1
|
Pang J, Xu D, Zhang X, Qu J, Jiang J, Suo J, Li T, Li Y, Peng Z. TIMP2-mediated mitochondrial fragmentation and glycolytic reprogramming drive renal fibrogenesis following ischemia-reperfusion injury. Free Radic Biol Med 2025; 232:244-259. [PMID: 39986488 DOI: 10.1016/j.freeradbiomed.2025.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/11/2025] [Accepted: 02/14/2025] [Indexed: 02/24/2025]
Abstract
Acute kidney injury (AKI) triggers renal structural and functional abnormalities through inflammatory and fibrotic signaling pathways, ultimately progressing to chronic kidney disease (CKD). The mechanisms underlying AKI-to-CKD transition are complex, with hypoxia, mitochondrial dysfunction, and metabolic reprogramming as critical contributors. Public data analysis demonstrated significant upregulation of tissue inhibitors of metalloproteinases (Timp2) in renal biopsy tissues of CKD patients. In both ischemia/reperfusion (I/R) and unilateral ureteral obstruction (UUO) models, Timp2 upregulation was observed. Tubule-specific Timp2 knockout markedly attenuated renal fibrosis. RNA-sequencing revealed Timp2's association with mitochondrial dynamics and glycolysis in I/R mice. Timp2 deletion improved mitochondrial morphology and suppressed glycolytic enzyme expression. In vitro, TGF-β1-treated Timp2-knockdown HK-2 cells exhibited inhibited Drp1 expression, restored Mfn2 levels, alleviated mitochondrial fragmentation, and elevated mitochondrial membrane potential. Additionally, Pfkfb3 and HIF-1α were downregulated, accompanied by reduced extracellular acidification rate (ECAR), PFK activity, and lactate production. Mechanistically, Timp2 interacts with the extracellular domain of Sdc4 in an autocrine manner, activating the Hedgehog (Hh) signaling pathway. Cyclopamine partially rescued Timp2 overexpression-induced mitochondrial dysfunction, suppressed Pfkfb3-mediated glycolysis, and diminished collagen deposition. This study is the first to demonstrate that Timp2 in TECs exacerbates Hh signaling, promoting mitochondrial fragmentation and metabolic reprogramming to accelerate I/R-induced renal fibrosis.
Collapse
Affiliation(s)
- Jingjing Pang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Dongxue Xu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China.
| | - Xiaoyu Zhang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Jiacheng Qu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Jun Jiang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Jinmeng Suo
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Tianlong Li
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Yiming Li
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China.
| | - Zhiyong Peng
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Intensive Care Unit of the Second Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China.
| |
Collapse
|
2
|
Luo YE, Abe-Teh Z, Alsaghir TY, Kuo LY, Yu F, Stoker BE, Appu AB, Zhou Y, Yue F, Kopinke D, Barton ER. Fibro-Adipogenic Progenitors require autocrine IGF-I in homeostatic and regenerating skeletal muscle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.11.648330. [PMID: 40291730 PMCID: PMC12027368 DOI: 10.1101/2025.04.11.648330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Fibro-Adipogenic Progenitors (FAPs) are mesenchymal stem cells that are vital for muscle homeostasis and regeneration but produce fibrosis and intramuscular fat under pathological conditions. Insulin-like Growth Factor-I (IGF-I) is a key regulator of muscle repair, satellite cell activity, macrophage polarization, and extracellular matrix (ECM) remodeling. We generated inducible FAP-specific Igf1 deficient (FID) mice to determine the necessity of FAP IGF-I. After BaCl 2 injury, FID mice exhibited impaired muscle regeneration, with fewer Pax7+ cells, increased macrophage accumulation, smaller fibers, reduced ECM, and depressed FAP proliferation. Following glycerol injury, FID muscles exhibited reduced adipocyte accumulation. Primary FAPs isolated from injured FID muscles had blunted growth, upregulation of immune-regulatory genes and downregulation of ECM and cell proliferation genes, with delayed responses to fibrogenic and to adipogenic media. FAP property alterations were already present in homeostatic muscle, indicated by scRNASeq, with decreased indices of protein translation and ECM production as well as increased markers of senescence, confirmed in vivo and in vitro . Overall, FAP IGF-I is a critical autocrine factor, with further paracrine consequences for muscle regenerative capacity.
Collapse
|
3
|
Zhang P, Zhou M, Zhu Y, Xie J, Huo Z, Zhang D, Li P, Guo J, Li G, Li X, Wang R, Jiang C. The Glycogen Synthase Kinase-3 Inhibitor CHIR99021 Reduces Fatty Infiltration and Muscle Atrophy After Rotator Cuff Tears: An In Vitro Experiment and In Vivo Mouse Model. Am J Sports Med 2025; 53:1184-1194. [PMID: 39989144 PMCID: PMC12084656 DOI: 10.1177/03635465251319549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 12/10/2024] [Indexed: 02/25/2025]
Abstract
BACKGROUND Rotator cuff tears (RCTs) can cause inflammation, muscle atrophy, and irreversible fatty infiltration, resulting in poor clinical outcomes. Effective therapeutic approaches to inhibit fatty infiltration in rotator cuff muscles remain limited. PURPOSE To identify pathways associated with fatty infiltration through RNA sequencing and to evaluate the therapeutic potential of the glycogen synthase kinase-3 (GSK-3) inhibitor CHIR99021 based on enrichment of the Akt/GSK-3 pathway identified by RNA sequencing. STUDY DESIGN Controlled laboratory study. METHODS Supraspinatus muscle biopsy specimens from 6 patients with chronic full-thickness RCTs were analyzed by RNA sequencing. Fibro-adipogenic progenitors (FAPs) or C2C12 myoblasts were cultured with different doses of CHIR99021 to assess their effects on adipogenic or myogenic differentiation, respectively. RNA sequencing identified cellular pathways in FAPs treated with or without CHIR99021. A mouse RCT model was established by detaching the supraspinatus tendon, followed by treatment with or without CHIR99021 administered intraperitoneally. Muscle atrophy and fatty infiltration were assessed histologically and through gene expression analysis at 1 and 4 weeks after surgery. RESULTS RNA sequencing analysis identified a marked upregulation of the Akt/GSK-3 signaling pathway specifically in patients' samples and FAPs with minimal fat accumulation. CHIR99021 suppressed adipogenic differentiation in FAPs and promoted myogenic differentiation in C2C12 cells. In the mouse RCT model, CHIR99021-treated mice exhibited reduced Oil Red O staining, a larger cross-sectional area, and less muscle weight loss in the supraspinatus muscle compared with the vehicle-treated mice. Gene expression analysis indicated increased myogenesis and reduced fatty infiltration at 1 and 4 weeks after surgery as well as increased expression levels of IL-6 and IL-15 in the CHIR99021 group compared with the control group at 1 week after surgery. CONCLUSION The Akt/GSK-3 pathway was enriched in supraspinatus muscle samples and FAPs with low fat accumulation, highlighting its potential as a therapeutic target. The GSK-3 inhibitor CHIR99021 was shown to alleviate fatty infiltration and muscle atrophy after RCTs in vitro and in vivo in a mouse model. CLINICAL RELEVANCE The GSK-3 inhibitor CHIR99021 shows potential for treating muscle degeneration after RCTs.
Collapse
Affiliation(s)
- Pu Zhang
- Fourth School of Clinical Medicine, Peking University, Beijing, China
| | - Meng Zhou
- Sports Medicine Service, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
- Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Yiming Zhu
- Sports Medicine Service, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Jianhao Xie
- Fourth School of Clinical Medicine, Peking University, Beijing, China
| | - Ziqi Huo
- Sports Medicine Service, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Dan Zhang
- Fourth School of Clinical Medicine, Peking University, Beijing, China
| | - Pinxue Li
- Fourth School of Clinical Medicine, Peking University, Beijing, China
| | - Jianxun Guo
- JST Sarcopenia Research Centre, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Guangping Li
- Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Xu Li
- Sports Medicine Service, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Renxian Wang
- JST Sarcopenia Research Centre, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Chunyan Jiang
- Fourth School of Clinical Medicine, Peking University, Beijing, China
- Sports Medicine Service, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
- Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Lan M, Qin Q, Xie Y, Zhang C, Liu Z, Xu X, Zhang J, Xu S, Yang J, Zhang H, Alatan S, Wang Z, Liu Z. Construction of ceRNA networks of lncRNA and miRNA associated with intramuscular fat deposition in Ujumqin sheep. Front Vet Sci 2025; 12:1559727. [PMID: 40177664 PMCID: PMC11963774 DOI: 10.3389/fvets.2025.1559727] [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: 01/13/2025] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction The molecular mechanisms underlying intramuscular fat (IMF) deposition are crucial for enhancing lamb meat quality. This process is regulated by a network of transcription factors. Exploring the role of non-coding RNAs, particularly lncRNAs and miRNAs, in IMF deposition can clarify its complex genetics and offer resources for breeding Inner Mongolian local breeds. Methods We evaluated carcass and lamb meat quality parameters using 60 six-month-old Ujumqin sheep with similar body weights. To investigate non-coding RNA's role in IMF deposition, we identified differentially expressed genes and pathways between the longissimus dorsi and femoral biceps. Additionally, we analyzed these genes and the lncRNA-miRNA-mRNA co-regulatory network in high- and low-IMF femoral biceps groups. Results We identified 11,529 mRNAs (747 differentially expressed), 9,874 lncRNAs (1,428 differentially expressed), and 761 miRNAs (12 differentially expressed). GO and KEGG enrichment analyses showed these genes are involved in lipid metabolism, fatty acid oxidation, and energy metabolism. We constructed a ceRNA network with 12 lncRNAs, 4 miRNAs, and 6 mRNAs. Notably, lncRNA MSTRG.13155.1 interacts with miR-1343-3p_R + 2, promoting IMF deposition by releasing HADHA gene expression. Dual-luciferase reporter assays confirmed MSTRG.13155.1 and HADHA as miR-1343-3p_R + 2 targets. RT-qPCR validated the expression trends of key mRNAs, miRNAs, and lncRNAs, consistent with sequencing results. Discussion Our comprehensive analysis of differentially expressed genes and pathways in Ujumqin sheep's longissimus dorsi and femoral biceps, along with high- and low-IMF groups, has revealed the complex genetics of IMF deposition. This offers valuable resources for Inner Mongolian local breed selection. The interaction between lncRNA MSTRG.13155.1 and miR-1343-3p_R + 2, and their regulation of HADHA expression, provides new insights into IMF deposition mechanisms. Future research can explore these mechanisms' universality and specificity across different breeds and environments.
Collapse
Affiliation(s)
- Mingxi Lan
- Animal Science Department, Inner Mongolia Agricultural University, Hohhot, China
| | - Qing Qin
- Animal Science Department, Inner Mongolia Agricultural University, Hohhot, China
| | - Yuchun Xie
- Animal Science Department, Inner Mongolia Agricultural University, Hohhot, China
- College of Animal Science and Technology, Hebei Science and Technology Normal University, Qinhuangdao, Hebei, China
| | - Chongyan Zhang
- Animal Science Department, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Sheep & Goat Genetics, Breeding and Reproduction in Inner Mongolia, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Sheep & Goat Genetics and Breeding of Ministry of Agriculture Inner Mongolia Agricultural University, Hohhot, China
| | - Zhichen Liu
- Animal Science Department, Inner Mongolia Agricultural University, Hohhot, China
| | - Xiaolong Xu
- Animal Science Department, Inner Mongolia Agricultural University, Hohhot, China
| | - Jingwen Zhang
- Animal Science Department, Inner Mongolia Agricultural University, Hohhot, China
| | - Songsong Xu
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ji Yang
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Haijun Zhang
- Erdos Agricultural and Animal Husbandry Science Research Institute, Ordos, China
| | - Suhe Alatan
- East Ujumqin Banner Hishig Animal Husbandry Development Co., Ltd., East Ujumqin Banner, China
| | - Zhixin Wang
- Animal Science Department, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Sheep & Goat Genetics, Breeding and Reproduction in Inner Mongolia, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Sheep & Goat Genetics and Breeding of Ministry of Agriculture Inner Mongolia Agricultural University, Hohhot, China
| | - Zhihong Liu
- Animal Science Department, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Sheep & Goat Genetics, Breeding and Reproduction in Inner Mongolia, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Sheep & Goat Genetics and Breeding of Ministry of Agriculture Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
5
|
Kang X, Zhao K, Huang Z, Fukada SI, Qi XW, Miao H. Pdgfrα + stromal cells, a key regulator for tissue homeostasis and dysfunction in distinct organs. Genes Dis 2025; 12:101264. [PMID: 39759120 PMCID: PMC11696774 DOI: 10.1016/j.gendis.2024.101264] [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: 08/14/2023] [Revised: 01/24/2024] [Accepted: 02/21/2024] [Indexed: 01/07/2025] Open
Abstract
Pdgfrα+ stromal cells are a group of cells specifically expressing Pdgfrα, which may be mentioned with distinct names in different tissues. Importantly, the findings from numerous studies suggest that these cells share exactly similar biomarkers and properties, show complex functions in regulating the microenvironment, and are critical to tissue regeneration, repair, and degeneration. Comparing the similarities and differences between distinct tissue-resident Pdgfrα+ stromal cells is helpful for us to more comprehensively and deeply understand the behaviors of these cells and to explore some common regulating mechanisms and therapeutical targets. In this review, we summarize previous and current findings on Pdgfrα+ stromal cells in various tissues and discuss the crosstalk between Pdgfrα+ stromal cells and microenvironment.
Collapse
Affiliation(s)
- Xia Kang
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, Sichuan 610000, China
| | - Kun Zhao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
| | - Zhu Huang
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, Sichuan 610000, China
| | - So-ichiro Fukada
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 564-0871, Japan
| | - Xiao-wei Qi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Hongming Miao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
- Jinfeng Laboratory, Chongqing 401329, China
| |
Collapse
|
6
|
Sun Z, Cheng X, Wang Z, Qiao C, Qian H, Yuan T, Lv Z, Sun W, Zhang H, Liu Y, Lu Z, Lin J, Lai C, Wang Y, Yang X, Wang X, Meng J, Bao N. Single-nucleus transcriptomics reveals subsets of degenerative myonuclei after rotator cuff tear-induced muscle atrophy. Cell Prolif 2025; 58:e13763. [PMID: 39435630 PMCID: PMC11882757 DOI: 10.1111/cpr.13763] [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: 03/21/2024] [Revised: 09/06/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
Rotator cuff tear (RCT) is the primary cause of shoulder pain and disability and frequently trigger muscle degeneration characterised by muscle atrophy, fatty infiltration and fibrosis. Single-nucleus RNA sequencing (snRNA-seq) was used to reveal the transcriptional changes in the supraspinatus muscle after RCT. Supraspinatus muscles were obtained from patients with habitual shoulder dislocation (n = 3) and RCT (n = 3). In response to the RCT, trajectory analysis showed progression from normal myonuclei to ANKRD1+ myonuclei, which captured atrophy-and fatty infiltration-related regulons (KLF5, KLF10, FOSL1 and BHLHE40). Transcriptomic alterations in fibro/adipogenic progenitors (FAPs) and muscle satellite cells (MuSCs) have also been studied. By predicting cell-cell interactions, we observed communication alterations between myofibers and muscle-resident cells following RCT. Our findings reveal the plasticity of muscle cells in response to RCT and offer valuable insights into the molecular mechanisms and potential therapeutic targets of RCT.
Collapse
Affiliation(s)
- Ziying Sun
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Xi Cheng
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Zheng Wang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Chenfeng Qiao
- Department of Orthopedics, Jinling Clinical Medical CollegeNanjing University of Chinese MedicineNanjingJiangsuPeople's Republic of China
| | - Hong Qian
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Tao Yuan
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Zhongyang Lv
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Wenshuang Sun
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Hanwen Zhang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Yuan Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Zhihao Lu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Jintao Lin
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Chengteng Lai
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Yang Wang
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Xiaojiang Yang
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Xingyun Wang
- Hongqiao International Institute of Medicine, Tongren HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Jia Meng
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- Department of Orthopedics, Jinling Clinical Medical CollegeNanjing University of Chinese MedicineNanjingJiangsuPeople's Republic of China
| | - Nirong Bao
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- Department of Orthopedics, Jinling Clinical Medical CollegeNanjing University of Chinese MedicineNanjingJiangsuPeople's Republic of China
| |
Collapse
|
7
|
Wu Z, Chen N, Takao D. The role of primary cilia in myoblast proliferation and cell cycle regulation during myogenesis. Cell Struct Funct 2025; 50:53-63. [PMID: 39805615 DOI: 10.1247/csf.24067] [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: 01/16/2025] Open
Abstract
The process of mammalian myogenesis is fundamental to understanding muscle development and holds broad relevance across multiple fields, from developmental biology to regenerative medicine. This review highlights two key aspects: myoblast proliferation and the role of cilia in this process. Myoblasts, as muscle precursor cells, must undergo tightly regulated cycles of proliferation and differentiation to ensure proper muscle growth and function. Recent research has uncovered an essential role for primary cilia, hair-like sensory organelles on the cell surface, in modulating signaling pathways crucial to myogenesis. Cilium-mediated signaling appears to regulate various stages of myogenesis, including the control of myoblast differentiation. Furthermore, primary cilia undergo multiple cycles of formation and disassembly during myogenesis, presumably enabling detailed, context-dependent regulation of their functions. In particular, the regulation of myoblast proliferation through cell cycle control by primary cilia is an important topic that requires further investigation. By examining the interactions between primary cilia and myoblasts, this review aims to provide new insights into the molecular and cellular mechanisms driving muscle development, with potential applications for understanding muscle-related diseases and advancing therapeutic strategies. Additionally, advancements in imaging and image analysis technologies have become indispensable for studying these processes at the cellular level. This review also addresses these technological advancements and current challenges.Key words: myogenesis, myoblast, proliferation, cilia, imaging.
Collapse
Affiliation(s)
- Zhichao Wu
- College of Animal Sciences and Technology and College of Veterinary Medicine, Huazhong Agricultural University
| | - Nuo Chen
- College of Animal Sciences and Technology and College of Veterinary Medicine, Huazhong Agricultural University
| | - Daisuke Takao
- College of Animal Sciences and Technology and College of Veterinary Medicine, Huazhong Agricultural University
- Hubei Hongshan Labolatory
| |
Collapse
|
8
|
Owen AM, Gonzalez-Velez S, Keeble AR, Thomas NT, Fry CS. Fork in the road: therapeutic and pathological actions for fibro-adipogenic progenitors following musculoskeletal injury. J Physiol 2025. [PMID: 39930980 DOI: 10.1113/jp286816] [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: 09/30/2024] [Accepted: 01/20/2025] [Indexed: 02/19/2025] Open
Abstract
Musculoskeletal injuries are a substantial source of global disability through weakness and loss of function, which can be attributable, in part, to deficits in skeletal muscle quality. Poor muscle quality, resulting from fibrotic pathology or fatty infiltration, strongly predicts lower rates of patient recovery following injury and higher rates of re-injury. The cellular sources of fibrosis and fatty infiltration within skeletal muscle are mesenchymal fibro-adipogenic progenitors (FAPs), which are central effectors to support muscle homeostasis, regeneration and growth. However, following acute or chronic musculoskeletal injury, FAPs can promote fibro/fatty pathology within muscle that is likely to limit recovery and repair. Given their indispensable role within skeletal muscle, FAPs have emerged as a compelling cellular target to promote tissue recovery following acute and chronic injury. This review provides insight into the aetiology of FAP activity following various musculoskeletal injuries, in addition to signalling components that effect FAP differentiation. Contrasting pathology with therapeutic potential, insight into disease- and injury-specific FAP activation further cements their role as crucial effectors to improve muscle function and enhance patient outcomes.
Collapse
Affiliation(s)
- Allison M Owen
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Sara Gonzalez-Velez
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Alexander R Keeble
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Nicholas T Thomas
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA
- Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Christopher S Fry
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA
- Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
9
|
Neyroud D, D'Lugos A, Trevino E, Callaway C, Lamm J, Laitano O, Poole B, Deyhle M, Brantley J, Le L, Judge A, Judge S. Local Inflammation Precedes Diaphragm Wasting and Fibrotic Remodelling in a Mouse Model of Pancreatic Cancer. J Cachexia Sarcopenia Muscle 2025; 16:e13668. [PMID: 39810606 PMCID: PMC11733308 DOI: 10.1002/jcsm.13668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/16/2024] [Accepted: 11/06/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Cancer cachexia represents a debilitating muscle wasting condition that is highly prevalent in gastrointestinal cancers, including pancreatic ductal adenocarcinoma (PDAC). Cachexia is estimated to contribute to ~30% of cancer-related deaths, with deterioration of respiratory muscles suspected to be a key contributor to cachexia-associated morbidity and mortality. In recent studies, we identified fibrotic remodelling of respiratory accessory muscles as a key feature of human PDAC cachexia. METHODS To gain insight into mechanisms driving respiratory muscle wasting and fibrotic remodelling in response to PDAC, we conducted temporal histological and transcriptomic analyses on diaphragm muscles harvested from mice-bearing orthotopic murine pancreatic (KPC) tumours at time points reflective of precachexia (D8 and D10), mild-moderate cachexia (D12 and D14) and advanced cachexia (endpoint). RESULTS During the precachexia phase, diaphragms showed significant leukocyte infiltration (+3-fold to +13-fold; D8-endpoint vs. Sham, p < 0.05) and transcriptomic enrichment of inflammatory processes associated with tissue injury that remained increased through endpoint. Diaphragm inflammation was followed by increases in PDGFR-ɑ+ fibroadipogenic progenitors (+2.5 to +3.8-fold; D10-endpoint vs. Sham, p < 0.05), fibre atrophy (-16% to -24%, D12 to endpoint vs. Sham, p < 0.05), ECM expansion (+1.5 to +1.8-fold; D14-endpoint vs. Sham, p < 0.05), collagen accumulation (+3.8-fold; endpoint vs. Sham, p = 0.0013) and reductions in breathing frequency (-55%, p = 0.0074) and diaphragm excursion (-43%, p = 0.0006). These biological processes were supported by changes in the diaphragm transcriptome. Ingenuity pathway analysis predicted factors involved in inflammatory responses to tissue injury, including TGF-β1, angiotensin and PDGF BB, as top upstream regulators activated in diaphragms prior to and throughout cachexia progression, while PGC-1α and the insulin receptor were among the top upstream regulators predicted to be suppressed. The transcriptomic dataset further revealed progressive disturbances to networks involved in lipid, glucose and oxidative metabolism, activation of the unfolded protein response and neuromuscular junction remodelling associated with denervation. CONCLUSIONS In summary, our data support leukocyte infiltration and expansion of PDGFRα mesenchymal progenitors as early events that precede wasting and fibrotic remodelling of the diaphragm in response to PDAC that may also underlie metabolic disturbances, weakness and respiratory complications.
Collapse
Affiliation(s)
- Daria Neyroud
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
- Myology InstituteUniversity of FloridaGainesvilleFloridaUSA
- Institute of Sports SciencesUniversity of LausanneLausanneSwitzerland
| | - Andrew C. D'Lugos
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
- Myology InstituteUniversity of FloridaGainesvilleFloridaUSA
| | - Enrique J. Trevino
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
| | - Chandler S. Callaway
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
- Myology InstituteUniversity of FloridaGainesvilleFloridaUSA
| | - Jacqueline Lamm
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
| | - Orlando Laitano
- Myology InstituteUniversity of FloridaGainesvilleFloridaUSA
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFloridaUSA
| | - Brittney Poole
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
- Myology InstituteUniversity of FloridaGainesvilleFloridaUSA
- Department of Physiology and Aging, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Michael R. Deyhle
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
- Myology InstituteUniversity of FloridaGainesvilleFloridaUSA
| | - Justina Brantley
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
| | - Lam Le
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
| | - Andrew R. Judge
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
- Myology InstituteUniversity of FloridaGainesvilleFloridaUSA
- Department of Physiology and Aging, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Sarah M. Judge
- Department of Physical TherapyUniversity of Florida Health Cancer CenterGainesvilleFloridaUSA
- Myology InstituteUniversity of FloridaGainesvilleFloridaUSA
- Department of Physiology and Aging, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
10
|
Fang F, Casserly M, Robbins J, Thomopoulos S. Hedgehog signaling directs cell differentiation and plays a critical role in tendon enthesis healing. NPJ Regen Med 2025; 10:3. [PMID: 39833191 PMCID: PMC11747568 DOI: 10.1038/s41536-025-00392-4] [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: 04/15/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
A high prevalence of rotator cuff tears presents a major clinical challenge. A better understanding of the molecular mechanisms underlying enthesis development and healing is needed for developing treatments. We recently identified hedgehog (Hh)-lineage cells critical for enthesis development and repair. This study revealed cell-cell communication within the Hh-lineage cell population. To further characterize the role of Hh signaling, we used mouse models to activate and inactivate the Hh pathway in enthesis progenitors. Activation of Hh target genes during enthesis development increased its mineralization and mechanical properties. Activation of Hh signaling at the injured mature enthesis promoted fibrocartilage formation, enhanced mineralization, and increased expression of chondrogenic and osteogenic markers, which implies that Hh signaling drives cell differentiation to regenerate the damaged enthesis. Conversely, deletion of Hh target genes impaired enthesis healing. In summary, this study revealed a new strategy for enthesis repair via activation of Hh signaling in endogenous cells.
Collapse
Affiliation(s)
- Fei Fang
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Matthew Casserly
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julia Robbins
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stavros Thomopoulos
- Department of Orthopedic Surgery, Columbia University, New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| |
Collapse
|
11
|
Chen Z, Wu X, Zheng D, Wang Y, Chai J, Zhang T, Wu P, Wei M, Zhou T, Long K, Li M, Jin L, Chen L. Single-Nucleus RNA Sequencing Reveals Cellular Transcriptome Features at Different Growth Stages in Porcine Skeletal Muscle. Cells 2025; 14:37. [PMID: 39791738 PMCID: PMC11720419 DOI: 10.3390/cells14010037] [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: 12/11/2024] [Revised: 12/29/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025] Open
Abstract
Porcine latissimus dorsi muscle (LDM) is a crucial source of pork products. Meat quality indicators, such as the proportion of muscle fibers and intramuscular fat (IMF) deposition, vary during the growth and development of pigs. Numerous studies have highlighted the heterogeneous nature of skeletal muscle, with phenotypic differences reflecting variations in cellular composition and transcriptional profiles. This study investigates the cellular-level transcriptional characteristics of LDM in large white pigs at two growth stages (170 days vs. 245 days) using single-nucleus RNA sequencing (snRNA-seq). We identified 56,072 cells across 12 clusters, including myofibers, fibro/adipogenic progenitor (FAP) cells, muscle satellite cells (MUSCs), and other resident cell types. The same cell types were present in the LDM at both growth stages, but their proportions and states differed. A higher proportion of FAPs was observed in the skeletal muscle of 245-day-old pigs. Additionally, these cells exhibited more active communication with other cell types compared to 170-day-old pigs. For instance, more interactions were found between FAPs and pericytes or endothelial cells in 245-day-old pigs, including collagen and integrin family signaling. Three subclasses of FAPs was identified, comprising FAPs_COL3A1+, FAPs_PDE4D+, and FAPs_EBF1+, while adipocytes were categorized into Ad_PDE4D+ and Ad_DGAT2+ subclasses. The proportions of these subclasses differed between the two age groups. We also constructed differentiation trajectories for FAPs and adipocytes, revealing that FAPs in 245-day-old pigs differentiated more toward fibrosis, a characteristic reminiscent of the high prevalence of skeletal muscle fibrosis in aging humans. Furthermore, the Ad_PDE4D+ adipocyte subclass, predominant in 245-day-old pigs, originated from FAPs_PDE4D+ expressing the same gene, while the Ad_DGAT2+ subclass stemmed from FAPs_EBF1+. In conclusion, our study elucidates transcriptional differences in skeletal muscle between two growth stages of pigs and provides insights into mechanisms relevant to pork meat quality and skeletal muscle diseases.
Collapse
Affiliation(s)
- Ziyu Chen
- Chongqing Academy of Animal Science, Chongqing 402460, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 625041, China
| | - Xiaoqian Wu
- Chongqing Academy of Animal Science, Chongqing 402460, China
| | - Dongbin Zheng
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 625041, China
| | - Yuling Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 625041, China
| | - Jie Chai
- Chongqing Academy of Animal Science, Chongqing 402460, China
| | - Tinghuan Zhang
- Chongqing Academy of Animal Science, Chongqing 402460, China
| | - Pingxian Wu
- Chongqing Academy of Animal Science, Chongqing 402460, China
- National Center of Technology Innovation for Pigs, Chongqing 402460, China
| | - Minghong Wei
- National Center of Technology Innovation for Pigs, Chongqing 402460, China
| | - Ting Zhou
- Sichuan Litian Animal Husbandry Co., Ltd., Dazhou 635711, China
| | - Keren Long
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 625041, China
| | - Mingzhou Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 625041, China
| | - Long Jin
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 625041, China
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Chengdu 625041, China
| | - Li Chen
- Chongqing Academy of Animal Science, Chongqing 402460, China
- Key Laboratory of Animal Resource Evaluation and Utilization (Pigs), Ministry of Agriculture and Rural Affairs, Chongqing 402460, China
| |
Collapse
|
12
|
Badralmaa Y, Natarajan V. Aberrant Wnt/β-catenin signaling in the mesenchymal stem cells of LZTFL1-depleted mice leads to increased adipogenesis, with implications for obesity. J Biol Chem 2025; 301:108057. [PMID: 39662832 PMCID: PMC11770550 DOI: 10.1016/j.jbc.2024.108057] [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: 03/26/2024] [Revised: 11/05/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024] Open
Abstract
Obesity is one of the main clinical characteristics associated with the heterogeneous genetic disorder Bardet-Biedl syndrome (BBS). Leucine zipper transcription factor like 1 (LZTFL1) is a member of the BBS gene family. Our work showed that Lztfl1knockout (LZKO) mice display the obesity phenotype as early as 3 months of age. Mesenchymal stem cells (MSCs) are multipotent stem cells that can differentiate into various cell types, including adipocytes. To understand the role of LZTFL1 in adipogenesis, we analyzed MSCs isolated from LZKO mouse compact bones (CB-MSCs). Compared to wildtype (WT), LZKO CB-MSCs had elongated primary cilia with tapered tips and increased levels of peroxisome proliferator-activated receptor γ (PPARγ), a key transcription factor that favors adipogenesis, and nuclear glucocorticoid receptor (GR), a transcription factor involved in Pparg activation. Also, LZKO CB-MSCs had a lower level of total β-catenin, a core factor of the antiadipogenic canonical Wnt/b-catenin signaling pathway involved in limiting the nuclear localization of GR. Interaction between caveolin1 (CAV1) and LRP6, the main receptor for canonical Wnt signaling, is known to be critical for Wnt pathway activation and β-catenin stabilization. Compared to WT cells, LZKO cells had elevated total, cell-surface, and lipid-raft-associated LRP6 and reduced CAV1, strongly indicating alterations in the components of the Wnt-signaling pathway. We show that in the absence of LZTFL1, adipogenesis-restraining Wnt/β-catenin signaling is inhibited, and adipogenesis-favorable factors are stimulated in CB-MSCs, leading to enhanced adipogenesis. Evidence provided here could help in understanding the mechanism and molecular basis of obesity in LZTFL1-defective patients.
Collapse
Affiliation(s)
- Yunden Badralmaa
- Laboratory of Molecular Cell Biology, Leidos Biomedical Research Inc, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Ven Natarajan
- Laboratory of Molecular Cell Biology, Leidos Biomedical Research Inc, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
| |
Collapse
|
13
|
Wang T, Zhou D, Hong Z. Sarcopenia and cachexia: molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2025; 6:e70030. [PMID: 39764565 PMCID: PMC11702502 DOI: 10.1002/mco2.70030] [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: 07/16/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 03/17/2025] Open
Abstract
Sarcopenia is defined as a muscle-wasting syndrome that occurs with accelerated aging, while cachexia is a severe wasting syndrome associated with conditions such as cancer and immunodeficiency disorders, which cannot be fully addressed through conventional nutritional supplementation. Sarcopenia can be considered a component of cachexia, with the bidirectional interplay between adipose tissue and skeletal muscle potentially serving as a molecular mechanism for both conditions. However, the underlying mechanisms differ. Recognizing the interplay and distinctions between these disorders is essential for advancing both basic and translational research in this area, enhancing diagnostic accuracy and ultimately achieving effective therapeutic solutions for affected patients. This review discusses the muscle microenvironment's changes contributing to these conditions, recent therapeutic approaches like lifestyle modifications, small molecules, and nutritional interventions, and emerging strategies such as gene editing, stem cell therapy, and gut microbiome modulation. We also address the challenges and opportunities of multimodal interventions, aiming to provide insights into the pathogenesis and molecular mechanisms of sarcopenia and cachexia, ultimately aiding in innovative strategy development and improved treatments.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Dong Zhou
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Zhen Hong
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| |
Collapse
|
14
|
Hogarth MW, Kurukunda MP, Ismat K, Uapinyoying P, Jaiswal JK. Exploring the therapeutic potential of fibroadipogenic progenitors in muscle disease. J Neuromuscul Dis 2025; 12:22143602241298545. [PMID: 39973455 PMCID: PMC11949306 DOI: 10.1177/22143602241298545] [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: 02/21/2025]
Abstract
Skeletal muscle relies on its inherent self-repair ability to withstand continuous mechanical damage. Myofiber-intrinsic processes facilitate the repair of damage to sarcolemma and sarcomeres, but it is the coordinated interaction between muscle-resident satellite and stromal cells that are crucial in the regeneration of muscles to replace the lost muscle fibers. Fibroadipogenic progenitors (FAPs), are muscle-resident mesenchymal cells that are notable for their role in creating the dynamic stromal niche required to support long-term muscle homeostasis and regeneration. While FAP-mediated extracellular matrix formation and the establishment of a homeostatic muscle niche are essential for maintaining muscle health, excessive accumulation of FAPs and their aberrant differentiation leads to the fibrofatty degeneration that is a hallmark of myopathies and muscular dystrophies. Recent advancements, including single-cell RNA sequencing and in vivo analysis of FAPs, are providing deeper insights into the functions and specialization of FAPs, shedding light on their roles in both health and disease. This review will explore the above insights, discussing how FAP dysregulation contributes to muscle diseases. It will offer a concise overview of potential therapeutic interventions targeting FAPs to restore disrupted interactions among FAPs and muscle-resident cells, ultimately addressing degenerative muscle loss in neuromuscular diseases.
Collapse
Affiliation(s)
- Marshall W Hogarth
- Center for Genetic Medicine Research, Children’s National Research Institute, Washington, DC, U.S.A
| | - Medha P Kurukunda
- Center for Genetic Medicine Research, Children’s National Research Institute, Washington, DC, U.S.A
| | - Karim Ismat
- Center for Genetic Medicine Research, Children’s National Research Institute, Washington, DC, U.S.A
| | - Prech Uapinyoying
- Center for Genetic Medicine Research, Children’s National Research Institute, Washington, DC, U.S.A
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, U.S.A
| | - Jyoti K Jaiswal
- Center for Genetic Medicine Research, Children’s National Research Institute, Washington, DC, U.S.A
- Department of Genomics and Precision Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC, U.S.A
| |
Collapse
|
15
|
Gorski T, Casartelli NC, Fitzgerald G, Horstman AMH, Masschelein E, Essers KJ, Maffiuletti NA, Sutter R, Leunig M, De Bock K. Intramuscular fatty infiltration and its correlation with muscle composition and function in hip osteoarthritis. Skelet Muscle 2024; 14:32. [PMID: 39696460 DOI: 10.1186/s13395-024-00364-0] [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: 05/14/2024] [Accepted: 11/24/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Hip osteoarthritis patients display higher levels of fatty infiltration (FI) in the gluteus minimus (GM) compared to other hip muscles. We investigated specific histological factors such as fiber type composition and collagen deposition, and functional outcomes like muscle strength and activation associated with FI in these patients. METHODS In twelve men (67 ± 6 y) undergoing total hip replacement (THR), hip and knee muscle strength and activation (electromyography, EMG) were assessed bilaterally. Magnetic resonance imaging (MRI) was used to compare the relative FI area and muscle cross sectional area (CSA) of the GM, rectus femoris (RF), tensor fascia latae (TFL) and vastus lateralis (VL). Adipocyte content, fiber type composition, grouping, fiber size, centrally nucleated fiber frequency, collagen deposition, satellite cell density and capillarization were assessed in intraoperative biopsies of the four muscles. Differences between GM and other muscles were assessed with repeated-measures one-way ANOVA followed by Dunnett's post-hoc test. Pearson coefficients were calculated for the correlations between FI measurements and the other histological and functional parameters. RESULTS Strength was lower in the affected limb. Knee extensor weakness was accompanied by lower VL muscle activation. Muscle CSA and FI did not differ between sides. In the affected limb, GM displayed larger relative FI area (MRI) compared to RF and VL. Biopsy adipocyte content was higher in GM than RF and TFL. Compared to the other hip muscles, GM displayed higher type 1 fiber content while its type 2X fiber content was lower. Fiber grouping levels were higher in GM than the other muscles. Collagen content was higher in GM than TFL and VL. FI in GM was associated with type 1 (r = 0.43) and type 2X (r = -0.34) fiber content, fiber grouping (r = 0.39), and collagen deposition (r = 0.37). FI in VL was negatively associated with maximal knee extension strength (r = -0.65). CONCLUSIONS In patients undergoing THR, the higher FI levels of GM compared to other hip muscles were associated with fiber type composition and grouping, and with higher collagen deposition. Experimental studies exploring these associations could potentially uncover new targets for the treatment of intramuscular FI and related impairments in muscle function. TRIAL REGISTRATION KEK number: 2016-01852, date of registration: 12-4-2017.
Collapse
MESH Headings
- Humans
- Male
- Aged
- Osteoarthritis, Hip/pathology
- Osteoarthritis, Hip/surgery
- Osteoarthritis, Hip/physiopathology
- Osteoarthritis, Hip/metabolism
- Osteoarthritis, Hip/diagnostic imaging
- Middle Aged
- Adipose Tissue/pathology
- Adipose Tissue/metabolism
- Adipose Tissue/diagnostic imaging
- Muscle, Skeletal/pathology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/physiopathology
- Muscle Strength
- Magnetic Resonance Imaging/methods
- Electromyography
- Arthroplasty, Replacement, Hip
Collapse
Affiliation(s)
- Tatiane Gorski
- Laboratory of Exercise and Health, ETH Zurich, Schwerzenbach, Switzerland
- Cytometry Facility, University of Zurich, Zurich, Switzerland
| | - Nicola C Casartelli
- Laboratory of Exercise and Health, ETH Zurich, Schwerzenbach, Switzerland
- Human Performance Lab, Schulthess Clinic, Zurich, Switzerland
| | - Gillian Fitzgerald
- Laboratory of Exercise and Health, ETH Zurich, Schwerzenbach, Switzerland
| | | | - Evi Masschelein
- Laboratory of Exercise and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Kalliopi J Essers
- Laboratory of Exercise and Health, ETH Zurich, Schwerzenbach, Switzerland
| | | | - Reto Sutter
- Department of Radiology, University Hospital Balgrist, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Michael Leunig
- Department of Orthopaedic Surgery, Schulthess Clinic, Zurich, Switzerland
| | - Katrien De Bock
- Laboratory of Exercise and Health, ETH Zurich, Schwerzenbach, Switzerland.
- Laboratory of Exercise and Health, Institute of Movement Sciences (D-HEST), ETH Zürich - Swiss Federal Institute of Technology, Schorenstrasse 16, Schwerzenbach, CH-8603, Switzerland.
| |
Collapse
|
16
|
Jin L, Li K, Li Z, Huang X, Wang L, Wang X, Di S, Cui S, Xu Y. Investigation into Critical Gut Microbes Influencing Intramuscular Fat Deposition in Min Pigs. Animals (Basel) 2024; 14:3123. [PMID: 39518846 PMCID: PMC11545367 DOI: 10.3390/ani14213123] [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: 09/05/2024] [Revised: 10/19/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
To determine the pivotal microorganisms affecting intramuscular fat (IMF) accumulation in Min pigs and to discern the extent of the influence exerted by various intestinal segments on IMF-related traits, we sequenced 16S rRNA from the contents of six intestinal segments from a high IMF group (Group H) and a low IMF group (Group L) of Min pigs weighing 90 ± 1 kg. We then compared their diversity and disparities in bacterial genera. Group H exhibited considerably higher α diversity in the jejunum and colon than Group L (p < 0.05). When 95% confidence levels were considered, the main β diversity components for the ileum, caecum, and colon within Groups H and L exhibited absolute segregation. Accordingly, 31 differentially abundant genera across Group H were pinpointed via LEfSe and the Wilcoxon test (p < 0.05) and subsequently scrutinised based on their distribution and abundance across distinct intestinal segments and their correlation with IMF phenotypes. The abundances of Terrisporobacter, Acetitomaculum, Bacteroides, Fibrobacter, Treponema, Akkermansia, Blautia, Clostridium sensu stricto 1, Turicibacter, Subdoligranulum, the [Eubacterium] siraeum group, and dgA 11 gut groups were positively correlated with IMF content (p < 0.05), whereas those of Bacillus, the Lachnospiraceae NK4A136 group, Streptococcus, Roseburia, Solobacterium, Veillonella, Lactobacillus, the Rikenellaceae RC9 gut group, Anaerovibrio, and the Lachnospiraceae AC2044 group were negatively associated with IMF content (p < 0.05). Employing PICRUSt2 for predicting intergenic metabolic pathways that differ among intestinal microbial communities revealed that within the 95% confidence interval the colonic microbiome was enriched with the most metabolic pathways, including those related to lipid metabolism. The diversity results, bacterial genus distributions, and metabolic pathway disparities revealed the colonic segment as an influential region for IMF deposition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Shiquan Cui
- College of Animal Science and Technology, Northeast Agricultural University, No. 600 Changjiang Road, Xiangfang District, Harbin 150030, China; (L.J.); (K.L.); (Z.L.); (X.H.); (L.W.); (X.W.); (S.D.)
| | - Yuan Xu
- College of Animal Science and Technology, Northeast Agricultural University, No. 600 Changjiang Road, Xiangfang District, Harbin 150030, China; (L.J.); (K.L.); (Z.L.); (X.H.); (L.W.); (X.W.); (S.D.)
| |
Collapse
|
17
|
Jung U, Kim M, Voy BH. Fibroadipogenic progenitors: a potential target for preventing breast muscle myopathies in broilers. Front Physiol 2024; 15:1458151. [PMID: 39193441 PMCID: PMC11347355 DOI: 10.3389/fphys.2024.1458151] [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: 07/02/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
Genetic selection for high growth rate, breast muscle yield, and feed efficiency in modern broilers has been a double-edged sword. While it has resulted in marked benefits in production, it has also introduced widespread incidence of breast muscle myopathies. Broiler myopathies are phenotypically characterized by myodegeneration and fibrofatty infiltration, which compromise meat quality. These lesions resemble those of various myopathies found in humans, such as Duchenne muscular dystrophy, Limb-girdle muscular dystrophy, and sarcopenia. Fibroadipogenic progenitors (FAPs) are interstitial muscle-resident mesenchymal stem cells that are named because of their ability to differentiate into both fibroblasts and adipocytes. This cell population has clearly been established to play a role in the development and progression of myopathies in mice and humans. Gene expression studies of wooden breast and other related disorders have implicated FAPs in broilers, but to our knowledge this cell population have not been characterized in chickens. In this review, we summarize the evidence that FAPs may be a novel, new target for interventions that reduce the incidence and development of chicken breast muscle myopathies.
Collapse
Affiliation(s)
| | | | - Brynn H. Voy
- Department of Animal Science, University of Tennessee, Knoxville, TN, United States
| |
Collapse
|
18
|
He K, Jiang H, Li W, Toutounchi S, Huang Y, Wu J, Ma X, Baehr W, Pignolo RJ, Ling K, Zhou X, Wang H, Hu J. Primary cilia mediate skeletogenic BMP and Hedgehog signaling in heterotopic ossification. Sci Transl Med 2024; 16:eabn3486. [PMID: 39047114 DOI: 10.1126/scitranslmed.abn3486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/04/2024] [Accepted: 07/02/2024] [Indexed: 07/27/2024]
Abstract
Heterotopic ossification (HO), defined as the formation of extraskeletal bone in muscle and soft tissues, is a diverse pathological process caused by either genetic mutations or inciting trauma. Fibrodysplasia ossificans progressiva (FOP) is a genetic form of HO caused by mutations in the bone morphogenetic protein (BMP) type I receptor gene activin A receptor type 1 (ACVR1). These mutations make ACVR1 hypersensitive to BMP and responsive to activin A. Hedgehog (Hh) signaling also contributes to HO development. However, the exact pathophysiology of how skeletogenic cells contribute to endochondral ossification in FOP remains unknown. Here, we showed that the wild-type or FOP-mutant ACVR1 localized in the cilia of stem cells from human exfoliated deciduous teeth with key FOP signaling components, including activin A receptor type 2A/2B, SMAD family member 1/5, and FK506-binding protein 12kD. Cilia suppression by deletion of intraflagellar transport 88 or ADP ribosylation factor like GTPase 3 effectively inhibited pathological BMP and Hh signaling, subdued aberrant chondro-osteogenic differentiation in primary mouse or human FOP cells, and diminished in vivo extraskeletal ossification in Acvr1Q207D, Sox2-Cre; Acvr1R206H/+ FOP mice and in burn tenotomy-treated wild-type mice. Our results provide a rationale for early and localized suppression of cilia in affected tissues after injury as a therapeutic strategy against either genetic or acquired HO.
Collapse
Affiliation(s)
- Kai He
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905 USA
| | - Heng Jiang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905 USA
| | - Weijun Li
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905 USA
- Mayo Clinic Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905 USA
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Mayo Clinic, Rochester, MN 55905 USA
| | - Saman Toutounchi
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905 USA
- Mayo Clinic Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905 USA
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Mayo Clinic, Rochester, MN 55905 USA
| | - Yan Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905 USA
| | - Jianfeng Wu
- Department of Orthopedics, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Xiaoyu Ma
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905 USA
| | - Wolfgang Baehr
- Department of Ophthalmology, University of Utah, Salt Lake City, UT 84132, USA
| | - Robert J Pignolo
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905 USA
- Mayo Clinic Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905 USA
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Mayo Clinic, Rochester, MN 55905 USA
| | - Kun Ling
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905 USA
| | - Xuhui Zhou
- Translational Research Center of Orthopedics, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Haitao Wang
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905 USA
- Mayo Clinic Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905 USA
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Mayo Clinic, Rochester, MN 55905 USA
| | - Jinghua Hu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905 USA
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
- Mayo Clinic Robert M. and Billie Kelley Pirnie Translational Polycystic Kidney Disease Center, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
19
|
Fernández-Simón E, Piñol-Jurado P, Gokul-Nath R, Unsworth A, Alonso-Pérez J, Schiava M, Nascimento A, Tasca G, Queen R, Cox D, Suarez-Calvet X, Díaz-Manera J. Single cell RNA sequencing of human FAPs reveals different functional stages in Duchenne muscular dystrophy. Front Cell Dev Biol 2024; 12:1399319. [PMID: 39045456 PMCID: PMC11264872 DOI: 10.3389/fcell.2024.1399319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 06/03/2024] [Indexed: 07/25/2024] Open
Abstract
Background: Duchenne muscular dystrophy is a genetic disease produced by mutations in the dystrophin gene characterized by early onset muscle weakness leading to severe and irreversible disability. Muscle degeneration involves a complex interplay between multiple cell lineages spatially located within areas of damage, termed the degenerative niche, including inflammatory cells, satellite cells (SCs) and fibro-adipogenic precursor cells (FAPs). FAPs are mesenchymal stem cell which have a pivotal role in muscle homeostasis as they can either promote muscle regeneration or contribute to muscle degeneration by expanding fibrotic and fatty tissue. Although it has been described that FAPs could have a different behavior in DMD patients than in healthy controls, the molecular pathways regulating their function as well as their gene expression profile are unknown. Methods: We used single-cell RNA sequencing (scRNAseq) with 10X Genomics and Illumina technology to elucidate the differences in the transcriptional profile of isolated FAPs from healthy and DMD patients. Results: Gene signatures in FAPs from both groups revealed transcriptional differences. Seurat analysis categorized cell clusters as proliferative FAPs, regulatory FAPs, inflammatory FAPs, and myofibroblasts. Differentially expressed genes (DEGs) between healthy and DMD FAPs included upregulated genes CHI3L1, EFEMP1, MFAP5, and TGFBR2 in DMD. Functional analysis highlighted distinctions in system development, wound healing, and cytoskeletal organization in control FAPs, while extracellular organization, degradation, and collagen degradation were upregulated in DMD FAPs. Validation of DEGs in additional samples (n = 9) using qPCR reinforced the specific impact of pathological settings on FAP heterogeneity, reflecting their distinct contribution to fibro or fatty degeneration in vivo. Conclusion: Using the single-cell RNA seq from human samples provide new opportunities to study cellular coordination to further understand the regulation of muscle homeostasis and degeneration that occurs in muscular dystrophies.
Collapse
Affiliation(s)
- Esther Fernández-Simón
- John Walton Muscular Dystrophy Research Centre, Newcastle University Translational and Clinical Research Institute and Newcastle Hospitals NHS Foundation Trust, NE1 3BZ, Newcastle Upon Tyne, United Kingdom
| | - Patricia Piñol-Jurado
- John Walton Muscular Dystrophy Research Centre, Newcastle University Translational and Clinical Research Institute and Newcastle Hospitals NHS Foundation Trust, NE1 3BZ, Newcastle Upon Tyne, United Kingdom
| | - Rasya Gokul-Nath
- John Walton Muscular Dystrophy Research Centre, Newcastle University Translational and Clinical Research Institute and Newcastle Hospitals NHS Foundation Trust, NE1 3BZ, Newcastle Upon Tyne, United Kingdom
| | - Adrienne Unsworth
- John Walton Muscular Dystrophy Research Centre, Newcastle University Translational and Clinical Research Institute and Newcastle Hospitals NHS Foundation Trust, NE1 3BZ, Newcastle Upon Tyne, United Kingdom
| | - Jorge Alonso-Pérez
- Bioinformatics Unit, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Marianela Schiava
- John Walton Muscular Dystrophy Research Centre, Newcastle University Translational and Clinical Research Institute and Newcastle Hospitals NHS Foundation Trust, NE1 3BZ, Newcastle Upon Tyne, United Kingdom
| | - Andres Nascimento
- Neuromuscular Disorders Unit, Neurology Department, Hospital Sant Joan de Deu, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Madrid, Spain
| | - Giorgio Tasca
- John Walton Muscular Dystrophy Research Centre, Newcastle University Translational and Clinical Research Institute and Newcastle Hospitals NHS Foundation Trust, NE1 3BZ, Newcastle Upon Tyne, United Kingdom
| | - Rachel Queen
- John Walton Muscular Dystrophy Research Centre, Newcastle University Translational and Clinical Research Institute and Newcastle Hospitals NHS Foundation Trust, NE1 3BZ, Newcastle Upon Tyne, United Kingdom
| | - Dan Cox
- John Walton Muscular Dystrophy Research Centre, Newcastle University Translational and Clinical Research Institute and Newcastle Hospitals NHS Foundation Trust, NE1 3BZ, Newcastle Upon Tyne, United Kingdom
| | - Xavier Suarez-Calvet
- Neuromuscular Disorders Unit, Neurology Department, Insitut de Recerca de l’Hospital de la Santa Creu I Sant Pau, Barcelona, Spain
- Neuromuscular Disease Unit, Neurology Department, Hospital Universitario Nuestra Señora de Candelaria, Fundación Canaria Instituto de Investigación Sanitaria de Canarias (FIISC), Tenerife, Spain
| | - Jordi Díaz-Manera
- John Walton Muscular Dystrophy Research Centre, Newcastle University Translational and Clinical Research Institute and Newcastle Hospitals NHS Foundation Trust, NE1 3BZ, Newcastle Upon Tyne, United Kingdom
- Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Madrid, Spain
- Neuromuscular Disorders Unit, Neurology Department, Insitut de Recerca de l’Hospital de la Santa Creu I Sant Pau, Barcelona, Spain
| |
Collapse
|
20
|
Nishimura Y. [Regulation of adipose progenitors and fibro-adipogenic progenitors through primary cilia]. Nihon Yakurigaku Zasshi 2024; 159:188-191. [PMID: 38684399 DOI: 10.1254/fpj.23108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
The primary cilium, an antenna-like structure of cell membrane, detects various signals and regulates cellular functions such as proliferation and differentiation. The impairment of primary cilium is associated with the etiologies of diseases including cancer, obesity, and congenital anomalies. In this review, novel functions of trichoplein, a suppressor of ciliogenesis, on the regulation of adipose progenitors and fibro-adipogenic progenitors are focused. Trichoplein-knockout mice show resistance to high-fat diet-induced obesity and accelerated regeneration after skeletal muscle injury. The primary cilia of adipose progenitors from trichoplein-knockout mice are elongated, leading to the inhibitions of the accumulation of lipid raft to the base of primary cilia and the phosphorylation of AKT. The primary cilia of fibro-adipogenic progenitors from trichoplein-knockout mice are also elongated, causing the increased expression of IL-13 through IL-33 receptor signaling. These mechanisms are involved in the resistance to diet-induced obesity and improved regeneration. These findings suggest that targeting the primary cilia of specific cells may be a novel therapeutic approach through modulating cellular functions.
Collapse
Affiliation(s)
- Yuhei Nishimura
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine
- Research Center for Cilia and Diseases, Mie University Graduate School of Medicine
| |
Collapse
|
21
|
Yi X, Feng M, Zhu J, Yu H, He Z, Zhang Z, Zhao T, Zhang Q, Pang W. Adipocyte Progenitor Pools Composition and Cellular Niches Affect Adipogenesis Divergence in Porcine Subcutaneous and Intramuscular Fat. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 38848240 DOI: 10.1021/acs.jafc.4c01044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
Intramuscular fat (IMF) contributed positively to pork quality, whereas subcutaneous fat (SCF) was often considered to be a detrimental factor impacting growth and carcass traits. Reducing SCF while maintaining optimal IMF levels requires a thorough understanding of the adipogenic differences between these two adipose depots. Our study explored the differences in adipogenesis between porcine IMF and SCF, and the results showed that subcutaneous adipocytes (SCAs) demonstrate a greater potential for adipogenic differentiation, both in vivo and in vitro. Lipidomic and transcriptomic analyses suggested that intramuscular adipocytes (IMAs) are more inclined to biosynthesize unsaturated fatty acids. Furthermore, single-cell RNA sequencing (scRNA-seq) was employed to dissect the intrinsic and microenvironmental discrepancies in adipogenesis between porcine IMF and SCF. Comparative analysis indicated that SCF was enriched with preadipocytes, exhibiting an enhanced adipogenic potential, while IMF was characterized by a higher abundance of stem cells. Furthermore, coculture analyses of porcine intramuscular adipogenic cells and myogenetic cells indicated that the niche of IMAs inhibited its adipogenic differentiation. Cell communication analysis identified 160 ligand-receptor pairs and channels between adipogenic and myogenetic cells in IMF. Collectively, our study elucidated two intrinsic and microenvironmental novel mechanisms underpinning the divergence in adipogenesis between porcine SCF and IMF.
Collapse
Affiliation(s)
- Xudong Yi
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Ming Feng
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jiahua Zhu
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - He Yu
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Zhaozhao He
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Ziyi Zhang
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Tiantian Zhao
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Que Zhang
- Department of Animal Science and Technology, Shandong Vocational Animal Science and Veterinary College, Weifang, Shandong 261061, China
| | - Weijun Pang
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| |
Collapse
|
22
|
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
|
23
|
Zhao T, Tian T, Yu H, Cao C, Zhang Z, He Z, Ma Z, Cai R, Li F, Pang W. Identification of porcine fast/slow myogenic exosomes and their regulatory effects on lipid accumulation in intramuscular adipocytes. J Anim Sci Biotechnol 2024; 15:73. [PMID: 38824596 PMCID: PMC11144342 DOI: 10.1186/s40104-024-01029-0] [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: 11/27/2023] [Accepted: 04/01/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND Pork quality is affected by the type of muscle fibers, which is closely related to meat color, tenderness and juiciness. Exosomes are tiny vesicles with a diameter of approximately 30-150 nm that are secreted by cells and taken up by recipient cells to mediate communication. Exosome-mediated muscle-fat tissue crosstalk is a newly discovered mechanism that may have an important effect on intramuscular fat deposition and with that on meat quality. Various of adipose tissue-derived exosomes have been discovered and identified, but the identification and function of muscle exosomes, especially porcine fast/slow myotube exosomes, remain unclear. Here, we first isolated and identified exosomes secreted from porcine extensor digitorum longus (EDL) and soleus (SOL), which represent fast and slow muscle, respectively, and further explored their effects on lipid accumulation in longissimus dorsi adipocytes. RESULTS Porcine SOL-derived exosomes (SOL-EXO) and EDL-derived exosomes (EDL-EXO) were first identified and their average particle sizes were approximately 84 nm with double-membrane disc- shapes as observed via transmission electron microscopy and scanning electron microscopy. Moreover, the intramuscular fat content of the SOL was greater than that of the EDL at 180 days of age, because SOL intramuscular adipocytes had a stronger lipid-accumulating capacity than those of the EDL. Raman spectral analysis revealed that SOL-EXO protein content was much greater than that of EDL-EXO. Proteomic sequencing identified 72 proteins that were significantly differentially expressed between SOL-EXO and EDL-EXO, 31 of which were downregulated and 41 of which were upregulated in SOL-EXO. CONCLUSIONS Our findings suggest that muscle-fat tissue interactions occur partly via SOL-EXO promoting adipogenic activity of intramuscular adipocytes.
Collapse
Affiliation(s)
- Tiantian Zhao
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Tingting Tian
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - He Yu
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Chaoyue Cao
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Ziyi Zhang
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Zhaozhao He
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Zeqiang Ma
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Rui Cai
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Fengna Li
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China
| | - Weijun Pang
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
24
|
Flores-Opazo M, Kopinke D, Helmbacher F, Fernández-Verdejo R, Tuñón-Suárez M, Lynch GS, Contreras O. Fibro-adipogenic progenitors in physiological adipogenesis and intermuscular adipose tissue remodeling. Mol Aspects Med 2024; 97:101277. [PMID: 38788527 PMCID: PMC11692456 DOI: 10.1016/j.mam.2024.101277] [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: 02/01/2024] [Revised: 04/27/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
Excessive accumulation of intermuscular adipose tissue (IMAT) is a common pathological feature in various metabolic and health conditions and can cause muscle atrophy, reduced function, inflammation, insulin resistance, cardiovascular issues, and unhealthy aging. Although IMAT results from fat accumulation in muscle, the mechanisms underlying its onset, development, cellular components, and functions remain unclear. IMAT levels are influenced by several factors, such as changes in the tissue environment, muscle type and origin, extent and duration of trauma, and persistent activation of fibro-adipogenic progenitors (FAPs). FAPs are a diverse and transcriptionally heterogeneous population of stromal cells essential for tissue maintenance, neuromuscular stability, and tissue regeneration. However, in cases of chronic inflammation and pathological conditions, FAPs expand and differentiate into adipocytes, resulting in the development of abnormal and ectopic IMAT. This review discusses the role of FAPs in adipogenesis and how they remodel IMAT. It highlights evidence supporting FAPs and FAP-derived adipocytes as constituents of IMAT, emphasizing their significance in adipose tissue maintenance and development, as well as their involvement in metabolic disorders, chronic pathologies and diseases. We also investigated the intricate molecular pathways and cell interactions governing FAP behavior, adipogenesis, and IMAT accumulation in chronic diseases and muscle deconditioning. Finally, we hypothesize that impaired cellular metabolic flexibility in dysfunctional muscles impacts FAPs, leading to IMAT. A deeper understanding of the biology of IMAT accumulation and the mechanisms regulating FAP behavior and fate are essential for the development of new therapeutic strategies for several debilitating conditions.
Collapse
Affiliation(s)
| | - Daniel Kopinke
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, 32610, FL, USA; Myology Institute, University of Florida College of Medicine, Gainesville, FL, USA.
| | | | - Rodrigo Fernández-Verdejo
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA; Laboratorio de Fisiología Del Ejercicio y Metabolismo (LABFEM), Escuela de Kinesiología, Facultad de Medicina, Universidad Finis Terrae, Chile.
| | - Mauro Tuñón-Suárez
- Laboratorio de Fisiología Del Ejercicio y Metabolismo (LABFEM), Escuela de Kinesiología, Facultad de Medicina, Universidad Finis Terrae, Chile.
| | - Gordon S Lynch
- Centre for Muscle Research, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Parkville 3010, Australia.
| | - Osvaldo Contreras
- Developmental and Regenerative Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, 2010, Australia; School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia.
| |
Collapse
|
25
|
Norris AM, Fierman KE, Campbell J, Pitale R, Shahraj M, Kopinke D. Studying intramuscular fat deposition and muscle regeneration: insights from a comparative analysis of mouse strains, injury models, and sex differences. Skelet Muscle 2024; 14:12. [PMID: 38812056 PMCID: PMC11134715 DOI: 10.1186/s13395-024-00344-4] [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: 04/01/2024] [Accepted: 05/13/2024] [Indexed: 05/31/2024] Open
Abstract
Intramuscular fat (IMAT) infiltration, pathological adipose tissue that accumulates between muscle fibers, is a shared hallmark in a diverse set of diseases including muscular dystrophies and diabetes, spinal cord and rotator cuff injuries, as well as sarcopenia. While the mouse has been an invaluable preclinical model to study skeletal muscle diseases, they are also resistant to IMAT formation. To better understand this pathological feature, an adequate pre-clinical model that recapitulates human disease is necessary. To address this gap, we conducted a comprehensive in-depth comparison between three widely used mouse strains: C57BL/6J, 129S1/SvlmJ and CD1. We evaluated the impact of strain, sex and injury type on IMAT formation, myofiber regeneration and fibrosis. We confirm and extend previous findings that a Glycerol (GLY) injury causes significantly more IMAT and fibrosis compared to Cardiotoxin (CTX). Additionally, females form more IMAT than males after a GLY injury, independent of strain. Of all strains, C57BL/6J mice, both females and males, are the most resistant to IMAT formation. In regard to injury-induced fibrosis, we found that the 129S strain formed the least amount of scar tissue. Surprisingly, C57BL/6J of both sexes demonstrated complete myofiber regeneration, while both CD1 and 129S1/SvlmJ strains still displayed smaller myofibers 21 days post injury. In addition, our data indicate that myofiber regeneration is negatively correlated with IMAT and fibrosis. Combined, our results demonstrate that careful consideration and exploration are needed to determine which injury type, mouse model/strain and sex to utilize as preclinical model especially for modeling IMAT formation.
Collapse
Affiliation(s)
- Alessandra M Norris
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Kiara E Fierman
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Jillian Campbell
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Rhea Pitale
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Muhammad Shahraj
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Daniel Kopinke
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
26
|
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
|
27
|
Rota Graziosi E, François S, Nasser F, Gauthier M, Oger M, Favier AL, Drouet M, Jullien N, Riccobono D. Comparison of Three Antagonists of Hedgehog Pathway to Promote Skeletal Muscle Regeneration after High Dose Irradiation. Radiat Res 2024; 201:429-439. [PMID: 38253061 DOI: 10.1667/rade-23-00140.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/01/2023] [Indexed: 01/24/2024]
Abstract
The current geopolitical context has brought the radiological nuclear risk to the forefront of concerns. High-dose localized radiation exposure leads to the development of a musculocutaneous radiation syndrome affecting the skin and subcutaneous muscles. Despite the implementation of a gold standard treatment based on an invasive surgical procedure coupled with autologous cell therapy, a muscular defect frequently persists. Targeting the modulation of the Hedgehog (Hh) signaling pathway appears to be a promising therapeutic approach. Activation of this pathway enhances cell survival and promotes proliferation after irradiation, while inhibition by Cyclopamine facilitates differentiation. In this study, we compared the effects of three antagonists of Hh, Cyclopamine (CA), Vismodegib (VDG) and Sonidegib (SDG) on differentiation. A stable cell line of murine myoblasts, C2C12, was exposed to X-ray radiation (5 Gy) and treated with CA, VDG or SDG. Analysis of proliferation, survival (apoptosis), morphology, myogenesis genes expression and proteins production were performed. According to the results, VDG does not have a significant impact on C2C12 cells. SDG increases the expression/production of differentiation markers to a similar extent as CA, while morphologically, SDG proves to be more effective than CA. To conclude, SDG can be used in the same way as CA but already has a marketing authorization with an indication against basal cell cancers, facilitating their use in vivo. This proof of concept demonstrates that SDG represents a promising alternative to CA to promotes differentiation of murine myoblasts. Future studies on isolated and cultured satellite cells and in vivo will test this proof of concept.
Collapse
Affiliation(s)
- Emmanuelle Rota Graziosi
- IRBA, French Armed Forces Biomedical Research Institute, Radiobiology unit, Brétigny-sur-Orge, France
| | - Sabine François
- IRBA, French Armed Forces Biomedical Research Institute, Radiobiology unit, Brétigny-sur-Orge, France
- INSERM, UMR1296, Radiations: Defense, Health, Environment, Lyon and Brétigny-sur-Orge, France
| | - Farah Nasser
- IRBA, French Armed Forces Biomedical Research Institute, Radiobiology unit, Brétigny-sur-Orge, France
| | - Michel Gauthier
- IRBA, French Armed Forces Biomedical Research Institute, Radiobiology unit, Brétigny-sur-Orge, France
| | - Myriam Oger
- IRBA, French Armed Forces Biomedical Research Institute, Imagery Unit, Department of Platforms and Technology Research, Brétigny-sur-Orge, France
| | - Anne-Laure Favier
- IRBA, French Armed Forces Biomedical Research Institute, Imagery Unit, Department of Platforms and Technology Research, Brétigny-sur-Orge, France
| | - Michel Drouet
- INSERM, UMR1296, Radiations: Defense, Health, Environment, Lyon and Brétigny-sur-Orge, France
- IRBA, French Armed Forces Biomedical Research Institute, Radiations Bioeffects Department, Brétigny-sur-Orge, France
| | - Nicolas Jullien
- IRBA, French Armed Forces Biomedical Research Institute, Radiobiology unit, Brétigny-sur-Orge, France
| | - Diane Riccobono
- INSERM, UMR1296, Radiations: Defense, Health, Environment, Lyon and Brétigny-sur-Orge, France
- IRBA, French Armed Forces Biomedical Research Institute, Radiations Bioeffects Department, Brétigny-sur-Orge, France
| |
Collapse
|
28
|
Guo Q, Luo Q, Song G. Control of muscle satellite cell function by specific exercise-induced cytokines and their applications in muscle maintenance. J Cachexia Sarcopenia Muscle 2024; 15:466-476. [PMID: 38375571 PMCID: PMC10995279 DOI: 10.1002/jcsm.13440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 01/05/2024] [Accepted: 01/14/2024] [Indexed: 02/21/2024] Open
Abstract
Exercise is recognized to play an observable role in improving human health, especially in promoting muscle hypertrophy and intervening in muscle mass loss-related diseases, including sarcopenia. Recent rapid advances have demonstrated that exercise induces the release of abundant cytokines from several tissues (e.g., liver, muscle, and adipose tissue), and multiple cytokines improve the functions or expand the numbers of adult stem cells, providing candidate cytokines for alleviating a wide range of diseases. Muscle satellite cells (SCs) are a population of muscle stem cells that are mitotically quiescent but exit from the dormancy state to become activated in response to physical stimuli, after which SCs undergo asymmetric divisions to generate new SCs (stem cell pool maintenance) and commit to later differentiation into myocytes (skeletal muscle replenishment). SCs are essential for the postnatal growth, maintenance, and regeneration of skeletal muscle. Emerging evidence reveals that exercise regulates muscle function largely via the exercise-induced cytokines that govern SC potential, but this phenomenon is complicated and confusing. This review provides a comprehensive integrative overview of the identified exercise-induced cytokines and the roles of these cytokines in SC function, providing a more complete picture regarding the mechanism of SC homeostasis and rejuvenation therapies for skeletal muscle.
Collapse
Affiliation(s)
- Qian Guo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqingChina
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqingChina
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqingChina
| |
Collapse
|
29
|
Wang L, Valencak TG, Shan T. Fat infiltration in skeletal muscle: Influential triggers and regulatory mechanism. iScience 2024; 27:109221. [PMID: 38433917 PMCID: PMC10907799 DOI: 10.1016/j.isci.2024.109221] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024] Open
Abstract
Fat infiltration in skeletal muscle (also known as myosteatosis) is now recognized as a distinct disease from sarcopenia and is directly related to declining muscle capacity. Hence, understanding the origins and regulatory mechanisms of fat infiltration is vital for maintaining skeletal muscle development and improving human health. In this article, we summarized the triggering factors such as aging, metabolic diseases and metabolic syndromes, nonmetabolic diseases, and muscle injury that all induce fat infiltration in skeletal muscle. We discussed recent advances on the cellular origins of fat infiltration and found several cell types including myogenic cells and non-myogenic cells that contribute to myosteatosis. Furthermore, we reviewed the molecular regulatory mechanism, detection methods, and intervention strategies of fat infiltration in skeletal muscle. Based on the current findings, our review will provide new insight into regulating function and lipid metabolism of skeletal muscle and treating muscle-related diseases.
Collapse
Affiliation(s)
- Liyi Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | | | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| |
Collapse
|
30
|
Kang X, Qian J, Shi YX, Bian XT, Zhang LD, Li GM, Wang LT, Zhao J, Dong ZY, Yang MM, Chen YJN, Tang KL, Miao HM. Exercise-induced Musclin determines the fate of fibro-adipogenic progenitors to control muscle homeostasis. Cell Stem Cell 2024; 31:212-226.e7. [PMID: 38232727 DOI: 10.1016/j.stem.2023.12.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 01/19/2024]
Abstract
The effects of exercise on fibro-adipogenic progenitors (FAPs) are unclear, and the direct molecular link is still unknown. In this study, we reveal that exercise reduces the frequency of FAPs and attenuates collagen deposition and adipose formation in injured or disused muscles through Musclin. Mechanistically, Musclin inhibits FAP proliferation and promotes apoptosis in FAPs by upregulating FILIP1L. Chromatin immunoprecipitation (ChIP)-qPCR confirms that FoxO3a is the transcription factor of FILIP1L. In addition, the Musclin/FILIP1L pathway facilitates the phagocytosis of apoptotic FAPs by macrophages through downregulating the expression of CD47. Genetic ablation of FILIP1L in FAPs abolishes the effects of exercise or Musclin on FAPs and the benefits on the reduction of fibrosis and fatty infiltration. Overall, exercise forms a microenvironment of myokines in muscle and prevents the abnormal accumulation of FAPs in a Musclin/FILIP1L-dependent manner. The administration of exogenous Musclin exerts a therapeutic effect, demonstrating a potential therapeutic approach for muscle atrophy or acute muscle injury.
Collapse
Affiliation(s)
- Xia Kang
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China; Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu 610000, Sichuan, China.
| | - Jin Qian
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
| | - You-Xing Shi
- Department of Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Xu-Ting Bian
- Department of Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Li-Dan Zhang
- Center for Medical Epigenetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400038, China
| | - Gao-Ming Li
- Department of Health Statistics, Army Medical University, Chongqing 400038, China
| | - Li-Ting Wang
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Jing Zhao
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Zhen-Yu Dong
- Department of Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Meng-Meng Yang
- Center for Medical Epigenetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400038, China
| | - Yu-Jia-Nan Chen
- Department of Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Kang-Lai Tang
- Department of Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, First Affiliated Hospital of Army Medical University, Chongqing 400038, China.
| | - Hong-Ming Miao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China; Jinfeng Laboratory, Chongqing 401329, China.
| |
Collapse
|
31
|
Kim JT, Chen J, Zhou Y, Son MJ, Jeon DH, Kwon JW, Lee GY, Lee HJ. Cycloastragenol inhibits adipogenesis and fat accumulation in vitro and in vivo through activating Hedgehog signaling. Food Sci Biotechnol 2024; 33:711-720. [PMID: 38274180 PMCID: PMC10805729 DOI: 10.1007/s10068-023-01403-0] [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: 06/21/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 01/27/2024] Open
Abstract
In this study, we investigated the effect of cycloastragenol (CAG), a triterpenoid isolated from Astragalus membranaceus roots, on regulating the adipogenesis and fat accumulation in vitro and in vivo. During the adipogenesis of 3T3-L1 cells, CAG inhibited lipid accumulation and the expression of key adipogenic factors, proliferator-activated receptor γ (PPARγ) and CCAAT enhancer binding protein α (C/EBPα) and increased the expression of Gli1, a key mediator in Hedgehog (Hh) signaling. In HFD-induced animal experiment, CAG significantly reduced body weight gain without affecting brown fat weight. In addition, CAG regulated the expression of PPARγ, C/EBPα, and Gli1 in visceral white adipose tissue (vWAT). We also confirmed the inhibitory effect of CAG on specifically targeting white adipose tissue (WAT) formation in stromal vascular fraction (SVF) cell differentiation. Taken together, these results suggest that CAG may be a potent phytochemical preventing adipogenesis and obesity via Hh signaling. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-023-01403-0.
Collapse
Affiliation(s)
- Jin Tae Kim
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 456-756 South Korea
| | - Jing Chen
- Institute for Advanced and Applied Chemical Synthesis, Jinan University, Guangzhou, 510632 China
| | - Yimeng Zhou
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 456-756 South Korea
| | - Moon Jeong Son
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 456-756 South Korea
| | - Dong Hyeon Jeon
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 456-756 South Korea
| | - Jung Won Kwon
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 456-756 South Korea
| | - Ga Yeon Lee
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 456-756 South Korea
| | - Hong Jin Lee
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 456-756 South Korea
| |
Collapse
|
32
|
Li E, Yan R, Qiao H, Sun J, Zou P, Chang J, Li S, Ma Q, Zhang R, Liao B. Combined transcriptomics and proteomics studies on the effect of electrical stimulation on spinal cord injury in rats. Heliyon 2024; 10:e23960. [PMID: 38226269 PMCID: PMC10788535 DOI: 10.1016/j.heliyon.2023.e23960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 11/20/2023] [Accepted: 12/19/2023] [Indexed: 01/17/2024] Open
Abstract
Electrical stimulation (ES) of the spinal cord is a promising therapy for functional rehabilitation after spinal cord injury (SCI). However, the specific mechanism of action is poorly understood. We designed and applied an implanted ES device in the SCI area in rats and determined the effect of ES on the treatment of motor dysfunction after SCI using behavioral scores. Additionally, we examined the molecular characteristics of the samples using proteomic and transcriptomic sequencing. The differential molecules between groups were identified using statistical analyses. Molecular, network, and pathway-based analyses were used to identify group-specific biological features. ES (0.5 mA, 0.1 ms, 50 Hz) had a positive effect on motor dysfunction and neuronal regeneration in rats after SCI. Six samples (three independent replicates in each group) were used for transcriptome sequencing; we obtained 1026 differential genes, comprising 274 upregulated genes and 752 downregulated genes. A total of 10 samples were obtained: four samples in the ES group and six samples in the SCI group; for the proteome sequencing, 48 differential proteins were identified, including 45 up-regulated and three down-regulated proteins. Combined transcriptomic and proteomic studies have shown that the main enrichment pathway is the hedgehog signaling pathway. Western blot results showed that the expression levels of Sonic hedgehog (SHH) (P < 0.001), Smoothened (SMO) (P = 0.0338), and GLI-1 (P < 0.01) proteins in the ES treatment group were significantly higher than those in the SCI group. The immunofluorescence results showed significantly increased expression of SHH (P = 0.0181), SMO (P = 0.021), and GLI-1 (P = 0.0126) in the ES group compared with that in the SCI group. In conclusion, ES after SCI had a positive effect on motor dysfunction and anti-inflammatory effects in rats. Moreover, transcriptomic and proteomic sequencing also provided unique perspectives on the complex relationships between ES on SCI, where the SHH signaling pathway plays a critical role. Our study provides a significant theoretical foundation for the clinical implementation of ES therapy in patients with SCI.
Collapse
Affiliation(s)
- Erliang Li
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Rongbao Yan
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Huanhuan Qiao
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Jin Sun
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Peng Zou
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jiaqi Chang
- School of Automation Science and Electrical Engineering, Beihang University, 37th Xueyuan Road, Beijing, China
| | - Shuang Li
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Qiong Ma
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Rui Zhang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Bo Liao
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
33
|
Garcia SM, Lau J, Diaz A, Chi H, Lizarraga M, Wague A, Montenegro C, Davies MR, Liu X, Feeley BT. Distinct human stem cell subpopulations drive adipogenesis and fibrosis in musculoskeletal injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.28.551038. [PMID: 38260367 PMCID: PMC10802239 DOI: 10.1101/2023.07.28.551038] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Fibroadipogenic progenitors (FAPs) maintain healthy skeletal muscle in homeostasis but drive muscle degeneration in chronic injuries by promoting adipogenesis and fibrosis. To uncover how these stem cells switch from a pro-regenerative to pro-degenerative role we perform single-cell mRNA sequencing of human FAPs from healthy and injured human muscles across a spectrum of injury, focusing on rotator cuff tears. We identify multiple subpopulations with progenitor, adipogenic, or fibrogenic gene signatures. We utilize full spectrum flow cytometry to identify distinct FAP subpopulations based on highly multiplexed protein expression. Injury severity increases adipogenic commitment of FAP subpopulations and is driven by the downregulation of DLK1. Treatment of FAPs both in vitro and in vivo with DLK1 reduces adipogenesis and fatty infiltration, suggesting that during injury, reduced DLK1 within a subpopulation of FAPs may drive degeneration. This work highlights how stem cells perform varied functions depending on tissue context, by dynamically regulating subpopulation fate commitment, which can be targeted improve patient outcomes after injury.
Collapse
|
34
|
Tung LW, Groppa E, Soliman H, Lin B, Chang C, Cheung CW, Ritso M, Guo D, Rempel L, Sinha S, Eisner C, Brassard J, McNagny K, Biernaskie J, Rossi F. Spatiotemporal signaling underlies progressive vascular rarefaction in myocardial infarction. Nat Commun 2023; 14:8498. [PMID: 38129410 PMCID: PMC10739910 DOI: 10.1038/s41467-023-44227-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
Therapeutic angiogenesis represents a promising avenue to revascularize the ischemic heart. Its limited success is partly due to our poor understanding of the cardiac stroma, specifically mural cells, and their response to ischemic injury. Here, we combine single-cell and positional transcriptomics to assess the behavior of mural cells within the healing heart. In response to myocardial infarction, mural cells adopt an altered state closely associated with the infarct and retain a distinct lineage from fibroblasts. This response is concurrent with vascular rarefaction and reduced vascular coverage by mural cells. Positional transcriptomics reveals that the infarcted heart is governed by regional-dependent and temporally regulated programs. While the remote zone acts as an important source of pro-angiogenic signals, the infarct zone is accentuated by chronic activation of anti-angiogenic, pro-fibrotic, and inflammatory cues. Together, our work unveils the spatiotemporal programs underlying cardiac repair and establishes an association between vascular deterioration and mural cell dysfunction.
Collapse
Affiliation(s)
- Lin Wei Tung
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Elena Groppa
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Borea Therapeutics, Scuola Internazionale Superiore di Studi Avanzati, Via Bonomea, 265, 34136, Trieste, Italy
| | - Hesham Soliman
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Aspect Biosystems, 1781 W 75th Ave, Vancouver, BC, V6P 6P2, Canada
- Faculty of Pharmaceutical Sciences, Minia University, Minia, Egypt
| | - Bruce Lin
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Chihkai Chang
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Chun Wai Cheung
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Morten Ritso
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - David Guo
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Lucas Rempel
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Sarthak Sinha
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Christine Eisner
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Julyanne Brassard
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Kelly McNagny
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Jeff Biernaskie
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Fabio Rossi
- School of Biomedical Engineering & Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
35
|
Song Y, Wei D, Raza SHA, Zhao Y, Jiang C, Song X, Wu H, Wang X, Luoreng Z, Ma Y. Research progress of intramuscular fat formation based on co-culture. Anim Biotechnol 2023; 34:3216-3236. [PMID: 36200856 DOI: 10.1080/10495398.2022.2127410] [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: 11/01/2022]
Abstract
Intramuscular fat (IMF) is closely related to the meat quality of livestock and poultry. As a new cell culture technique in vitro, cell co-culture has been gradually applied to the related research of IMF formation because it can simulate the changes of microenvironment in vivo during the process of IMF cell formation. In the co-culture model, in addition to studying the effects of skeletal muscle cells on the proliferation and differentiation of IMF, we can also consider the role of many secretion factors in the formation of IMF, thus making the cell research in vitro closer to the real level in vivo. This paper reviewed the generation and origin of IMF, summarized the existing co-culture methods and systems, and discussed the advantages and disadvantages of each method as well as the challenges faced in the establishment of the system, with emphasis on the current status of research on the formation of IMF for human and animal based on co-culture technology.
Collapse
Affiliation(s)
- Yaping Song
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Dawei Wei
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | | | - Yiang Zhao
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Chao Jiang
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Xiaoyu Song
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Hao Wu
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Xingping Wang
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Zhuoma Luoreng
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Yun Ma
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| |
Collapse
|
36
|
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
|
37
|
Xu D, Wan B, Qiu K, Wang Y, Zhang X, Jiao N, Yan E, Wu J, Yu R, Gao S, Du M, Liu C, Li M, Fan G, Yin J. Single-Cell RNA-Sequencing Provides Insight into Skeletal Muscle Evolution during the Selection of Muscle Characteristics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2305080. [PMID: 37870215 PMCID: PMC10724408 DOI: 10.1002/advs.202305080] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/27/2023] [Indexed: 10/24/2023]
Abstract
Skeletal muscle comprises a large, heterogeneous assortment of cell populations that interact to maintain muscle homeostasis, but little is known about the mechanism that controls myogenic development in response to artificial selection. Different pig (Sus scrofa) breeds exhibit distinct muscle phenotypes resulting from domestication and selective breeding. Using unbiased single-cell transcriptomic sequencing analysis (scRNA-seq), the impact of artificial selection on cell profiles is investigated in neonatal skeletal muscle of pigs. This work provides panoramic muscle-resident cell profiles and identifies novel and breed-specific cells, mapping them on pseudotime trajectories. Artificial selection has elicited significant changes in muscle-resident cell profiles, while conserving signs of generational environmental challenges. These results suggest that fibro-adipogenic progenitors serve as a cellular interaction hub and that specific transcription factors identified here may serve as candidate target regulons for the pursuit of a specific muscle phenotype. Furthermore, a cross-species comparison of humans, mice, and pigs illustrates the conservation and divergence of mammalian muscle ontology. The findings of this study reveal shifts in cellular heterogeneity, novel cell subpopulations, and their interactions that may greatly facilitate the understanding of the mechanism underlying divergent muscle phenotypes arising from artificial selection.
Collapse
Affiliation(s)
- Doudou Xu
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Boyang Wan
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Kai Qiu
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Yubo Wang
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Xin Zhang
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
- Molecular Design Breeding Frontier Science Center of the Ministry of EducationBeijingChina
| | - Ning Jiao
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Enfa Yan
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Jiangwei Wu
- Key Laboratory of Animal GeneticsBreeding and Reproduction of Shaanxi ProvinceCollege of Animal Science and TechnologyNorthwest A&F UniversityYangling712100China
| | - Run Yu
- Beijing National Day SchoolBeijing100039China
| | - Shuai Gao
- Key Laboratory of Animal GeneticsCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Min Du
- Nutrigenomics and Growth Biology LaboratoryDepartment of Animal Sciences and School of Molecular BioscienceWashington State UniversityPullmanWA99164USA
| | | | - Mingzhou Li
- Institute of Animal Genetics and BreedingCollege of Animal Science and TechnologySichuan Agricultural UniversityChengdu625014China
| | - Guoping Fan
- Department of Human GeneticsDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
| | - Jingdong Yin
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
- Molecular Design Breeding Frontier Science Center of the Ministry of EducationBeijingChina
| |
Collapse
|
38
|
You W, Xu Z, Chen W, Yang X, Liu S, Wang L, Tu Y, Zhou Y, Valencak TG, Wang Y, Kuang S, Shan T. Cellular and Transcriptional Dynamics during Brown Adipose Tissue Regeneration under Acute Injury. RESEARCH (WASHINGTON, D.C.) 2023; 6:0268. [PMID: 38434240 PMCID: PMC10907023 DOI: 10.34133/research.0268] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 10/18/2023] [Indexed: 03/05/2024]
Abstract
Brown adipose tissue (BAT) is the major site of non-shivering thermogenesis and crucial for systemic metabolism. Under chronic cold exposures and high-fat diet challenges, BAT undergoes robust remodeling to adapt to physiological demands. However, whether and how BAT regenerates after acute injuries are poorly understood. Here, we established a novel BAT injury and regeneration model (BAT-IR) in mice and performed single-cell RNA sequencing (scRNA-seq) and bulk RNA-seq to determine cellular and transcriptomic dynamics during BAT-IR. We further defined distinct fibro-adipogenic and myeloid progenitor populations contributing to BAT regeneration. Cell trajectory and gene expression analyses uncovered the involvement of MAPK, Wnt, and Hedgehog (Hh) signaling pathways in BAT regeneration. We confirmed the role of Hh signaling in BAT development through Myf5Cre-mediated conditional knockout (cKO) of the Sufu gene to activate Hh signaling in BAT and muscle progenitors. Our BAT-IR model therefore provides a paradigm to identify conserved cellular and molecular mechanisms underlying BAT development and remodeling.
Collapse
Affiliation(s)
- Wenjing You
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Ziye Xu
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- Department of Laboratory Medicine, the First Affiliated Hospital, College of Medicine,
Zhejiang University, Hangzhou, China
| | - Wentao Chen
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Xin Yang
- Department of Animal Sciences,
Purdue University, West Lafayette, IN, USA
| | - Shiqi Liu
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Liyi Wang
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Yuang Tu
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Yanbing Zhou
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | | | - Yizhen Wang
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Shihuan Kuang
- Department of Animal Sciences,
Purdue University, West Lafayette, IN, USA
| | - Tizhong Shan
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| |
Collapse
|
39
|
Hwang SH, White KA, Somatilaka BN, Wang B, Mukhopadhyay S. Context-dependent ciliary regulation of hedgehog pathway repression in tissue morphogenesis. PLoS Genet 2023; 19:e1011028. [PMID: 37943875 PMCID: PMC10662714 DOI: 10.1371/journal.pgen.1011028] [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: 07/10/2023] [Revised: 11/21/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023] Open
Abstract
A fundamental problem in tissue morphogenesis is identifying how subcellular signaling regulates mesoscale organization of tissues. The primary cilium is a paradigmatic organelle for compartmentalized subcellular signaling. How signaling emanating from cilia orchestrates tissue organization-especially, the role of cilia-generated effectors in mediating diverse morpho-phenotypic outcomes-is not well understood. In the hedgehog pathway, bifunctional GLI transcription factors generate both GLI-activators (GLI-A) and GLI-repressors (GLI-R). The formation of GLI-A/GLI-R requires cilia. However, how these counterregulatory effectors coordinate cilia-regulated morphogenetic pathways is unclear. Here we determined GLI-A/GLI-R requirements in phenotypes arising from lack of hedgehog pathway repression (derepression) during mouse neural tube and skeletal development. We studied hedgehog pathway repression by the GPCR GPR161, and the ankyrin repeat protein ANKMY2 that direct cAMP/protein kinase-A signaling by cilia in GLI-R generation. We performed genetic epistasis between Gpr161 or Ankmy2 mutants, and Gli2/Gli3 knockouts, Gli3R knock-in and knockout of Smoothened, the hedgehog pathway transducer. We also tested the role of cilia-generated signaling using a Gpr161 ciliary localization knock-in mutant that is cAMP signaling competent. We found that the cilia-dependent derepression phenotypes arose in three modes: lack of GLI-R only, excess GLI-A formation only, or dual regulation of either lack of GLI-R or excess GLI-A formation. These modes were mostly independent of Smoothened. The cAMP signaling-competent non-ciliary Gpr161 knock-in recapitulated Gpr161 loss-of-function tissue phenotypes solely from lack of GLI-R only. Our results show complex tissue-specific GLI-effector requirements in morphogenesis and point to tissue-specific GLI-R thresholds generated by cilia in hedgehog pathway repression. Broadly, our study sets up a conceptual framework for rationalization of different modes of signaling generated by the primary cilium in mediating morphogenesis in diverse tissues.
Collapse
Affiliation(s)
- Sun-Hee Hwang
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Kevin Andrew White
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Bandarigoda Nipunika Somatilaka
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Present address, Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Baolin Wang
- Department of Genetic Medicine, Weill Medical College of Cornell University, New York, New York, United States of America
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| |
Collapse
|
40
|
Pass CG, Palzkill V, Tan J, Kim K, Thome T, Yang Q, Fazzone B, Robinson ST, O’Malley KA, Yue F, Scali ST, Berceli SA, Ryan TE. Single-Nuclei RNA-Sequencing of the Gastrocnemius Muscle in Peripheral Artery Disease. Circ Res 2023; 133:791-809. [PMID: 37823262 PMCID: PMC10599805 DOI: 10.1161/circresaha.123.323161] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND Lower extremity peripheral artery disease (PAD) is a growing epidemic with limited effective treatment options. Here, we provide a single-nuclei atlas of PAD limb muscle to facilitate a better understanding of the composition of cells and transcriptional differences that comprise the diseased limb muscle. METHODS We obtained gastrocnemius muscle specimens from 20 patients with PAD and 12 non-PAD controls. Nuclei were isolated and single-nuclei RNA-sequencing was performed. The composition of nuclei was characterized by iterative clustering via principal component analysis, differential expression analysis, and the use of known marker genes. Bioinformatics analysis was performed to determine differences in gene expression between PAD and non-PAD nuclei, as well as subsequent analysis of intercellular signaling networks. Additional histological analyses of muscle specimens accompany the single-nuclei RNA-sequencing atlas. RESULTS Single-nuclei RNA-sequencing analysis indicated a fiber type shift with patients with PAD having fewer type I (slow/oxidative) and more type II (fast/glycolytic) myonuclei compared with non-PAD, which was confirmed using immunostaining of muscle specimens. Myonuclei from PAD displayed global upregulation of genes involved in stress response, autophagy, hypoxia, and atrophy. Subclustering of myonuclei also identified populations that were unique to PAD muscle characterized by metabolic dysregulation. PAD muscles also displayed unique transcriptional profiles and increased diversity of transcriptomes in muscle stem cells, regenerating myonuclei, and fibro-adipogenic progenitor cells. Analysis of intercellular communication networks revealed fibro-adipogenic progenitors as a major signaling hub in PAD muscle, as well as deficiencies in angiogenic and bone morphogenetic protein signaling which may contribute to poor limb function in PAD. CONCLUSIONS This reference single-nuclei RNA-sequencing atlas provides a comprehensive analysis of the cell composition, transcriptional signature, and intercellular communication pathways that are altered in the PAD condition.
Collapse
Affiliation(s)
- Caroline G. Pass
- Department of Applied Physiology and Kinesiology (C.G.P., V.P., J.T., K.K., T.T., Q.Y., T.E.R.), The University of Florida, Gainesville
| | - Victoria Palzkill
- Department of Applied Physiology and Kinesiology (C.G.P., V.P., J.T., K.K., T.T., Q.Y., T.E.R.), The University of Florida, Gainesville
| | - Jianna Tan
- Department of Applied Physiology and Kinesiology (C.G.P., V.P., J.T., K.K., T.T., Q.Y., T.E.R.), The University of Florida, Gainesville
| | - Kyoungrae Kim
- Department of Applied Physiology and Kinesiology (C.G.P., V.P., J.T., K.K., T.T., Q.Y., T.E.R.), The University of Florida, Gainesville
| | - Trace Thome
- Department of Applied Physiology and Kinesiology (C.G.P., V.P., J.T., K.K., T.T., Q.Y., T.E.R.), The University of Florida, Gainesville
| | - Qingping Yang
- Department of Applied Physiology and Kinesiology (C.G.P., V.P., J.T., K.K., T.T., Q.Y., T.E.R.), The University of Florida, Gainesville
| | - Brian Fazzone
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy (B.F., S.T.R., K.A.O., S.T.S., S.A.B.), The University of Florida, Gainesville
- Malcom Randall VA Medical Center, Gainesville, FL (B.F., S.T.R., K.A.O., S.T.S., S.A.B.)
| | - Scott T. Robinson
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy (B.F., S.T.R., K.A.O., S.T.S., S.A.B.), The University of Florida, Gainesville
- Malcom Randall VA Medical Center, Gainesville, FL (B.F., S.T.R., K.A.O., S.T.S., S.A.B.)
| | - Kerri A. O’Malley
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy (B.F., S.T.R., K.A.O., S.T.S., S.A.B.), The University of Florida, Gainesville
- Malcom Randall VA Medical Center, Gainesville, FL (B.F., S.T.R., K.A.O., S.T.S., S.A.B.)
| | - Feng Yue
- Department of Animal Sciences (F.Y.), The University of Florida, Gainesville
- Myology Institute (F.Y., T.E.R.), The University of Florida, Gainesville
| | - Salvatore T. Scali
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy (B.F., S.T.R., K.A.O., S.T.S., S.A.B.), The University of Florida, Gainesville
- Malcom Randall VA Medical Center, Gainesville, FL (B.F., S.T.R., K.A.O., S.T.S., S.A.B.)
| | - Scott A. Berceli
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy (B.F., S.T.R., K.A.O., S.T.S., S.A.B.), The University of Florida, Gainesville
- Malcom Randall VA Medical Center, Gainesville, FL (B.F., S.T.R., K.A.O., S.T.S., S.A.B.)
| | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology (C.G.P., V.P., J.T., K.K., T.T., Q.Y., T.E.R.), The University of Florida, Gainesville
- Center for Exercise Science (T.E.R.), The University of Florida, Gainesville
- Myology Institute (F.Y., T.E.R.), The University of Florida, Gainesville
| |
Collapse
|
41
|
Roberson EC, Tran NK, Godambe AN, Mark H, Nguimtsop M, Rust T, Ung E, Barker LJ, Fitch RD, Wallingford JB. Hedgehog signaling is required for endometrial remodeling and myometrial homeostasis in the cycling mouse uterus. iScience 2023; 26:107993. [PMID: 37810243 PMCID: PMC10551904 DOI: 10.1016/j.isci.2023.107993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 08/24/2023] [Accepted: 09/16/2023] [Indexed: 10/10/2023] Open
Abstract
Decades of work demonstrate that the mammalian estrous cycle is controlled by cycling steroid hormones. However, the signaling mechanisms that act downstream, linking hormonal action to the physical remodeling of the cycling uterus, remain unclear. To address this issue, we analyzed gene expression at all stages of the mouse estrous cycle. Strikingly, we found that several genetic programs well-known to control tissue morphogenesis in developing embryos displayed cyclical patterns of expression. We find that most of the genetic architectures of Hedgehog signaling (ligands, receptors, effectors, and transcription factors) are transcribed cyclically in the uterus, and that conditional disruption of the Hedgehog receptor smoothened not only elicits a failure of normal cyclical thickening of the endometrial lining but also induces aberrant deformation of the uterine smooth muscle. Together, our data shed light on the mechanisms underlying normal uterine remodeling specifically and cyclical gene expression generally.
Collapse
Affiliation(s)
- Elle C. Roberson
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical School, Aurora, CO 80045, USA
| | - Ngan Kim Tran
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Anushka N. Godambe
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Harrison Mark
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Michelle Nguimtsop
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Trinity Rust
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Elizabeth Ung
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical School, Aurora, CO 80045, USA
| | - LeCaine J. Barker
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical School, Aurora, CO 80045, USA
| | - Rebecca D. Fitch
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - John B. Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
42
|
Zhang T, Li J, Li X, Liu Y. Intermuscular adipose tissue in obesity and related disorders: cellular origins, biological characteristics and regulatory mechanisms. Front Endocrinol (Lausanne) 2023; 14:1280853. [PMID: 37920255 PMCID: PMC10619759 DOI: 10.3389/fendo.2023.1280853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/01/2023] [Indexed: 11/04/2023] Open
Abstract
Intermuscular adipose tissue (IMAT) is a unique adipose depot interspersed between muscle fibers (myofibers) or muscle groups. Numerous studies have shown that IMAT is strongly associated with insulin resistance and muscular dysfunction in people with metabolic disease, such as obesity and type 2 diabetes. Moreover, IMAT aggravates obesity-related muscle metabolism disorders via secretory factors. Interestingly, researchers have discovered that intermuscular brown adipocytes in rodent models provide new hope for obesity treatment by acting on energy dissipation, which inspired researchers to explore the underlying regulation of IMAT formation. However, the molecular and cellular properties and regulatory processes of IMAT remain debated. Previous studies have suggested that muscle-derived stem/progenitor cells and other adipose tissue progenitors contribute to the development of IMAT. Adipocytes within IMAT exhibit features that are similar to either white adipocytes or uncoupling protein 1 (UCP1)-positive brown adipocytes. Additionally, given the heterogeneity of skeletal muscle, which comprises myofibers, satellite cells, and resident mesenchymal progenitors, it is plausible that interplay between these cellular components actively participate in the regulation of intermuscular adipogenesis. In this context, we review recent studies associated with IMAT to offer insights into the cellular origins, biological properties, and regulatory mechanisms of IMAT. Our aim is to provide novel ideas for the therapeutic strategy of IMAT and the development of new drugs targeting IMAT-related metabolic diseases.
Collapse
Affiliation(s)
- Ting Zhang
- Center of Obesity and Metabolic Diseases, Department of General Surgery, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University & The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University & The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
- Medical Research Center, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University & The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
| | - Jun Li
- Department of Orthopedics, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University & The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
| | - Xi Li
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Yanjun Liu
- Center of Obesity and Metabolic Diseases, Department of General Surgery, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University & The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University & The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
| |
Collapse
|
43
|
Loomis T, Smith LR. Thrown for a loop: fibro-adipogenic progenitors in skeletal muscle fibrosis. Am J Physiol Cell Physiol 2023; 325:C895-C906. [PMID: 37602412 PMCID: PMC11932532 DOI: 10.1152/ajpcell.00245.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 08/22/2023]
Abstract
Fibro-adipogenic progenitors (FAPs) are key regulators of skeletal muscle regeneration and homeostasis. However, dysregulation of these cells leads to fibro-fatty infiltration across various muscle diseases. FAPs are the key source of extracellular matrix (ECM) deposition in muscle, and disruption to this process leads to a pathological accumulation of ECM, known as fibrosis. The replacement of contractile tissue with fibrotic ECM functionally impairs the muscle and increases muscle stiffness. FAPs and fibrotic muscle form a progressively degenerative feedback loop where, as a muscle becomes fibrotic, it induces a fibrotic FAP phenotype leading to further development of fibrosis. In this review, we summarize FAPs' role in fibrosis in terms of their activation, heterogeneity, contributions to fibrotic degeneration, and role across musculoskeletal diseases. We also discuss current research on potential therapeutic avenues to attenuate fibrosis by targeting FAPs.
Collapse
Affiliation(s)
- Taryn Loomis
- Biomedical Engineering Graduate Group, University of California, Davis, California, United States
| | - Lucas R Smith
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, California, United States
- Department of Physical Medicine and Rehabilitation, University of California, Davis, California, United States
| |
Collapse
|
44
|
Derderian C, Canales GI, Reiter JF. Seriously cilia: A tiny organelle illuminates evolution, disease, and intercellular communication. Dev Cell 2023; 58:1333-1349. [PMID: 37490910 PMCID: PMC10880727 DOI: 10.1016/j.devcel.2023.06.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/18/2023] [Accepted: 06/30/2023] [Indexed: 07/27/2023]
Abstract
The borders between cell and developmental biology, which have always been permeable, have largely dissolved. One manifestation is the blossoming of cilia biology, with cell and developmental approaches (increasingly complemented by human genetics, structural insights, and computational analysis) fruitfully advancing understanding of this fascinating, multifunctional organelle. The last eukaryotic common ancestor probably possessed a motile cilium, providing evolution with ample opportunity to adapt cilia to many jobs. Over the last decades, we have learned how non-motile, primary cilia play important roles in intercellular communication. Reflecting their diverse motility and signaling functions, compromised cilia cause a diverse range of diseases collectively called "ciliopathies." In this review, we highlight how cilia signal, focusing on how second messengers generated in cilia convey distinct information; how cilia are a potential source of signals to other cells; how evolution may have shaped ciliary function; and how cilia research may address thorny outstanding questions.
Collapse
Affiliation(s)
- Camille Derderian
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Gabriela I Canales
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Jeremy F Reiter
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
45
|
Lilly AC, Astsaturov I, Golemis EA. Intrapancreatic fat, pancreatitis, and pancreatic cancer. Cell Mol Life Sci 2023; 80:206. [PMID: 37452870 PMCID: PMC10349727 DOI: 10.1007/s00018-023-04855-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/18/2023]
Abstract
Pancreatic cancer is typically detected at an advanced stage, and is refractory to most forms of treatment, contributing to poor survival outcomes. The incidence of pancreatic cancer is gradually increasing, linked to an aging population and increasing rates of obesity and pancreatitis, which are risk factors for this cancer. Sources of risk include adipokine signaling from fat cells throughout the body, elevated levels of intrapancreatic intrapancreatic adipocytes (IPAs), inflammatory signals arising from pancreas-infiltrating immune cells and a fibrotic environment induced by recurring cycles of pancreatic obstruction and acinar cell lysis. Once cancers become established, reorganization of pancreatic tissue typically excludes IPAs from the tumor microenvironment, which instead consists of cancer cells embedded in a specialized microenvironment derived from cancer-associated fibroblasts (CAFs). While cancer cell interactions with CAFs and immune cells have been the topic of much investigation, mechanistic studies of the source and function of IPAs in the pre-cancerous niche are much less developed. Intriguingly, an extensive review of studies addressing the accumulation and activity of IPAs in the pancreas reveals that unexpectedly diverse group of factors cause replacement of acinar tissue with IPAs, particularly in the mouse models that are essential tools for research into pancreatic cancer. Genes implicated in regulation of IPA accumulation include KRAS, MYC, TGF-β, periostin, HNF1, and regulators of ductal ciliation and ER stress, among others. These findings emphasize the importance of studying pancreas-damaging factors in the pre-cancerous environment, and have significant implications for the interpretation of data from mouse models for pancreatic cancer.
Collapse
Affiliation(s)
- Anna C Lilly
- Program in Cancer Signaling and Microenvironment, Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA, 19111, USA
- Molecular & Cell Biology & Genetics (MCBG) Program, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Igor Astsaturov
- Program in Cancer Signaling and Microenvironment, Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA, 19111, USA
- The Marvin & Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Erica A Golemis
- Program in Cancer Signaling and Microenvironment, Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA, 19111, USA.
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA.
| |
Collapse
|
46
|
Norris AM, Appu AB, Johnson CD, Zhou LY, McKellar DW, Renault MA, Hammers D, Cosgrove BD, Kopinke D. Hedgehog signaling via its ligand DHH acts as cell fate determinant during skeletal muscle regeneration. Nat Commun 2023; 14:3766. [PMID: 37355632 PMCID: PMC10290686 DOI: 10.1038/s41467-023-39506-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 06/16/2023] [Indexed: 06/26/2023] Open
Abstract
Successful muscle regeneration relies on the interplay of multiple cell populations. However, the signals required for this coordinated intercellular crosstalk remain largely unknown. Here, we describe how the Hedgehog (Hh) signaling pathway controls the fate of fibro/adipogenic progenitors (FAPs), the cellular origin of intramuscular fat (IMAT) and fibrotic scar tissue. Using conditional mutagenesis and pharmacological Hh modulators in vivo and in vitro, we identify DHH as the key ligand that acts as a potent adipogenic brake by preventing the adipogenic differentiation of FAPs. Hh signaling also impacts muscle regeneration, albeit indirectly through induction of myogenic factors in FAPs. Our results also indicate that ectopic and sustained Hh activation forces FAPs to adopt a fibrogenic fate resulting in widespread fibrosis. In this work, we reveal crucial post-developmental functions of Hh signaling in balancing tissue regeneration and fatty fibrosis. Moreover, they provide the exciting possibility that mis-regulation of the Hh pathway with age and disease could be a major driver of pathological IMAT formation.
Collapse
Affiliation(s)
- Alessandra M Norris
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Ambili Bai Appu
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Connor D Johnson
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Lylybell Y Zhou
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - David W McKellar
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Marie-Ange Renault
- Biology of Cardiovascular Diseases, INSERM, University of Bordeaux, Pessac, France
| | - David Hammers
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Benjamin D Cosgrove
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Daniel Kopinke
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
47
|
Parson JC, Biltz NK, Meyer GA. Decellularization-Based Quantification of Skeletal Muscle Fatty Infiltration. J Vis Exp 2023. [PMID: 37358301 PMCID: PMC10837739 DOI: 10.3791/65461] [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] [Indexed: 06/27/2023] Open
Abstract
Fatty infiltration is the accumulation of adipocytes between myofibers in skeletal muscle and is a prominent feature of many myopathies, metabolic disorders, and dystrophies. Clinically in human populations, fatty infiltration is assessed using noninvasive methods, including computed tomography (CT), magnetic resonance imaging (MRI), and ultrasound (US). Although some studies have used CT or MRI to quantify fatty infiltration in mouse muscle, costs and insufficient spatial resolution remain challenging. Other small animal methods utilize histology to visualize individual adipocytes; however, this methodology suffers from sampling bias in heterogeneous pathology. This protocol describes the methodology to qualitatively view and quantitatively measure fatty infiltration comprehensively throughout intact mouse muscle and at the level of individual adipocytes using decellularization. The protocol is not limited to specific muscles or specific species and can be extended to human biopsy. Additionally, gross qualitative and quantitative assessments can be made with standard laboratory equipment for little cost, making this procedure more accessible across research laboratories.
Collapse
Affiliation(s)
- Jacob C Parson
- Program in Physical Therapy, Washington University in St. Louis
| | - Nicole K Biltz
- Program in Physical Therapy, Washington University in St. Louis
| | - Gretchen A Meyer
- Program in Physical Therapy, Washington University in St. Louis; Departments of Neurology, Orthopaedic Surgery and Biomedical Engineering, Washington University in St. Louis;
| |
Collapse
|
48
|
Tu HQ, Li S, Xu YL, Zhang YC, Li PY, Liang LY, Song GP, Jian XX, Wu M, Song ZQ, Li TT, Hu HB, Yuan JF, Shen XL, Li JN, Han QY, Wang K, Zhang T, Zhou T, Li AL, Zhang XM, Li HY. Rhythmic cilia changes support SCN neuron coherence in circadian clock. Science 2023; 380:972-979. [PMID: 37262147 DOI: 10.1126/science.abm1962] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 04/13/2023] [Indexed: 06/03/2023]
Abstract
The suprachiasmatic nucleus (SCN) drives circadian clock coherence through intercellular coupling, which is resistant to environmental perturbations. We report that primary cilia are required for intercellular coupling among SCN neurons to maintain the robustness of the internal clock in mice. Cilia in neuromedin S-producing (NMS) neurons exhibit pronounced circadian rhythmicity in abundance and length. Genetic ablation of ciliogenesis in NMS neurons enabled a rapid phase shift of the internal clock under jet-lag conditions. The circadian rhythms of individual neurons in cilia-deficient SCN slices lost their coherence after external perturbations. Rhythmic cilia changes drive oscillations of Sonic Hedgehog (Shh) signaling and clock gene expression. Inactivation of Shh signaling in NMS neurons phenocopied the effects of cilia ablation. Thus, cilia-Shh signaling in the SCN aids intercellular coupling.
Collapse
Affiliation(s)
- Hai-Qing Tu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Sen Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Yu-Ling Xu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Yu-Cheng Zhang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Pei-Yao Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Li-Yun Liang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Guang-Ping Song
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Xiao-Xiao Jian
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Min Wu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Zeng-Qing Song
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Ting-Ting Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Huai-Bin Hu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Jin-Feng Yuan
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Xiao-Lin Shen
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Jia-Ning Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Qiu-Ying Han
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Kai Wang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Tao Zhang
- Laboratory Animal Center, Academy of Military Medical Sciences, Beijing, China
| | - Tao Zhou
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Ai-Ling Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xue-Min Zhang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hui-Yan Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
49
|
Tao D, Zhang L, Ding Y, Tang N, Xu X, Li G, Niu P, Yue R, Wang X, Shen Y, Sun Y. Primary cilia support cartilage regeneration after injury. Int J Oral Sci 2023; 15:22. [PMID: 37268650 DOI: 10.1038/s41368-023-00223-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 03/24/2023] [Indexed: 06/04/2023] Open
Abstract
In growing children, growth plate cartilage has limited self-repair ability upon fracture injury always leading to limb growth arrest. Interestingly, one type of fracture injuries within the growth plate achieve amazing self-healing, however, the mechanism is unclear. Using this type of fracture mouse model, we discovered the activation of Hedgehog (Hh) signaling in the injured growth plate, which could activate chondrocytes in growth plate and promote cartilage repair. Primary cilia are the central transduction mediator of Hh signaling. Notably, ciliary Hh-Smo-Gli signaling pathways were enriched in the growth plate during development. Moreover, chondrocytes in resting and proliferating zone were dynamically ciliated during growth plate repair. Furthermore, conditional deletion of the ciliary core gene Ift140 in cartilage disrupted cilia-mediated Hh signaling in growth plate. More importantly, activating ciliary Hh signaling by Smoothened agonist (SAG) significantly accelerated growth plate repair after injury. In sum, primary cilia mediate Hh signaling induced the activation of stem/progenitor chondrocytes and growth plate repair after fracture injury.
Collapse
Affiliation(s)
- Dike Tao
- Department of Implantology, School & Hospital of Stomatology, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Lei Zhang
- Department of Implantology, School & Hospital of Stomatology, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Yunpeng Ding
- Department of Implantology, School & Hospital of Stomatology, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Na Tang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoqiao Xu
- Department of Implantology, School & Hospital of Stomatology, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Gongchen Li
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Tongji University, Shanghai, China
| | - Pingping Niu
- Department of Implantology, School & Hospital of Stomatology, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Rui Yue
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Xiaogang Wang
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China
| | - Yidong Shen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yao Sun
- Department of Implantology, School & Hospital of Stomatology, Tongji University, Shanghai, China.
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China.
| |
Collapse
|
50
|
Graca FA, Stephan A, Minden-Birkenmaier BA, Shirinifard A, Wang YD, Demontis F, Labelle M. Platelet-derived chemokines promote skeletal muscle regeneration by guiding neutrophil recruitment to injured muscles. Nat Commun 2023; 14:2900. [PMID: 37217480 DOI: 10.1038/s41467-023-38624-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 05/09/2023] [Indexed: 05/24/2023] Open
Abstract
Skeletal muscle regeneration involves coordinated interactions between different cell types. Injection of platelet-rich plasma is circumstantially considered an aid to muscle repair but whether platelets promote regeneration beyond their role in hemostasis remains unexplored. Here, we find that signaling via platelet-released chemokines is an early event necessary for muscle repair in mice. Platelet depletion reduces the levels of the platelet-secreted neutrophil chemoattractants CXCL5 and CXCL7/PPBP. Consequently, early-phase neutrophil infiltration to injured muscles is impaired whereas later inflammation is exacerbated. Consistent with this model, neutrophil infiltration to injured muscles is compromised in male mice with Cxcl7-knockout platelets. Moreover, neo-angiogenesis and the re-establishment of myofiber size and muscle strength occurs optimally in control mice post-injury but not in Cxcl7ko mice and in neutrophil-depleted mice. Altogether, these findings indicate that platelet-secreted CXCL7 promotes regeneration by recruiting neutrophils to injured muscles, and that this signaling axis could be utilized therapeutically to boost muscle regeneration.
Collapse
Affiliation(s)
- Flavia A Graca
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Anna Stephan
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Benjamin A Minden-Birkenmaier
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Department of Oncology, Division of Molecular Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Abbas Shirinifard
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yong-Dong Wang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Fabio Demontis
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| | - Myriam Labelle
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
- Department of Oncology, Division of Molecular Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|