University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA.
- Departments of Microbiology, Immunology & Molecular Genetics, The School of Medicine, UTHSCSA, San Antonio, TX, 78229, USA.
| | - Armen N Akopian
- Integrated Biomedical Sciences (IBMS) Program, The School of Medicine, The University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA.
- Departments of Endodontics, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA.
| |
Collapse
|
25
|
Cocchiararo I, Cattaneo O, Rajendran J, Chabry F, Cornut M, Soldati H, Bigot A, Mamchaoui K, Gibertini S, Bouche A, Ham DJ, Laumonier T, Prola A, Castets P. Identification of a muscle-specific isoform of VMA21 as a potent actor in X-linked myopathy with excessive autophagy pathogenesis. Hum Mol Genet 2023; 32:3374-3389. [PMID: 37756622 PMCID: PMC10695681 DOI: 10.1093/hmg/ddad164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/23/2023] [Accepted: 09/21/2023] [Indexed: 09/29/2023] Open
Abstract
Defective lysosomal acidification is responsible for a large range of multi-systemic disorders associated with impaired autophagy. Diseases caused by mutations in the VMA21 gene stand as exceptions, specifically affecting skeletal muscle (X-linked Myopathy with Excessive Autophagy, XMEA) or liver (Congenital Disorder of Glycosylation). VMA21 chaperones vacuolar (v-) ATPase assembly, which is ubiquitously required for proper lysosomal acidification. The reason VMA21 deficiencies affect specific, but divergent tissues remains unknown. Here, we show that VMA21 encodes a yet-unreported long protein isoform, in addition to the previously described short isoform, which we name VMA21-120 and VMA21-101, respectively. In contrast to the ubiquitous pattern of VMA21-101, VMA21-120 was predominantly expressed in skeletal muscle, and rapidly up-regulated upon differentiation of mouse and human muscle precursors. Accordingly, VMA21-120 accumulated during development, regeneration and denervation of mouse skeletal muscle. In contrast, neither induction nor blockade of autophagy, in vitro and in vivo, strongly affected VMA21 isoform expression. Interestingly, VMA21-101 and VMA21-120 both localized to the sarcoplasmic reticulum of muscle cells, and interacted with the v-ATPase. While VMA21 deficiency impairs autophagy, VMA21-101 or VMA21-120 overexpression had limited impact on autophagic flux in muscle cells. Importantly, XMEA-associated mutations lead to both VMA21-101 deficiency and loss of VMA21-120 expression. These results provide important insights into the clinical diversity of VMA21-related diseases and uncover a muscle-specific VMA21 isoform that potently contributes to XMEA pathogenesis.
Collapse
Affiliation(s)
- Ilaria Cocchiararo
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1 rue Michel Servet, CH-1211 Geneva, Switzerland
| | - Olivia Cattaneo
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1 rue Michel Servet, CH-1211 Geneva, Switzerland
| | - Jayasimman Rajendran
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1 rue Michel Servet, CH-1211 Geneva, Switzerland
| | - Florent Chabry
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1 rue Michel Servet, CH-1211 Geneva, Switzerland
| | - Mélanie Cornut
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1 rue Michel Servet, CH-1211 Geneva, Switzerland
| | - Hadrien Soldati
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1 rue Michel Servet, CH-1211 Geneva, Switzerland
| | - Anne Bigot
- Centre de Recherche en Myologie, Inserm, Institut de Myologie, Sorbonne Université, 47 Bd de l'Hôpital, 75013 Paris, France
| | - Kamel Mamchaoui
- Centre de Recherche en Myologie, Inserm, Institut de Myologie, Sorbonne Université, 47 Bd de l'Hôpital, 75013 Paris, France
| | - Sara Gibertini
- Neuromuscular Diseases and Neuroimmunology Unit, Muscle Cell Biology Lab, Fondazione IRCCS Istituto Neurologico “C. Besta”, Via Amadeo 42, 20133 Milano, Italy
| | - Axelle Bouche
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1 rue Michel Servet, CH-1211 Geneva, Switzerland
- Department of Orthopaedic Surgery, Geneva University Hospitals and Faculty of Medicine, University Medical Center, 1 rue Michel Servet, 1211, Geneva, Switzerland
| | - Daniel J Ham
- Biozentrum, University of Basel, Spitalstrasse 41, 4056 Basel, Switzerland
| | - Thomas Laumonier
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1 rue Michel Servet, CH-1211 Geneva, Switzerland
- Department of Orthopaedic Surgery, Geneva University Hospitals and Faculty of Medicine, University Medical Center, 1 rue Michel Servet, 1211, Geneva, Switzerland
| | - Alexandre Prola
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1 rue Michel Servet, CH-1211 Geneva, Switzerland
| | - Perrine Castets
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1 rue Michel Servet, CH-1211 Geneva, Switzerland
| |
Collapse
|
26
|
Yaghi OK, Hanna BS, Langston PK, Michelson DA, Jayewickreme T, Marin-Rodero M, Benoist C, Mathis D. A discrete 'early-responder' stromal-cell subtype orchestrates immunocyte recruitment to injured tissue. Nat Immunol 2023; 24:2053-2067. [PMID: 37932455 PMCID: PMC10792729 DOI: 10.1038/s41590-023-01669-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/05/2023] [Indexed: 11/08/2023]
Abstract
Following acute injury, stromal cells promote tissue regeneration by a diversity of mechanisms. Time-resolved single-cell RNA sequencing of muscle mesenchymal stromal cells (MmSCs) responding to acute injury identified an 'early-responder' subtype that spiked on day 1 and expressed a notable array of transcripts encoding immunomodulators. IL-1β, TNF-α and oncostatin M each strongly and rapidly induced MmSCs transcribing this immunomodulatory program. Macrophages amplified the program but were not strictly required for its induction. Transfer of the inflammatory MmSC subtype, tagged with a unique surface marker, into healthy hindlimb muscle induced inflammation primarily driven by neutrophils and macrophages. Among the abundant inflammatory transcripts produced by this subtype, Cxcl5 was stroma-specific and highly upregulated with injury. Depletion of this chemokine early after injury revealed a substantial impact on recruitment of neutrophils, a prolongation of inflammation to later times and an effect on tissue regeneration. Mesenchymal stromal cell subtypes expressing a comparable inflammatory program were found in a mouse model of muscular dystrophy and in several other tissues and pathologies in both mice and humans. These 'early-responder' mesenchymal stromal cells, already in place, permit rapid and coordinated mobilization and amplification of critical cell collaborators in response to injury.
Collapse
Affiliation(s)
- Omar K Yaghi
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Bola S Hanna
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - P Kent Langston
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Daniel A Michelson
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Teshika Jayewickreme
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Miguel Marin-Rodero
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Christophe Benoist
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA, USA.
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
27
|
Tarban N, Papp AB, Deák D, Szentesi P, Halász H, Patsalos A, Csernoch L, Sarang Z, Szondy Z. Loss of adenosine A3 receptors accelerates skeletal muscle regeneration in mice following cardiotoxin-induced injury. Cell Death Dis 2023; 14:706. [PMID: 37898628 PMCID: PMC10613231 DOI: 10.1038/s41419-023-06228-7] [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: 07/05/2023] [Revised: 10/13/2023] [Accepted: 10/18/2023] [Indexed: 10/30/2023]
Abstract
Skeletal muscle regeneration is a complex process orchestrated by multiple interacting steps. An increasing number of reports indicate that inflammatory responses play a central role in linking initial muscle injury responses to timely muscle regeneration following injury. The nucleoside adenosine has been known for a long time as an endogenously produced anti-inflammatory molecule that is generated in high amounts during tissue injury. It mediates its physiological effects via four types of adenosine receptors. From these, adenosine A3 receptors (A3Rs) are not expressed by the skeletal muscle but are present on the surface of various inflammatory cells. In the present paper, the effect of the loss of A3Rs was investigated on the regeneration of the tibialis anterior (TA) muscle in mice following cardiotoxin-induced injury. Here we report that regeneration of the skeletal muscle from A3R-/- mice is characterized by a stronger initial inflammatory response resulting in a larger number of transmigrating inflammatory cells to the injury site, faster clearance of cell debris, enhanced proliferation and faster differentiation of the satellite cells (the muscle stem cells), and increased fusion of the generated myoblasts. This leads to accelerated skeletal muscle tissue repair and the formation of larger myofibers. Though the infiltrating immune cells expressed A3Rs and showed an increased inflammatory profile in the injured A3R-/- muscles, bone marrow transplantation experiments revealed that the increased response of the tissue-resident cells to tissue injury is responsible for the observed phenomenon. Altogether our data indicate that A3Rs are negative regulators of injury-related regenerative inflammation and consequently also that of the muscle fiber growth in the TA muscle. Thus, inhibiting A3Rs might have a therapeutic value during skeletal muscle regeneration following injury.
Collapse
Affiliation(s)
- Nastaran Tarban
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Debrecen, Hungary
| | - Albert Bálint Papp
- Doctoral School of Dental Sciences, University of Debrecen, Debrecen, Hungary
| | - Dávid Deák
- Laboratory Animal Facility, Life Science Building, University of Debrecen, Debrecen, Hungary
| | - Péter Szentesi
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Hajnalka Halász
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Debrecen, Hungary
| | - Andreas Patsalos
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St, Petersburg, FL, USA
| | - László Csernoch
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Sarang
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsuzsa Szondy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
- Division of Dental Biochemistry, Department of Basic Medical Sciences, Faculty of Dentistry, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
28
|
Wang X, Zhou L. The multifaceted role of macrophages in homeostatic and injured skeletal muscle. Front Immunol 2023; 14:1274816. [PMID: 37954602 PMCID: PMC10634307 DOI: 10.3389/fimmu.2023.1274816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/13/2023] [Indexed: 11/14/2023] Open
Abstract
Skeletal muscle is essential for body physical activity, energy metabolism, and temperature maintenance. It has excellent capabilities to maintain homeostasis and to regenerate after injury, which indispensably relies on muscle stem cells, satellite cells (MuSCs). The quiescence, activation, and differentiation of MuSCs are tightly regulated in homeostatic and regenerating muscles. Among the important regulators are intramuscular macrophages, which are functionally heterogeneous with different subtypes present in a spatiotemporal manner to regulate the balance of different MuSC statuses. During chronic injury and aging, intramuscular macrophages often undergo aberrant activation, which in turn disrupts muscle homeostasis and regenerative repair. Growing evidence suggests that the aberrant activation is mainly triggered by altered muscle microenvironment. The trained immunity that affects myeloid progenitors during hematopoiesis may also contribute. Aged immune system may contribute, in part, to the aging-related sarcopenia and compromised skeletal muscle injury repair. As macrophages are actively involved in the progression of many muscle diseases, manipulating their functional activation has become a promising therapeutic approach, which requires comprehensive knowledge of the cellular and molecular mechanisms underlying the diverse activation. To this end, we discuss here the current knowledge of multifaceted role of macrophages in skeletal muscle homeostasis, injury, and repair.
Collapse
Affiliation(s)
- Xingyu Wang
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | | |
Collapse
|
29
|
Edouard P, Reurink G, Mackey AL, Lieber RL, Pizzari T, Järvinen TAH, Gronwald T, Hollander K. Traumatic muscle injury. Nat Rev Dis Primers 2023; 9:56. [PMID: 37857686 DOI: 10.1038/s41572-023-00469-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/22/2023] [Indexed: 10/21/2023]
Abstract
Traumatic muscle injury represents a collection of skeletal muscle pathologies caused by trauma to the muscle tissue and is defined as damage to the muscle tissue that can result in a functional deficit. Traumatic muscle injury can affect people across the lifespan and can result from high stresses and strains to skeletal muscle tissue, often due to muscle activation while the muscle is lengthening, resulting in indirect and non-contact muscle injuries (strains or ruptures), or from external impact, resulting in direct muscle injuries (contusion or laceration). At a microscopic level, muscle fibres can repair focal damage but must be completely regenerated after full myofibre necrosis. The diagnosis of muscle injury is based on patient history and physical examination. Imaging may be indicated to eliminate differential diagnoses. The management of muscle injury has changed within the past 5 years from initial rest, immobilization and (over)protection to early activation and progressive loading using an active approach. One challenge of muscle injury management is that numerous medical treatment options, such as medications and injections, are often used or proposed to try to accelerate muscle recovery despite very limited efficacy evidence. Another challenge is the prevention of muscle injury owing to the multifactorial and complex nature of this injury.
Collapse
Affiliation(s)
- Pascal Edouard
- Université Jean Monnet, Lyon 1, Université Savoie Mont-Blanc, Inter-university Laboratory of Human Movement Biology, Saint-Etienne, France.
- Department of Clinical and Exercise Physiology, Sports Medicine Unit, University Hospital of Saint-Etienne, Faculty of Medicine, Saint-Etienne, France.
| | - Gustaaf Reurink
- Department of Orthopedic Surgery and Sports Medicine, Academic Medical Center, University of Amsterdam, Amsterdam Movement Sciences, Amsterdam, Netherlands
- Academic Center for Evidence-based Sports Medicine (ACES), Academic Medical Center, Amsterdam, Netherlands
- The Sports Physicians Group, Onze Lieve Vrouwe Gasthuis, Amsterdam, Netherlands
| | - Abigail L Mackey
- Institute of Sports Medicine Copenhagen, Department of Orthopaedic Surgery M, Bispebjerg Hospital, Copenhagen, Denmark
- Center for Healthy Aging, Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Richard L Lieber
- Shirley Ryan AbilityLab, Chicago, IL, USA
- Departments of Physical Medicine and Rehabilitation and Biomedical Engineering, Northwestern University, Chicago, IL, USA
- Hines VA Medical Center, Maywood, IL, USA
| | - Tania Pizzari
- La Trobe Sport and Exercise Medicine Research Centre, La Trobe University, Melbourne, Victoria, Australia
| | - Tero A H Järvinen
- Tampere University and Tampere University Hospital, Tampere, Finland
| | - Thomas Gronwald
- Institute of Interdisciplinary Exercise Science and Sports Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Karsten Hollander
- Institute of Interdisciplinary Exercise Science and Sports Medicine, MSH Medical School Hamburg, Hamburg, Germany
| |
Collapse
|
30
|
Gregg SR, Barshick MR, Johnson SE. Intravenous Injection of Sodium Hyaluronate Diminishes Basal Inflammatory Gene Expression in Equine Skeletal Muscle. Animals (Basel) 2023; 13:3030. [PMID: 37835636 PMCID: PMC10571686 DOI: 10.3390/ani13193030] [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: 08/18/2023] [Revised: 09/08/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
Following strenuous exercise, skeletal muscle experiences an acute inflammatory state that initiates the repair process. Systemic hyaluronic acid (HA) is injected to horses routinely as a joint anti-inflammatory. To gain insight into the effects of HA on skeletal muscle, adult Thoroughbred geldings (n = 6) were injected with a commercial HA product weekly for 3 weeks prior to performing a submaximal exercise test. Gluteal muscle (GM) biopsies were obtained before and 1 h after exercise for gene expression analysis and HA localization. The results from RNA sequencing demonstrate differences in gene expression between non-injected controls (CON; n = 6) and HA horses. Prior to exercise, HA horses contained fewer (p < 0.05) transcripts associated with leukocyte activity and cytokine production than CON. The performance of exercise resulted in the upregulation (p < 0.05) of several cytokine genes and their signaling intermediates, indicating that HA does not suppress the normal inflammatory response to exercise. The transcript abundance for marker genes of neutrophils (NCF2) and macrophages (CD163) was greater (p < 0.05) post-exercise and was unaffected by HA injection. The anti-inflammatory effects of HA on muscle are indirect as no differences (p > 0.05) in the relative amount of the macromolecule was observed between the CON and HA fiber extracellular matrix (ECM). However, exercise tended (p = 0.10) to cause an increase in ECM size suggestive of muscle damage and remodeling. The finding was supported by the increased (p < 0.05) expression of CTGF, TGFβ1, MMP9, TIMP4 and Col4A1. Collectively, the results validate HA as an anti-inflammatory aid that does not disrupt the normal post-exercise muscle repair process.
Collapse
Affiliation(s)
| | | | - Sally E. Johnson
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA; (S.R.G.); (M.R.B.)
| |
Collapse
|
31
|
Feng F, Cui B, Fang L, Lan T, Luo K, Xu X, Lu Z. DDAH1 Protects against Cardiotoxin-Induced Muscle Injury and Regeneration. Antioxidants (Basel) 2023; 12:1754. [PMID: 37760057 PMCID: PMC10525962 DOI: 10.3390/antiox12091754] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Nitric oxide (NO) is an important biological signaling molecule affecting muscle regeneration. The activity of NO synthase (NOS) is regulated by dimethylarginine dimethylaminohydrolase 1 (DDAH1) through degradation of the endogenous NOS inhibitor asymmetric dimethylarginine (ADMA). To investigate the role of DDAH1 in muscle injury and regeneration, muscle-specific Ddah1-knockout mice (Ddah1MKO) and their littermates (Ddah1f/f) were used to examine the progress of cardiotoxin (CTX)-induced muscle injury and subsequent muscle regeneration. After CTX injection, Ddah1MKO mice developed more severe muscle injury than Ddah1f/f mice. Muscle regeneration was also delayed in Ddah1MKO mice on Day 5 after CTX injection. These phenomena were associated with higher serum ADMA and LDH levels as well as a great induction of inflammatory response, oxidative stress and cell apoptosis in the gastrocnemius (GA) muscle of Ddah1MKO mice. In the GA muscle of CTX-treated mice, Ddah1 deficiency decreased the protein expression of M-cadherin, myogenin, Bcl-2, peroxiredoxin 3 (PRDX3) and PRDX5, and increased the protein expression of MyoD, TNFα, Il-6, iNOS and Bax. In summary, our data suggest that DDAH1 exerts a protective role in muscle injury and regeneration.
Collapse
Affiliation(s)
- Fei Feng
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China;
| | - Bingqing Cui
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China; (B.C.); (K.L.)
| | - Li Fang
- Department of Endocrinology, Dongtai Renmin Hospital, Dongtai 224233, China;
| | - Ting Lan
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China; (B.C.); (K.L.)
| | - Kai Luo
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China; (B.C.); (K.L.)
| | - Xin Xu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China;
| | - Zhongbing Lu
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China; (B.C.); (K.L.)
| |
Collapse
|
32
|
Bernard C, Jomard C, Chazaud B, Gondin J. Kinetics of skeletal muscle regeneration after mild and severe muscle damage induced by electrically-evoked lengthening contractions. FASEB J 2023; 37:e23107. [PMID: 37534948 DOI: 10.1096/fj.202201708rr] [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: 10/19/2022] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 08/04/2023]
Abstract
Post-injury skeletal muscle regeneration requires interactions between myogenic and non-myogenic cells. Our knowledge on the regeneration process is mainly based on models using toxic, chemical, or physical (e.g., based on either muscle freezing or crushing) injury. Strikingly, the time course and magnitude of changes in the number of cells involved in muscle regeneration have been poorly described in relation to mild and severe muscle damage induced by electrically-evoked lengthening contractions. We investigated for the first time the kinetics and magnitude of changes in mononuclear cells in relation to the extent of muscle damage. Mild and severe injury were induced in vivo in the mouse gastrocnemius muscle by 1 and 30 electrically-evoked lengthening contractions, respectively. Several days after muscle damage, functional analysis of maximal torque production and histological investigations were performed to assess the related cellular changes. Torque recovery was faster after mild injury than after severe muscle damage. More necrotic and regenerating myofibers were observed after severe muscle damage as compared with mild injury, illustrating an association between functional and histological alterations. The kinetics of changes in muscle stem cells (total, proliferating, and differentiating), endothelial cells, fibro-adipogenic progenitors (FAPs), and macrophages in the regenerating muscle was similar in mild and severe models. However, the magnitude of changes in the number of differentiating muscle stem cells, hematopoietic cells, among which macrophages, and FAPs was higher in severe muscle damage. Collectively, our results show that the amount of myogenic and non-myogenic cells varies according to the extent of skeletal muscle injury to ensure efficient skeletal muscle regeneration while the kinetics of changes is independent of muscle tissue alterations. The possibility to experimentally modulate the extent of muscle damage will be useful to further investigate the cellular and molecular events involved in muscle regeneration.
Collapse
Affiliation(s)
- Clara Bernard
- Faculté de Médecine et de Pharmacie, Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Charline Jomard
- Faculté de Médecine et de Pharmacie, Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Bénédicte Chazaud
- Faculté de Médecine et de Pharmacie, Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Julien Gondin
- Faculté de Médecine et de Pharmacie, Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| |
Collapse
|
33
|
Johnson AL, Kamal M, Parise G. The Role of Supporting Cell Populations in Satellite Cell Mediated Muscle Repair. Cells 2023; 12:1968. [PMID: 37566047 PMCID: PMC10417507 DOI: 10.3390/cells12151968] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023] Open
Abstract
Skeletal muscle has a high capacity to repair and remodel in response to damage, largely through the action of resident muscle stem cells, termed satellite cells. Satellite cells are required for the proper repair of skeletal muscle through a process known as myogenesis. Recent investigations have observed relationships between satellite cells and other cell types and structures within the muscle microenvironment. These findings suggest that the crosstalk between inflammatory cells, fibrogenic cells, bone-marrow-derived cells, satellite cells, and the vasculature is essential for the restoration of muscle homeostasis. This review will discuss the influence of the cells and structures within the muscle microenvironment on satellite cell function and muscle repair.
Collapse
Affiliation(s)
| | | | - Gianni Parise
- Department of Kinesiology, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
34
|
Stec MJ, Su Q, Adler C, Zhang L, Golann DR, Khan NP, Panagis L, Villalta SA, Ni M, Wei Y, Walls JR, Murphy AJ, Yancopoulos GD, Atwal GS, Kleiner S, Halasz G, Sleeman MW. A cellular and molecular spatial atlas of dystrophic muscle. Proc Natl Acad Sci U S A 2023; 120:e2221249120. [PMID: 37410813 PMCID: PMC10629561 DOI: 10.1073/pnas.2221249120] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 05/24/2023] [Indexed: 07/08/2023] Open
Abstract
Asynchronous skeletal muscle degeneration/regeneration is a hallmark feature of Duchenne muscular dystrophy (DMD); however, traditional -omics technologies that lack spatial context make it difficult to study the biological mechanisms of how asynchronous regeneration contributes to disease progression. Here, using the severely dystrophic D2-mdx mouse model, we generated a high-resolution cellular and molecular spatial atlas of dystrophic muscle by integrating spatial transcriptomics and single-cell RNAseq datasets. Unbiased clustering revealed nonuniform distribution of unique cell populations throughout D2-mdx muscle that were associated with multiple regenerative timepoints, demonstrating that this model faithfully recapitulates the asynchronous regeneration observed in human DMD muscle. By probing spatiotemporal gene expression signatures, we found that propagation of inflammatory and fibrotic signals from locally damaged areas contributes to widespread pathology and that querying expression signatures within discrete microenvironments can identify targetable pathways for DMD therapy. Overall, this spatial atlas of dystrophic muscle provides a valuable resource for studying DMD disease biology and therapeutic target discovery.
Collapse
Affiliation(s)
| | - Qi Su
- Regeneron Pharmaceuticals, Tarrytown, NY10591
| | | | - Lance Zhang
- Regeneron Pharmaceuticals, Tarrytown, NY10591
| | | | | | | | - S. Armando Villalta
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA92697
- Institute for Immunology, University of California Irvine, Irvine, CA92697
- Department of Neurology, University of California Irvine, Irvine, CA92697
| | - Min Ni
- Regeneron Pharmaceuticals, Tarrytown, NY10591
| | - Yi Wei
- Regeneron Pharmaceuticals, Tarrytown, NY10591
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Coulis G, Jaime D, Guerrero-Juarez C, Kastenschmidt JM, Farahat PK, Nguyen Q, Pervolarakis N, McLinden K, Thurlow L, Movahedi S, Hughes BS, Duarte J, Sorn A, Montoya E, Mozaffar I, Dragan M, Othy S, Joshi T, Hans CP, Kimonis V, MacLean AL, Nie Q, Wallace LM, Harper SQ, Mozaffar T, Hogarth MW, Bhattacharya S, Jaiswal JK, Golann DR, Su Q, Kessenbrock K, Stec M, Spencer MJ, Zamudio JR, Villalta SA. Single-cell and spatial transcriptomics identify a macrophage population associated with skeletal muscle fibrosis. SCIENCE ADVANCES 2023; 9:eadd9984. [PMID: 37418531 PMCID: PMC10328414 DOI: 10.1126/sciadv.add9984] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 06/05/2023] [Indexed: 07/09/2023]
Abstract
Macrophages are essential for skeletal muscle homeostasis, but how their dysregulation contributes to the development of fibrosis in muscle disease remains unclear. Here, we used single-cell transcriptomics to determine the molecular attributes of dystrophic and healthy muscle macrophages. We identified six clusters and unexpectedly found that none corresponded to traditional definitions of M1 or M2 macrophages. Rather, the predominant macrophage signature in dystrophic muscle was characterized by high expression of fibrotic factors, galectin-3 (gal-3) and osteopontin (Spp1). Spatial transcriptomics, computational inferences of intercellular communication, and in vitro assays indicated that macrophage-derived Spp1 regulates stromal progenitor differentiation. Gal-3+ macrophages were chronically activated in dystrophic muscle, and adoptive transfer assays showed that the gal-3+ phenotype was the dominant molecular program induced within the dystrophic milieu. Gal-3+ macrophages were also elevated in multiple human myopathies. These studies advance our understanding of macrophages in muscular dystrophy by defining their transcriptional programs and reveal Spp1 as a major regulator of macrophage and stromal progenitor interactions.
Collapse
Affiliation(s)
- Gerald Coulis
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | - Diego Jaime
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | | | - Jenna M. Kastenschmidt
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | - Philip K. Farahat
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | - Quy Nguyen
- Department of Biological Chemistry, University of California Irvine, Irvine, CA USA
| | | | - Katherine McLinden
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Lauren Thurlow
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Saba Movahedi
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Brandon S. Hughes
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Jorge Duarte
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Andrew Sorn
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Elizabeth Montoya
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Izza Mozaffar
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Morgan Dragan
- Department of Biological Chemistry, University of California Irvine, Irvine, CA USA
| | - Shivashankar Othy
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | - Trupti Joshi
- Department of Health Management and Informatics, University of Missouri, Columbia, MO, USA
| | - Chetan P. Hans
- Department of Cardiovascular Medicine, University of Missouri, Columbia, MO USA
| | - Virginia Kimonis
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
| | - Adam L. MacLean
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - Qing Nie
- Department of Mathematics, Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Lindsay M. Wallace
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Scott Q. Harper
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Tahseen Mozaffar
- Department of Neurology, University of California Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA, USA
| | - Marshall W. Hogarth
- Children’s National Hospital, Research Center for Genetic Medicine, Washington, DC, USA
| | - Surajit Bhattacharya
- Children’s National Hospital, Research Center for Genetic Medicine, Washington, DC, USA
| | - Jyoti K. Jaiswal
- Children’s National Hospital, Research Center for Genetic Medicine, Washington, DC, USA
| | | | - Qi Su
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | - Kai Kessenbrock
- Department of Biological Chemistry, University of California Irvine, Irvine, CA USA
| | - Michael Stec
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | - Melissa J. Spencer
- Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Jesse R. Zamudio
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - S. Armando Villalta
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
- Department of Neurology, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
36
|
Mázala DAG, Hindupur R, Moon YJ, Shaikh F, Gamu IH, Alladi D, Panci G, Weiss-Gayet M, Chazaud B, Partridge TA, Novak JS, Jaiswal JK. Altered muscle niche contributes to myogenic deficit in the D2-mdx model of severe DMD. Cell Death Discov 2023; 9:224. [PMID: 37402716 PMCID: PMC10319851 DOI: 10.1038/s41420-023-01503-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/06/2023] [Accepted: 06/19/2023] [Indexed: 07/06/2023] Open
Abstract
Lack of dystrophin expression is the underlying genetic basis for Duchenne muscular dystrophy (DMD). However, disease severity varies between patients, based on specific genetic modifiers. D2-mdx is a model for severe DMD that exhibits exacerbated muscle degeneration and failure to regenerate even in the juvenile stage of the disease. We show that poor regeneration of juvenile D2-mdx muscles is associated with an enhanced inflammatory response to muscle damage that fails to resolve efficiently and supports the excessive accumulation of fibroadipogenic progenitors (FAPs), leading to increased fibrosis. Unexpectedly, the extent of damage and degeneration in juvenile D2-mdx muscle is significantly reduced in adults, and is associated with the restoration of the inflammatory and FAP responses to muscle injury. These improvements enhance regenerative myogenesis in the adult D2-mdx muscle, reaching levels comparable to the milder B10-mdx model of DMD. Ex vivo co-culture of healthy satellite cells (SCs) with juvenile D2-mdx FAPs reduces their fusion efficacy. Wild-type juvenile D2 mice also manifest regenerative myogenic deficit and glucocorticoid treatment improves their muscle regeneration. Our findings indicate that aberrant stromal cell responses contribute to poor regenerative myogenesis and greater muscle degeneration in juvenile D2-mdx muscles and reversal of this reduces pathology in adult D2-mdx muscle, identifying these responses as a potential therapeutic target for the treatment of DMD.
Collapse
Affiliation(s)
- Davi A G Mázala
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, DC, 20012, USA
- Department of Kinesiology, College of Health Professions, Towson University, Towson, MD, 21252, USA
| | - Ravi Hindupur
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, DC, 20012, USA
| | - Young Jae Moon
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, DC, 20012, USA
- Department of Biochemistry and Orthopaedic Surgery, Jeonbuk National University Medical School and Hospital, Jeonju, 54907, Republic of Korea
| | - Fatima Shaikh
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, DC, 20012, USA
| | - Iteoluwakishi H Gamu
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, DC, 20012, USA
| | - Dhruv Alladi
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, DC, 20012, USA
| | - Georgiana Panci
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, INSERM U1513, CNRS UMR 5261, Université Claude Bernard Lyon 1, Univ Lyon, Lyon, France
| | - Michèle Weiss-Gayet
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, INSERM U1513, CNRS UMR 5261, Université Claude Bernard Lyon 1, Univ Lyon, Lyon, France
| | - Bénédicte Chazaud
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, INSERM U1513, CNRS UMR 5261, Université Claude Bernard Lyon 1, Univ Lyon, Lyon, France
| | - Terence A Partridge
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, DC, 20012, USA
- Departments of Pediatrics and Genomics and Precision Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20052, USA
| | - James S Novak
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, DC, 20012, USA.
- Departments of Pediatrics and Genomics and Precision Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20052, USA.
| | - Jyoti K Jaiswal
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, DC, 20012, USA.
- Departments of Pediatrics and Genomics and Precision Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20052, USA.
| |
Collapse
|
37
|
Molinaro C, Scalise M, Leo I, Salerno L, Sabatino J, Salerno N, De Rosa S, Torella D, Cianflone E, Marino F. Polarizing Macrophage Functional Phenotype to Foster Cardiac Regeneration. Int J Mol Sci 2023; 24:10747. [PMID: 37445929 DOI: 10.3390/ijms241310747] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
There is an increasing interest in understanding the connection between the immune and cardiovascular systems, which are highly integrated and communicate through finely regulated cross-talking mechanisms. Recent evidence has demonstrated that the immune system does indeed have a key role in the response to cardiac injury and in cardiac regeneration. Among the immune cells, macrophages appear to have a prominent role in this context, with different subtypes described so far that each have a specific influence on cardiac remodeling and repair. Similarly, there are significant differences in how the innate and adaptive immune systems affect the response to cardiac damage. Understanding all these mechanisms may have relevant clinical implications. Several studies have already demonstrated that stem cell-based therapies support myocardial repair. However, the exact role that cardiac macrophages and their modulation may have in this setting is still unclear. The current need to decipher the dual role of immunity in boosting both heart injury and repair is due, at least for a significant part, to unresolved questions related to the complexity of cardiac macrophage phenotypes. The aim of this review is to provide an overview on the role of the immune system, and of macrophages in particular, in the response to cardiac injury and to outline, through the modulation of the immune response, potential novel therapeutic strategies for cardiac regeneration.
Collapse
Affiliation(s)
- Claudia Molinaro
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Mariangela Scalise
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Isabella Leo
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Luca Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Jolanda Sabatino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Nadia Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Fabiola Marino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| |
Collapse
|
38
|
Lindquist KA, Shein SA, Hovhannisyan AH, Mecklenburg J, Zou Y, Lai Z, Tumanov AV, Akopian AN. Association of inflammation and tissue damage induced biological processes in masseter muscle with the resolution of chronic myalgia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.21.537828. [PMID: 37131723 PMCID: PMC10153356 DOI: 10.1101/2023.04.21.537828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Biological processes linked to intramuscular inflammation during myogenous temporomandibular disorder (TMDM) are largely unknown. We mimicked this inflammation by intra-masseteric muscle (MM) injections of complete Freund’s adjuvant (CFA) or collagenase type 2 (Col), which emulates tissue damage. CFA triggered mechanical hypersensitivity at 1d post-injection was mainly linked to processes controlling chemotactic activity of monocytes and neutrophils. At 5d post-CFA, when hypersensitivity was resolved, there was minimal inflammation whereas tissue repair processes were pronounced. Low dose Col (0.2U) also produced acute orofacial hypersensitivity that was linked to tissue repair, but not inflammatory processes. High dose Col (10U) triggered prolonged orofacial hypersensitivity with inflammatory processes dominating at 1d post-injection. At pre-resolution time point (6d), tissue repair processes were underway and a significant increase in pro-inflammatory gene expressions compared to 1d post-injection were detected. RNA-seq and flow cytometry showed that immune processes in MM were linked to accumulation of macrophages, natural killer and natural killer T cells, dendritic cells and T-cells. Altogether, CFA and Col treatments induced different immune processes in MM. Importantly, orofacial hypersensitivity resolution was preceded with repairs of muscle cell and extracellular matrix, an elevation in immune system gene expression and accumulation of distinct immune cells in MM.
Collapse
|
39
|
Coulis G, Jaime D, Guerrero-Juarez C, Kastenschmidt JM, Farahat PK, Nguyen Q, Pervolarakis N, McLinden K, Thurlow L, Movahedi S, Duarte J, Sorn A, Montoya E, Mozaffar I, Dragan M, Othy S, Joshi T, Hans CP, Kimonis V, MacLean AL, Nie Q, Wallace LM, Harper SQ, Mozaffar T, Hogarth MW, Bhattacharya S, Jaiswal JK, Golann DR, Su Q, Kessenbrock K, Stec M, Spencer MJ, Zamudio JR, Villalta SA. Single-cell and spatial transcriptomics identify a macrophage population associated with skeletal muscle fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.18.537253. [PMID: 37131694 PMCID: PMC10153153 DOI: 10.1101/2023.04.18.537253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The monocytic/macrophage system is essential for skeletal muscle homeostasis, but its dysregulation contributes to the pathogenesis of muscle degenerative disorders. Despite our increasing knowledge of the role of macrophages in degenerative disease, it still remains unclear how macrophages contribute to muscle fibrosis. Here, we used single-cell transcriptomics to determine the molecular attributes of dystrophic and healthy muscle macrophages. We identified six novel clusters. Unexpectedly, none corresponded to traditional definitions of M1 or M2 macrophage activation. Rather, the predominant macrophage signature in dystrophic muscle was characterized by high expression of fibrotic factors, galectin-3 and spp1. Spatial transcriptomics and computational inferences of intercellular communication indicated that spp1 regulates stromal progenitor and macrophage interactions during muscular dystrophy. Galectin-3 + macrophages were chronically activated in dystrophic muscle and adoptive transfer assays showed that the galectin-3 + phenotype was the dominant molecular program induced within the dystrophic milieu. Histological examination of human muscle biopsies revealed that galectin-3 + macrophages were also elevated in multiple myopathies. These studies advance our understanding of macrophages in muscular dystrophy by defining the transcriptional programs induced in muscle macrophages, and reveal spp1 as a major regulator of macrophage and stromal progenitor interactions.
Collapse
Affiliation(s)
- Gerald Coulis
- Department of Physiology and Biophysics, University of California Irvine, USA
- Institute for Immunology, University of California Irvine, USA
| | - Diego Jaime
- Department of Physiology and Biophysics, University of California Irvine, USA
- Institute for Immunology, University of California Irvine, USA
| | - Christian Guerrero-Juarez
- Department of Mathematics, University of California Irvine, USA
- Department of Developmental and Cell Biology, University of California Irvine, USA
| | - Jenna M. Kastenschmidt
- Department of Physiology and Biophysics, University of California Irvine, USA
- Institute for Immunology, University of California Irvine, USA
| | - Philip K. Farahat
- Department of Physiology and Biophysics, University of California Irvine, USA
- Institute for Immunology, University of California Irvine, USA
| | - Quy Nguyen
- Department of Biological Chemistry, University of California Irvine, USA
| | | | - Katherine McLinden
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, USA
| | - Lauren Thurlow
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, USA
| | - Saba Movahedi
- Department of Physiology and Biophysics, University of California Irvine, USA
| | - Jorge Duarte
- Department of Physiology and Biophysics, University of California Irvine, USA
| | - Andrew Sorn
- Department of Physiology and Biophysics, University of California Irvine, USA
| | - Elizabeth Montoya
- Department of Physiology and Biophysics, University of California Irvine, USA
| | - Izza Mozaffar
- Department of Physiology and Biophysics, University of California Irvine, USA
| | - Morgan Dragan
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, USA
| | - Shivashankar Othy
- Department of Physiology and Biophysics, University of California Irvine, USA
- Institute for Immunology, University of California Irvine, USA
| | - Trupti Joshi
- Department of Health Management and Informatics, University of Missouri, Columbia, USA
| | - Chetan P. Hans
- Department of Cardiovascular Medicine, University of Missouri, Columbia, USA
| | | | - Adam L. MacLean
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, USA
| | - Qing Nie
- Department of Mathematics, University of California Irvine, USA
- Department of Developmental and Cell Biology, University of California Irvine, USA
| | - Lindsay M. Wallace
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital
| | - Scott Q. Harper
- Department of Pediatrics, The Ohio State University, Columbus, OH, USA
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital
| | - Tahseen Mozaffar
- Department of Neurology, University of California Irvine, USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, USA
| | - Marshall W. Hogarth
- Children’s National Hospital, Research Center for Genetic Medicine, Washington, DC, USA
| | - Surajit Bhattacharya
- Children’s National Hospital, Research Center for Genetic Medicine, Washington, DC, USA
| | - Jyoti K. Jaiswal
- Children’s National Hospital, Research Center for Genetic Medicine, Washington, DC, USA
| | | | - Qi Su
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Kai Kessenbrock
- Department of Biological Chemistry, University of California Irvine, USA
| | - Michael Stec
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | - Jesse R. Zamudio
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, USA
| | - S. Armando Villalta
- Department of Physiology and Biophysics, University of California Irvine, USA
- Institute for Immunology, University of California Irvine, USA
- Department of Neurology, University of California Irvine, USA
| |
Collapse
|
40
|
Mázala DAG, Hindupur R, Moon YJ, Shaikh F, Gamu IH, Alladi D, Panci G, Weiss-Gayet M, Chazaud B, Partridge TA, Novak JS, Jaiswal JK. Altered muscle niche contributes to myogenic deficit in the D2- mdx model of severe DMD. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.27.534413. [PMID: 37034785 PMCID: PMC10081277 DOI: 10.1101/2023.03.27.534413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Lack of dystrophin is the genetic basis for the Duchenne muscular dystrophy (DMD). However, disease severity varies between patients, based on specific genetic modifiers. D2- mdx is a model for severe DMD that exhibits exacerbated muscle degeneration and failure to regenerate even in the juvenile stage of the disease. We show that poor regeneration of juvenile D2- mdx muscles is associated with enhanced inflammatory response to muscle damage that fails to resolve efficiently and supports excessive accumulation of fibroadipogenic progenitors (FAPs). Unexpectedly, the extent of damage and degeneration of juvenile D2- mdx muscle is reduced in adults and is associated with the restoration of the inflammatory and FAP responses to muscle injury. These improvements enhance myogenesis in the adult D2- mdx muscle, reaching levels comparable to the milder (B10- mdx ) mouse model of DMD. Ex vivo co-culture of healthy satellite cells (SCs) with the juvenile D2- mdx FAPs reduced their fusion efficacy and in vivo glucocorticoid treatment of juvenile D2 mouse improved muscle regeneration. Our findings indicate that aberrant stromal cell response contributes to poor myogenesis and greater muscle degeneration in dystrophic juvenile D2- mdx muscles and reversal of this reduces pathology in adult D2- mdx mouse muscle, identifying these as therapeutic targets to treat dystrophic DMD muscles.
Collapse
Affiliation(s)
- Davi A. G. Mázala
- Center for Genetic Medicine Research, Children’s National Research Institute, Children’s National Research and Innovation Campus, Children’s National Hospital, Washington, D.C., 20012, USA
- Department of Kinesiology, College of Health Professions, Towson University, Towson, MD, 21252, USA
| | - Ravi Hindupur
- Center for Genetic Medicine Research, Children’s National Research Institute, Children’s National Research and Innovation Campus, Children’s National Hospital, Washington, D.C., 20012, USA
| | - Young Jae Moon
- Center for Genetic Medicine Research, Children’s National Research Institute, Children’s National Research and Innovation Campus, Children’s National Hospital, Washington, D.C., 20012, USA
- Department of Biochemistry and Orthopaedic Surgery, Jeonbuk National University Medical School and Hospital, Jeonju, 54907, Republic of Korea
| | - Fatima Shaikh
- Center for Genetic Medicine Research, Children’s National Research Institute, Children’s National Research and Innovation Campus, Children’s National Hospital, Washington, D.C., 20012, USA
| | - Iteoluwakishi H. Gamu
- Center for Genetic Medicine Research, Children’s National Research Institute, Children’s National Research and Innovation Campus, Children’s National Hospital, Washington, D.C., 20012, USA
| | - Dhruv Alladi
- Center for Genetic Medicine Research, Children’s National Research Institute, Children’s National Research and Innovation Campus, Children’s National Hospital, Washington, D.C., 20012, USA
| | - Georgiana Panci
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, INSERM U1513, CNRS UMR 5261, Université Claude Bernard Lyon 1, Univ Lyon, Lyon, France
| | - Michèle Weiss-Gayet
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, INSERM U1513, CNRS UMR 5261, Université Claude Bernard Lyon 1, Univ Lyon, Lyon, France
| | - Bénédicte Chazaud
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, INSERM U1513, CNRS UMR 5261, Université Claude Bernard Lyon 1, Univ Lyon, Lyon, France
| | - Terence A. Partridge
- Center for Genetic Medicine Research, Children’s National Research Institute, Children’s National Research and Innovation Campus, Children’s National Hospital, Washington, D.C., 20012, USA
- Departments of Pediatrics and Genomics and Precision Medicine, The George Washington University School of Medicine and Health Sciences, Washington, D.C., 20052, USA
| | - James S. Novak
- Center for Genetic Medicine Research, Children’s National Research Institute, Children’s National Research and Innovation Campus, Children’s National Hospital, Washington, D.C., 20012, USA
- Departments of Pediatrics and Genomics and Precision Medicine, The George Washington University School of Medicine and Health Sciences, Washington, D.C., 20052, USA
| | - Jyoti K. Jaiswal
- Center for Genetic Medicine Research, Children’s National Research Institute, Children’s National Research and Innovation Campus, Children’s National Hospital, Washington, D.C., 20012, USA
- Departments of Pediatrics and Genomics and Precision Medicine, The George Washington University School of Medicine and Health Sciences, Washington, D.C., 20052, USA
| |
Collapse
|
41
|
Groppa E, Martini P, Derakhshan N, Theret M, Ritso M, Tung LW, Wang YX, Soliman H, Hamer MS, Stankiewicz L, Eisner C, Erwan LN, Chang C, Yi L, Yuan JH, Kong S, Weng C, Adams J, Chang L, Peng A, Blau HM, Romualdi C, Rossi FMV. Spatial compartmentalization of signaling imparts source-specific functions on secreted factors. Cell Rep 2023; 42:112051. [PMID: 36729831 DOI: 10.1016/j.celrep.2023.112051] [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: 10/04/2020] [Revised: 09/08/2022] [Accepted: 01/16/2023] [Indexed: 02/03/2023] Open
Abstract
Efficient regeneration requires multiple cell types acting in coordination. To better understand the intercellular networks involved and how they change when regeneration fails, we profile the transcriptome of hematopoietic, stromal, myogenic, and endothelial cells over 14 days following acute muscle damage. We generate a time-resolved computational model of interactions and identify VEGFA-driven endothelial engagement as a key differentiating feature in models of successful and failed regeneration. In addition, the analysis highlights that the majority of secreted signals, including VEGFA, are simultaneously produced by multiple cell types. To test whether the cellular source of a factor determines its function, we delete VEGFA from two cell types residing in close proximity: stromal and myogenic progenitors. By comparing responses to different types of damage, we find that myogenic and stromal VEGFA have distinct functions in regeneration. This suggests that spatial compartmentalization of signaling plays a key role in intercellular communication networks.
Collapse
Affiliation(s)
- Elena Groppa
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada; Borea Therapeutics, Scuola Internazionale Superiore di Studi Avanzati, Via Bonomea 265, Trieste, Italy
| | - Paolo Martini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Biology, University of Padova, via U. Bassi 58B, Padova, Italy
| | - Nima Derakhshan
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Marine Theret
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Morten Ritso
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Lin Wei Tung
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Yu Xin Wang
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Hesham Soliman
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada; Faculty of Pharmaceutical Sciences, Minia University, Minia, Egypt; Aspect Biosystems, 1781 W 75th Avenue, Vancouver, BC, Canada
| | - Mark Stephen Hamer
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Laura Stankiewicz
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Christine Eisner
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Le Nevé Erwan
- Department of Pediatrics, Université Laval, Laval, QC, Canada
| | - Chihkai Chang
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Lin Yi
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Jack H Yuan
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Sunny Kong
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Curtis Weng
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Josephine Adams
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Lucas Chang
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Anne Peng
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada
| | - Helen M Blau
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Chiara Romualdi
- Department of Biology, University of Padova, via U. Bassi 58B, Padova, Italy
| | - Fabio M V Rossi
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, Canada.
| |
Collapse
|
42
|
Tu H, Li YL. Inflammation balance in skeletal muscle damage and repair. Front Immunol 2023; 14:1133355. [PMID: 36776867 PMCID: PMC9909416 DOI: 10.3389/fimmu.2023.1133355] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 01/12/2023] [Indexed: 01/27/2023] Open
Abstract
Responding to tissue injury, skeletal muscles undergo the tissue destruction and reconstruction accompanied with inflammation. The immune system recognizes the molecules released from or exposed on the damaged tissue. In the local minor tissue damage, tissue-resident macrophages sequester pro-inflammatory debris to prevent initiation of inflammation. In most cases of the skeletal muscle injury, however, a cascade of inflammation will be initiated through activation of local macrophages and mast cells and recruitment of immune cells from blood circulation to the injured site by recongnization of damage-associated molecular patterns (DAMPs) and activated complement system. During the inflammation, macrophages and neutrophils scavenge the tissue debris to release inflammatory cytokines and the latter stimulates myoblast fusion and vascularization to promote injured muscle repair. On the other hand, an abundance of released inflammatory cytokines and chemokines causes the profound hyper-inflammation and mobilization of immune cells to trigger a vicious cycle and lead to the cytokine storm. The cytokine storm results in the elevation of cytolytic and cytotoxic molecules and reactive oxygen species (ROS) in the damaged muscle to aggravates the tissue injury, including the healthy bystander tissue. Severe inflammation in the skeletal muscle can lead to rhabdomyolysis and cause sepsis-like systemic inflammation response syndrome (SIRS) and remote organ damage. Therefore, understanding more details on the involvement of inflammatory factors and immune cells in the skeletal muscle damage and repair can provide the new precise therapeutic strategies, including attenuation of the muscle damage and promotion of the muscle repair.
Collapse
|
43
|
Panci G, E M Kneppers A, Mounier R, Chazaud B, Juban G. Co-cultures of Macrophages with Muscle Stem Cells with Fibroadipogenic Precursor Cells from Regenerating Skeletal Muscle. Methods Mol Biol 2023; 2640:57-71. [PMID: 36995587 DOI: 10.1007/978-1-0716-3036-5_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Adult muscle stem cells rebuild myofibers after damage. Although they are highly powerful to implement the adult myogenic program, they need environmental cues provided by surrounding cells for efficient and complete regeneration. Muscle stem cell environment includes fibroadipogenic precursors, vascular cells, and macrophages. A way to decipher the complexity of the interactions muscle stem cells establish with their neighborhood is to co-culture cells freshly isolated from the muscle and assess the impact of one cell type on the behavior/fate of the other cell type. Here, we present a protocol allowing the isolation of primary muscle stem cells, macrophages, and fibroadipogenic precursors by Fluorescence Activated Cell Sorting (FACS) or Magnetic Cell Separation (MACS), together with co-culture methods using a specific setup for a short time window to keep as much as possible the in vivo properties of the isolated cells.
Collapse
Affiliation(s)
- Georgiana Panci
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, CNRS, UMR5261, INSERM U1315, Université Claude Bernard Lyon 1, Lyon, France
| | - Anita E M Kneppers
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, CNRS, UMR5261, INSERM U1315, Université Claude Bernard Lyon 1, Lyon, France
| | - Rémi Mounier
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, CNRS, UMR5261, INSERM U1315, Université Claude Bernard Lyon 1, Lyon, France
| | - Bénédicte Chazaud
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, CNRS, UMR5261, INSERM U1315, Université Claude Bernard Lyon 1, Lyon, France.
| | - Gaëtan Juban
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, CNRS, UMR5261, INSERM U1315, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
44
|
Ahmadi M, Karlsen A, Mehling J, Soendenbroe C, Mackey AL, Hyldahl RD. Aging is associated with an altered macrophage response during human skeletal muscle regeneration. Exp Gerontol 2022; 169:111974. [PMID: 36228835 DOI: 10.1016/j.exger.2022.111974] [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: 08/02/2022] [Revised: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 12/15/2022]
Abstract
Skeletal muscle injury in aged rodents is characterized by an asynchronous infiltration of pro- and anti-inflammatory macrophage waves, leading to improper and incomplete regeneration. It is unclear whether this aberration also occurs in aged human muscle. In this study, we quantified the macrophage responses in a human model of muscle damage and regeneration induced by electrical stimulation in 7 young and 21 older adults. At baseline, total resident macrophage (CD68+/DAPI+) content was not different between young and old subjects, but pro-inflammatory (CD206-/CD68+/DAPI+) macrophage content was lower in the old. Following damage, muscle Infiltration of CD206-/CD68+/DAPI+ macrophages was lower in old relative to young subjects. Further, only the increase in CD206-/CD68+ macrophages correlated with the change in muscle satellite cell content. Our data show that older individuals have a compromised macrophage response during muscle regeneration, pointing to an altered inflammatory response as a potential mechanism for reduced muscle regenerative efficacy in aged humans.
Collapse
Affiliation(s)
- Mohadeseh Ahmadi
- Department of Exercise Sciences, Brigham Young University, Provo, UT 84602, USA
| | - Anders Karlsen
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark; Center for Healthy Aging, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jack Mehling
- Department of Exercise Sciences, Brigham Young University, Provo, UT 84602, USA
| | - Casper Soendenbroe
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark; Center for Healthy Aging, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Abigail L Mackey
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark; Center for Healthy Aging, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Robert D Hyldahl
- Department of Exercise Sciences, Brigham Young University, Provo, UT 84602, USA.
| |
Collapse
|
45
|
Yeh TS, Lei TH, Barnes MJ, Zhang L. Astragalosides Supplementation Enhances Intrinsic Muscle Repair Capacity Following Eccentric Exercise-Induced Injury. Nutrients 2022; 14:4339. [PMID: 36297022 PMCID: PMC9608496 DOI: 10.3390/nu14204339] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 10/26/2023] Open
Abstract
Astragalosides have been shown to enhance endurance exercise capacity in vivo and promote muscular hypertrophy in vitro. However, it remains unknown whether astragalosides supplementation can alter inflammatory response and enhance muscle recovery after damage in humans. We therefore aimed to evaluate the effect of astragalosides supplementation on muscle's intrinsic capacity to regenerate and repair itself after exercise-induced damage. Using a randomized double-blind placebo-controlled cross-over design, eleven male participants underwent 7 days of astragalosides supplementation (in total containing 4 mg of astragalosides per day) or a placebo control, following an eccentric exercise protocol. Serum blood samples and variables related to muscle function were collected prior to and immediately following the muscle damage protocol and also at 2 h, and 1, 2, 3, 5, and 7 days of the recovery period, to assess the pro-inflammatory cytokine response, the secretion of muscle regenerative factors, and muscular strength. Astragalosides supplementation reduced biomarkers of skeletal muscle damage (serum CK, LDH, and Mb), when compared to the placebo, at 1, 2, and 3 days following the muscle damage protocol. Astragalosides supplementation suppressed the secretion of IL-6 and TNF-α, whilst increasing the release of IGF-1 during the initial stages of muscle recovery. Furthermore, following astragaloside supplementation, muscular strength returned to baseline 2 days earlier than the placebo. Astragalosides supplementation shortens the duration of inflammation, enhances the regeneration process and restores muscle strength following eccentric exercise-induced injury.
Collapse
Affiliation(s)
- Tzu-Shao Yeh
- School of Public Health, Nantong University, Nantong 226019, China
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Tze-Huan Lei
- College of Physical Education, Hubei Normal University, Huangshi 435002, China
| | - Matthew J. Barnes
- School of Sport, Exercise and Nutrition, Massey University, Palmerston North 4410, New Zealand
| | - Lei Zhang
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| |
Collapse
|
46
|
Bernard C, Zavoriti A, Pucelle Q, Chazaud B, Gondin J. Role of macrophages during skeletal muscle regeneration and hypertrophy-Implications for immunomodulatory strategies. Physiol Rep 2022; 10:e15480. [PMID: 36200266 PMCID: PMC9535344 DOI: 10.14814/phy2.15480] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/12/2022] [Indexed: 06/16/2023] Open
Abstract
Skeletal muscle is a plastic tissue that regenerates ad integrum after injury and adapts to raise mechanical loading/contractile activity by increasing its mass and/or myofiber size, a phenomenon commonly refers to as skeletal muscle hypertrophy. Both muscle regeneration and hypertrophy rely on the interactions between muscle stem cells and their neighborhood, which include inflammatory cells, and particularly macrophages. This review first summarizes the role of macrophages in muscle regeneration in various animal models of injury and in response to exercise-induced muscle damage in humans. Then, the potential contribution of macrophages to skeletal muscle hypertrophy is discussed on the basis of both animal and human experiments. We also present a brief comparative analysis of the role of macrophages during muscle regeneration versus hypertrophy. Finally, we summarize the current knowledge on the impact of different immunomodulatory strategies, such as heat therapy, cooling, massage, nonsteroidal anti-inflammatory drugs and resolvins, on skeletal muscle regeneration and their potential impact on muscle hypertrophy.
Collapse
Affiliation(s)
- Clara Bernard
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du MuscleUniversité Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Université LyonLyonFrance
| | - Aliki Zavoriti
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du MuscleUniversité Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Université LyonLyonFrance
| | - Quentin Pucelle
- Université de Versailles Saint‐Quentin‐En‐YvelinesVersaillesFrance
| | - Bénédicte Chazaud
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du MuscleUniversité Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Université LyonLyonFrance
| | - Julien Gondin
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du MuscleUniversité Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Université LyonLyonFrance
| |
Collapse
|
47
|
Teixeira E, Garcia J, Bovolini A, Carvalho A, Pacheco J, Duarte JA. Sedentary Behaviour Impairs Skeletal Muscle Repair Modulating the Inflammatory Response. J Funct Morphol Kinesiol 2022; 7:jfmk7040076. [PMID: 36278737 PMCID: PMC9589940 DOI: 10.3390/jfmk7040076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/24/2022] [Accepted: 09/24/2022] [Indexed: 11/24/2022] Open
Abstract
This study investigated whether sedentary behaviour modulates skeletal-muscle repair and tissue inflammatory response after cardiotoxin (CTX)-induced injury. Singly caged rats spent 8 weeks either as a sedentary group (SED, n = 15) or as a control group (EX, n = 15)—caged with running wheels for voluntary running. All rats had each tibial anterior muscle infused either with CTX (CTX; right muscle) or saline solution (Sham; left muscle) and were sacrificed (n = 5 per group) on the 1st, 7th, and 15th day post-injection (dpi). Histological and immunohistochemical analyses were used to calculate myotube percentage and fibrosis accretion, and quantify the number of neutrophils and M1 and M2 macrophage subtypes. The SED group showed an increased number of both neutrophils and M1 macrophages (7th and 15th dpi) compared to the EX group (p < 0.01). The EX group showed an increased number of M2 macrophages on the 1st dpi. On the 7th dpi, the SED group showed a lower myotube percentage compared to the EX group (p < 0.01) and on the 15th dpi showed only 54% of normal undamaged fibres compared to 90% from the EX group (p < 0.01). The SED group showed increased fibrosis on both the 7th and 15th dpi. Our results show that sedentary behaviour affects the inflammatory response, enhancing and prolonging the Th1 phase, and delays and impairs the SMR process.
Collapse
Affiliation(s)
- Eduardo Teixeira
- Research Center in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sports, University of Porto (FADEUP) and Laboratory for Integrative and Translational Research in Population Health (ITR), 4200-450 Porto, Portugal
- Faculty of Psychology, Education and Sports, Lusófona University of Porto, 4000-098 Porto, Portugal
- Escola Superior Desporto e Lazer, Instituto Politécnico de Viana do Castelo, 4960-320 Melgaço, Portugal
- Correspondence:
| | - Juliana Garcia
- Centre for the Research and Technology of Agro-Environment and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
- AquaValor-Centro de Valorização e Transferência de Tecnologia da Água-Associação, 5400-342 Chaves, Portugal
| | - António Bovolini
- Instituto Politécnico da Guarda, Unidade de Investigação para o Desenvolvimento do Interior (UDI), 6300-559 Guarda, Portugal
| | - Ana Carvalho
- Research Center in Sports Sciences, Health Sciences and Human Development (CIDESD), University Institute of Maia, 4475-690 Maia, Portugal
| | - Júlio Pacheco
- Research Center in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sports, University of Porto (FADEUP) and Laboratory for Integrative and Translational Research in Population Health (ITR), 4200-450 Porto, Portugal
| | - José A. Duarte
- Toxicology Research Unit (TOXRUN), University Institute of Health Sciences (IUCS), Advanced Polytechnic and University Cooperative (CESPU), CRL, 4585-116 Gandra, Portugal
| |
Collapse
|
48
|
Crescioli C. Vitamin D, exercise, and immune health in athletes: A narrative review. Front Immunol 2022; 13:954994. [PMID: 36211340 PMCID: PMC9539769 DOI: 10.3389/fimmu.2022.954994] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022] Open
Abstract
Vitamin D exerts important extra-skeletal effects, exhibiting an exquisite immune regulatory ability, affecting both innate and adaptive immune responses through the modulation of immunocyte function and signaling. Remarkably, the immune function of working skeletal muscle, which is fully recognized to behave as a secretory organ with immune capacity, is under the tight control of vitamin D as well. Vitamin D status, meaning hormone sufficiency or insufficiency, can push toward strengthening/stabilization or decline of immune surveillance, with important consequences for health. This aspect is particularly relevant when considering the athletic population: while exercising is, nowadays, the recommended approach to maintain health and counteract inflammatory processes, “too much” exercise, often experienced by athletes, can increase inflammation, decrease immune surveillance, and expose them to a higher risk of diseases. When overexercise intersects with hypovitaminosis D, the overall effects on the immune system might converge into immune depression and higher vulnerability to diseases. This paper aims to provide an overview of how vitamin D shapes human immune responses, acting on the immune system and skeletal muscle cells; some aspects of exercise-related immune modifications are addressed, focusing on athletes. The crossroad where vitamin D and exercise meet can profile whole-body immune response and health.
Collapse
|
49
|
Chen YF, Lee CW, Wu HH, Lin WT, Lee OK. Immunometabolism of macrophages regulates skeletal muscle regeneration. Front Cell Dev Biol 2022; 10:948819. [PMID: 36147742 PMCID: PMC9485946 DOI: 10.3389/fcell.2022.948819] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
Sarcopenia is an age-related progressive loss of skeletal muscle mass, quality, and strength disease. In addition, sarcopenia is tightly correlated with age-associated pathologies, such as sarcopenic obesity and osteoporosis. Further understanding of disease mechanisms and the therapeutic strategies in muscle regeneration requires a deeper knowledge of the interaction of skeletal muscle and other cells in the muscle tissue. Skeletal muscle regeneration is a complex process that requires a series of highly coordinated events involving communication between muscle stem cells and niche cells, such as muscle fibro/adipogenic progenitors and macrophages. Macrophages play a critical role in tissue regeneration and the maintenance of muscle homeostasis by producing growth factors and cytokines that regulate muscle stem cells and myofibroblast activation. Furthermore, the aging-related immune dysregulation associated with the release of trophic factors and the polarization in macrophages transiently affect the inflammatory phase and impair muscle regeneration. In this review, we focus on the role and regulation of macrophages in skeletal muscle regeneration and homeostasis. The aim of this review is to highlight the important roles of macrophages as a therapeutic target in age-related sarcopenia and the increasing understanding of how macrophages are regulated will help to advance skeletal muscle regeneration.
Collapse
Affiliation(s)
- Yu-Fan Chen
- Center for Translational Genomics Research, China Medical University Hospital, Taichung, Taiwan
| | - Chien-Wei Lee
- Center for Translational Genomics Research, China Medical University Hospital, Taichung, Taiwan
| | - Hao-Hsiang Wu
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Stem Cell Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Ting Lin
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Doctoral Degree Program of Translational Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Oscar K. Lee
- Center for Translational Genomics Research, China Medical University Hospital, Taichung, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Stem Cell Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Orthopedics, China Medical University Hospital, Taichung, Taiwan
- *Correspondence: Oscar K. Lee,
| |
Collapse
|
50
|
Kokubo K, Onodera A, Kiuchi M, Tsuji K, Hirahara K, Nakayama T. Conventional and pathogenic Th2 cells in inflammation, tissue repair, and fibrosis. Front Immunol 2022; 13:945063. [PMID: 36016937 PMCID: PMC9395650 DOI: 10.3389/fimmu.2022.945063] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/18/2022] [Indexed: 12/15/2022] Open
Abstract
Type 2 helper T (Th2) cells, a subset of CD4+ T cells, play an important role in the host defense against pathogens and allergens by producing Th2 cytokines, such as interleukin-4 (IL-4), IL-5, and IL-13, to trigger inflammatory responses. Emerging evidence reveals that Th2 cells also contribute to the repair of injured tissues after inflammatory reactions. However, when the tissue repair process becomes chronic, excessive, or uncontrolled, pathological fibrosis is induced, leading to organ failure and death. Thus, proper control of Th2 cells is needed for complete tissue repair without the induction of fibrosis. Recently, the existence of pathogenic Th2 (Tpath2) cells has been revealed. Tpath2 cells produce large amounts of Th2 cytokines and induce type 2 inflammation when activated by antigen exposure or tissue injury. In recent studies, Tpath2 cells are suggested to play a central role in the induction of type 2 inflammation whereas the role of Tpath2 cells in tissue repair and fibrosis has been less reported in comparison to conventional Th2 cells. In this review, we discuss the roles of conventional Th2 cells and pathogenic Th2 cells in the sequence of tissue inflammation, repair, and fibrosis.
Collapse
Affiliation(s)
- Kota Kokubo
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Atsushi Onodera
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Institute for Advanced Academic Research, Chiba University, Chiba, Japan
| | - Masahiro Kiuchi
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kaori Tsuji
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kiyoshi Hirahara
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- *Correspondence: Kiyoshi Hirahara, ; Toshinori Nakayama,
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- AMED-CREST, AMED, Chiba, Japan
- *Correspondence: Kiyoshi Hirahara, ; Toshinori Nakayama,
| |
Collapse
|