1
|
Zhang Y, Hong S, Zhang F, Yao K, Jin S, Gao S, Liu Y, Li Y, Zhang C. Immunoproteasome subunit PSMB8 promotes skeletal muscle regeneration by regulating macrophage phenotyping switch in mice. Am J Physiol Cell Physiol 2025; 328:C1716-C1729. [PMID: 40241316 DOI: 10.1152/ajpcell.00965.2024] [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: 01/07/2025] [Accepted: 04/07/2025] [Indexed: 04/18/2025]
Abstract
Immunoproteasomes regulate the degradation of ubiquitin-coupled proteins and cell differentiation. However, its precise role in skeletal muscle regeneration remains unclear. In this study, we found that expression of the immunoproteasome subunit, PSMB8, increased significantly in young muscles after cardiotoxin-induced injury, whereas its expression was downregulated in injured aged mice. Genetic knockout or pharmacological inhibition of the immunoproteasome subunit, PSMB8, resulted in impaired muscle regeneration and increased interstitial fibrosis. PSMB8 inhibition by short interfering RNA (siRNA) or inhibitor decreased the differentiation ability of myoblasts. There was increased infiltration of inflammatory cells, especially Ly6Chi proinflammatory macrophages, in Psmb8 deficient muscles. In vitro, Psmb8-deficient macrophages expressed higher levels of proinflammatory cytokines and lower levels of anti-inflammatory cytokines after phagocytosis of myoblast debris, which was associated with increased activation of the NF-κB signaling pathway. Inhibition of the NF-κB pathway improves the regeneration ability and attenuates interstitial fibrosis in Psmb8-deficient muscles after injury. The overexpression of Psmb8 by adenovirus could also improve the regenerative ability of aged muscles.NEW & NOTEWORTHY The immunoproteasome subunit, PSMB8, is essential for efficient muscle regeneration and may be a new therapeutic target for age-related muscle atrophy.
Collapse
Affiliation(s)
- Yanhong Zhang
- Beijing Anzhen Hospital, Capital Medical UniversityBeijingPeople's Republic of China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, People's Republic of China
| | - Shiyao Hong
- Beijing Anzhen Hospital, Capital Medical UniversityBeijingPeople's Republic of China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, People's Republic of China
| | - Fan Zhang
- Beijing Anzhen Hospital, Capital Medical UniversityBeijingPeople's Republic of China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, People's Republic of China
| | - Kexin Yao
- Beijing Anzhen Hospital, Capital Medical UniversityBeijingPeople's Republic of China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, People's Republic of China
| | - Shuhui Jin
- Beijing Anzhen Hospital, Capital Medical UniversityBeijingPeople's Republic of China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, People's Republic of China
| | - Shijuan Gao
- Beijing Anzhen Hospital, Capital Medical UniversityBeijingPeople's Republic of China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, People's Republic of China
| | - Yan Liu
- Beijing Anzhen Hospital, Capital Medical UniversityBeijingPeople's Republic of China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, People's Republic of China
| | - Yulin Li
- Beijing Anzhen Hospital, Capital Medical UniversityBeijingPeople's Republic of China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, People's Republic of China
| | - Congcong Zhang
- Beijing Anzhen Hospital, Capital Medical UniversityBeijingPeople's Republic of China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, People's Republic of China
| |
Collapse
|
2
|
Francos-Quijorna I, López-González N, Caro-Canton M, Sánchez-Fernández A, Hernández-Mir G, López-Vales R. Lack of effects of Resolvin D1 after spinal cord injury in mice. Exp Neurol 2025; 388:115226. [PMID: 40120661 DOI: 10.1016/j.expneurol.2025.115226] [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: 12/24/2024] [Revised: 03/09/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Inflammation is a fundamental component of the body's response to injury or infection and is responsible for restoring tissue homeostasis and starting the wound healing process. To avoid excessive tissue damage, it is important to efficiently resolve inflammation once it is no longer necessary. In recent years, the discovery of pro-resolving lipid mediators derived from polyunsaturated fatty acids, such as Resolvin D1 (RvD1), has shed light on the resolution of inflammation. However, the impact of RvD1 on Spinal Cord Injury (SCI) remains unexplored. In this study, we provide direct evidence that the administration of RvD1 for one week after SCI fails to enhance resolution of inflammation and does not improve functional and histological outcomes. Our transcriptomic analysis reveals that RvD1 does not modulate inflammatory response pathways in the injured spinal cord but leads to significant changes in the expression of genes related to ribosomal function and extracellular matrix pathways. Unlike SCI, RvD1 treatment ameliorated neurological deficits in experimental autoimmune encephalomyelitis. Our findings represent the first report demonstrating that RvD1 treatment does not exert therapeutic actions in the context of SCI and suggest that this pro-resolving agonist may exert therapeutic actions in certain but not in all conditions involving an inflammatory component.
Collapse
Affiliation(s)
- Isaac Francos-Quijorna
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Institut de Neurociències, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Bellaterra, 08193, Catalonia, Spain
| | - Néstor López-González
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Institut de Neurociències, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Bellaterra, 08193, Catalonia, Spain
| | - Marc Caro-Canton
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Institut de Neurociències, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Bellaterra, 08193, Catalonia, Spain
| | - Alba Sánchez-Fernández
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Institut de Neurociències, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Bellaterra, 08193, Catalonia, Spain
| | - Gerard Hernández-Mir
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine, Queen Mary University of London, London E1 2AT, UK
| | - Rubèn López-Vales
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Institut de Neurociències, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Bellaterra, 08193, Catalonia, Spain.
| |
Collapse
|
3
|
Ivy A, Bess SN, Agrawal S, Kochar V, Stokes AL, Muldoon TJ, Nelson CE. A dual-fluorescence assay for gene delivery vehicle screening in macrophages with an inflammation-inducible reporter construct. BMC METHODS 2025; 2:8. [PMID: 40352095 PMCID: PMC12062070 DOI: 10.1186/s44330-025-00030-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 05/01/2025] [Indexed: 05/14/2025]
Abstract
Background Macrophages are a promising target for therapeutics in various applications such as regenerative medicine and immunotherapy for cancer. Due to their plastic nature, macrophages can switch from a non-activated state to activated with the smallest environmental change. For macrophages to be effective in their respective applications, screening for phenotypic changes is necessary to elucidate the cell response to different delivery vehicles, vaccines, small molecules, and other stimuli. Methods We created a sensitive and dynamic high-throughput screening method for macrophages based on the activation of NF-κB. For this reporter, we placed an mRFP1 fluorescence gene under the control of an inflammatory promoter, which recruits NF-κB response elements to promote expression during the inflammatory response in macrophages. We characterized the inflammatory reporter based on key markers of an inflammatory response in macrophages including TNF-α cytokine release and immunostaining for inflammatory and non-inflammatory cell surface markers. We compared gene delivery and inflammation of several clinically relevant viral vehicles and commercially available non-viral vehicles. Statistical analysis between groups was performed with a one-way ANOVA with post-hoc Tukey's test. Results The reporter macrophages demonstrated a dynamic range after LPS stimulation with an EC50 of 0.61 ng/mL that was highly predictive of TNF-α release. Flow cytometry revealed heterogeneity between groups but confirmed population level shifts in pro-inflammatory markers. Finally, we demonstrated utility of the reporter by showing divergent effects with various leading gene delivery vehicles. Discussion This screening technique developed here provides a dynamic, high-throughput screening technique for determining inflammatory response by mouse macrophages to specific stimuli. The method presented here provides insight into the inflammatory response in mouse macrophages to different viral and non-viral gene delivery methods and provides a tool for high-throughput screening of novel vehicles. Supplementary Information The online version contains supplementary material available at 10.1186/s44330-025-00030-x.
Collapse
Affiliation(s)
- Allie Ivy
- Department of Biomedical Engineering, University of Arkansas, 120 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
| | - Shelby N. Bess
- Department of Biomedical Engineering, University of Arkansas, 120 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
| | - Shilpi Agrawal
- Department of Biomedical Engineering, University of Arkansas, 120 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
| | - Varun Kochar
- Department of Biomedical Engineering, University of Arkansas, 120 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
| | - Abbey L. Stokes
- Department of Biomedical Engineering, University of Arkansas, 120 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
| | - Timothy J. Muldoon
- Department of Biomedical Engineering, University of Arkansas, 120 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR USA
| | - Christopher E. Nelson
- Department of Biomedical Engineering, University of Arkansas, 120 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR USA
| |
Collapse
|
4
|
Cojocaru AI, Kefi K, Masson JD, Tiret L, Relaix F, Taglietti V. Forskolin treatment enhances muscle regeneration and shows therapeutic potential with limitations in Duchenne muscular dystrophy. Skelet Muscle 2025; 15:12. [PMID: 40329365 PMCID: PMC12057055 DOI: 10.1186/s13395-025-00381-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 04/20/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Duchenne Muscular Dystrophy (DMD) is a progressive neuromuscular disorder characterized by impaired muscle repair. Forskolin (FSK), an adenylyl cyclase activator, has shown potential in enhancing muscle regeneration and limiting muscle stem cell senescence. This study aimed to evaluate the effects of FSK on muscle repair, fibrosis, inflammation, and long-term muscle function in DMD using a preclinical rat model. METHODS BaCl2-induced muscle injury was performed on 6-month-old DMD (R-DMDdel52) and wild-type (WT) rats. FSK was supplied via short-term and long-term administration. Muscle tissues were harvested 14 days post-injury for histological analysis, including hematoxylin and eosin and Sirius red staining. Immunofluorescence was used to assess fibroadipogenic progenitors (FAPs), regeneration, muscle stem cells, and macrophage phenotypes. Moreover, we performed a study by chronically administering FSK to DMD rats from 1 to 7 months of age, either intraperitoneally (IP) or subcutaneously (SC). Functional assessments included grip strength test, in vivo muscle force measurements, plethysmography and electrocardiograms. Post-sacrifice, Tibialis anterior, diaphragm and heart tissues were histologically analyzed, to evaluate muscle architecture, fibrosis, and histopathological indices. RESULTS FSK treatment significantly improved muscle histology and reduced fibrosis in both uninjured and injured DMD muscles by decreasing the number of FAPs. Long-term FSK treatment in the acute injury model enhanced muscle regeneration, increased MuSC proliferation, and reduced senescence. FSK also modulated inflammation by reducing pro-inflammatory macrophages and promoting a shift to a restorative phenotype. However, despite these histological improvements, FSK treatment from 1 to 7 months resulted in limited functional benefits and worsened ventricular histology in the heart. CONCLUSIONS FSK shows promising results in improving muscle regeneration and reducing fibrosis in DMD, but concerns remain regarding its limited chronic functional benefits and potential adverse effects on cardiac tissue. Our results highlight the need for optimized adenylyl cyclase activators for therapeutic use in DMD patients.
Collapse
Affiliation(s)
| | - Kaouthar Kefi
- Univ Paris-Est Créteil, INSERM, U955 IMRB, F-94010, Créteil, France
| | | | - Laurent Tiret
- Univ Paris-Est Créteil, INSERM, U955 IMRB, F-94010, Créteil, France
- École Nationale Vétérinaire d'Alfort, U955 IMRB, F-94700, Maisons-Alfort, France
| | - Frederic Relaix
- Univ Paris-Est Créteil, INSERM, U955 IMRB, F-94010, Créteil, France.
- École Nationale Vétérinaire d'Alfort, U955 IMRB, F-94700, Maisons-Alfort, France.
- EFS, U955 IMRB, F-94010, Créteil, France.
- AP-HP, Hopital Mondor, Service d' histologie, 94010, Creteil, France.
| | | |
Collapse
|
5
|
Ye K, Li J, Huo Z, Xu J, Dai Q, Qiao K, Cao Y, Yan L, Liu W, Hu Y, Xu L, Su R, Zhu Y, Mi Y. Down-regulating HDAC2-LTA4H pathway ameliorates renal ischemia-reperfusion injury. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167889. [PMID: 40324735 DOI: 10.1016/j.bbadis.2025.167889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 05/02/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND The activation of histone deacetylase 2 (HDAC2) is the main pathogenesis of acute kidney injury (AKI), one of the leading causes of end-stage kidney disease. However, the regulatory role of HDAC2 upregulation on inflammation in AKI is still unclear. RESULTS In this study, we found that treatment with HDAC2 inhibitor BRD6688 could mitigate the degree of mesangial sclerosis, interstitial infiltration and tubular atrophy, reduce the concentration of blood urea nitrogen (BUN) and serum creatinine (Scr), improve the proliferation, anti-apoptotic, anti-oxidative stress and angiogenesis effects of renal cells. Our results mainly indicated that renal HDAC2 activity was increased by casein kinase 2 (CK2) in renal ischemia reperfusion (I/R) models, and HDAC2 genetic ablation in HREpiC cells suppressed the leukotriene B4 (LTB4) production. Renal leukotriene A4 hydrolase (LTA4H) activity was increased in AKI mice in a HDAC2-dependent manner. LTB4 could induce monocytes to differentiate into M1 macrophages, while BRD6688 could suppress this effect and force the M1 macrophages polarize to M2 macrophages. CONCLUSION Inhibition of HDAC2 activities by BRD6688 could suppress the progression of renal I/R injury through the regulation of LTA4H and macrophage polarization.
Collapse
Affiliation(s)
- Kai Ye
- Clinical School of the Second People's Hospital, Tianjin Medical University, Tianjin 300192, China; Tianjin Institute of Hepatology, Tianjin Second People's Hospital, Tianjin 300192, China; Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin 300192, China
| | - Jixuan Li
- Department of internal medicine, Tianjin Fourth Hospital, Tianjin 300222, China
| | - Zhixiao Huo
- Clinical School of the Second People's Hospital, Tianjin Medical University, Tianjin 300192, China; Tianjin Institute of Hepatology, Tianjin Second People's Hospital, Tianjin 300192, China; Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin 300192, China
| | - Jian Xu
- Clinical School of the Second People's Hospital, Tianjin Medical University, Tianjin 300192, China; Tianjin Institute of Hepatology, Tianjin Second People's Hospital, Tianjin 300192, China; Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin 300192, China
| | - Qinghai Dai
- Clinical School of the Second People's Hospital, Tianjin Medical University, Tianjin 300192, China; Tianjin Institute of Hepatology, Tianjin Second People's Hospital, Tianjin 300192, China; Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin 300192, China
| | - Kunyan Qiao
- Clinical School of the Second People's Hospital, Tianjin Medical University, Tianjin 300192, China; Tianjin Institute of Hepatology, Tianjin Second People's Hospital, Tianjin 300192, China; Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin 300192, China
| | - Yu Cao
- Clinical School of the Second People's Hospital, Tianjin Medical University, Tianjin 300192, China; Tianjin Institute of Hepatology, Tianjin Second People's Hospital, Tianjin 300192, China; Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin 300192, China
| | - Lihua Yan
- Clinical School of the Second People's Hospital, Tianjin Medical University, Tianjin 300192, China; Tianjin Institute of Hepatology, Tianjin Second People's Hospital, Tianjin 300192, China; Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin 300192, China
| | - Wei Liu
- Clinical School of the Second People's Hospital, Tianjin Medical University, Tianjin 300192, China; Tianjin Institute of Hepatology, Tianjin Second People's Hospital, Tianjin 300192, China; Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin 300192, China
| | - Yue Hu
- Clinical School of the Second People's Hospital, Tianjin Medical University, Tianjin 300192, China; Tianjin Institute of Hepatology, Tianjin Second People's Hospital, Tianjin 300192, China; Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin 300192, China
| | - Liang Xu
- Clinical School of the Second People's Hospital, Tianjin Medical University, Tianjin 300192, China; Tianjin Institute of Hepatology, Tianjin Second People's Hospital, Tianjin 300192, China; Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin 300192, China.
| | - Rui Su
- Clinical School of the Second People's Hospital, Tianjin Medical University, Tianjin 300192, China; Tianjin Institute of Hepatology, Tianjin Second People's Hospital, Tianjin 300192, China; Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin 300192, China.
| | - Yu Zhu
- Department of Clinical Laboratory, The Third Central Hospital of Tianjin, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin 300170, China
| | - Yuqiang Mi
- Clinical School of the Second People's Hospital, Tianjin Medical University, Tianjin 300192, China; Tianjin Institute of Hepatology, Tianjin Second People's Hospital, Tianjin 300192, China; Tianjin Integrated Traditional Chinese and Western Medicine Institute of Infectious Diseases, Tianjin 300192, China.
| |
Collapse
|
6
|
Zuo W, Peng J, Guo W, Wu J. Unraveling the protein-metabolite network of sarcopenia in plasma: A large-scale Mendelian randomization study. Arch Gerontol Geriatr 2025; 132:105788. [PMID: 40009978 DOI: 10.1016/j.archger.2025.105788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/08/2025] [Accepted: 02/17/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND Some plasma molecules may have an effect on sarcopenia, but it is not fully understood. We aimed to comprehensively explore the causal effects of plasma proteins and metabolites on sarcopenia traits, and to unravel their network. METHODS A two-sample Mendelian randomization design was adopted. The levels of 4,907 plasma proteins from 35,559 Icelanders, and 1,400 plasma metabolites from 8,299 Europeans, were set as exposures. Low handgrip strength, appendicular lean mass, and usual walking pace from Europeans were set as outcomes. Inverse-variance weighted (IVW) and four other methods, along with sensitivity analyzes, were performed to estimate the causal effects. Enrichment and pathway analyzes were conducted to present their characteristics. IVW was used to estimate the bidirectional relationships between sarcopenia-related proteins and metabolites, and to visualize them within a network. RESULTS We identified 76 relationships between proteins and sarcopenia traits. The absolute values of causal effects (βIVW) ranging from 0.01 to 0.35. IL2, AIF1, GDNF, CXCL13, LRRTM3, and SLPI were the top six proteins ranked by causal effects. Additionally, 22 relationships between metabolites and sarcopenia traits were identified, with absolute values of βIVW ranging from 0.02 to 0.22. Sulfate and serine/pyruvate ratio had the highest values. The network diagram showed some key nodes, such as ISOC1, GSTA1, tryptophan and 5α-androstan-3α,17β-diol monosulfate. CONCLUSIONS This work unraveled a molecular network of sarcopenia in plasma for the first time and identified some key proteins and metabolites. It may help to understand the mechanisms of sarcopenia, providing new insights for predicting, diagnosing and treating sarcopenia.
Collapse
Affiliation(s)
- Wenhang Zuo
- Center of Gerontology and Geriatrics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, PR China
| | - Jin Peng
- Center of Gerontology and Geriatrics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, PR China
| | - Wen Guo
- Center of Gerontology and Geriatrics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, PR China
| | - Jinhui Wu
- Center of Gerontology and Geriatrics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, PR China.
| |
Collapse
|
7
|
Wiyono AV, Ardinal AP, Raharjo PP. Unraveling the significance of innate inflammation in vascular disease. Int Rev Immunol 2025:1-16. [PMID: 40255209 DOI: 10.1080/08830185.2025.2489346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 02/06/2025] [Accepted: 03/31/2025] [Indexed: 04/22/2025]
Abstract
Atheroma formation is initiated by the activation of endothelial and smooth muscle cells, as well as immune cells, including neutrophils, lymphocytes, monocytes, macrophages, and dendritic cells. Monocytes, macrophages, and neutrophils are the innate immune cells that provide a rapid initial line of defence against vascular disease. These cells have a short lifespan and cannot retain memories, making them potential therapeutic targets for the inflammatory process associated with atherosclerosis. In addition, macrophages comprise the majority of vessel wall infiltrates and are, therefore, implicated in all stages of atherosclerosis progression. Neutrophils are the most common type of leukocyte found in circulation, and their high levels of matrix-degrading protease explain their significance in fibrous cap destabilization. However, the activation of immune cells becomes more complex by various microenvironmental stimuli and cytokines, which ultimately transform immune cells into their pro-inflammatory state. Different types of macrophage subsets with distinct functions in inflammation, such as M1 macrophages, cause an increase in pro-inflammatory cytokines and produce reactive oxygen species and nitric oxide, further worsening the disease. This review aims to shed light on immune-mediated inflammation in cardiovascular disease by focusing on the role of macrophage subsets in vascular inflammation and plaque stability, as well as the interaction between neutrophils and monocyte-macrophages.
Collapse
Affiliation(s)
- Alice Valeria Wiyono
- Faculty of Life Sciences & Medicine, King's College London, London, UK
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Rumah Sakit Umum Pusat Hasan Sadikin, Bandung, Indonesia
| | | | - Pradana Pratomo Raharjo
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Rumah Sakit Umum Pusat Hasan Sadikin, Bandung, Indonesia
| |
Collapse
|
8
|
Chen M, Chen J, Liu Y, Wang X, Yao M, Chen J, Zhang J, Huang Q. Senescent Macrophages Promote Age-Related Revascularization Impairment by Increasing Antiangiogenic VEGF-A165B Expression. Aging Cell 2025:e70059. [PMID: 40243169 DOI: 10.1111/acel.70059] [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: 08/20/2024] [Revised: 03/05/2025] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Peripheral arterial disease is a common vascular disease in the elderly. Therapeutic revascularization, including angiogenic and arteriogenic therapy, is a promising treatment approach for peripheral arterial disease. However, the progress of clinical trials is not ideal, possibly due to insufficiency of preclinical models, such as not taking into account the effect of aging on vascular regeneration. Macrophages are crucial in angiogenesis and arteriogenesis. The aging microenvironment typically makes recruited monocytes and macrophages more susceptible to senescence. However, the feature of macrophages in ischemic hindlimb muscle of old individuals and their underlying role remains unclear. In this study, we reveal that macrophages of ischemic skeletal muscle in old mice are more senescent and proinflammatory. By transplanting macrophages into mice following hindlimb ischemia, we find senescent macrophages inhibit revascularization. Mechanistically, these senescent macrophages induce endothelial dysfunction via increasing vascular endothelial growth factor A-165B (VEGF-A165B) expression and secretion, and eventually impair revascularization. Notably, plasma VEGF-A165B levels are elevated in old patients with PAD and positively associated with a lower ankle brachial index (ABI). Our study suggests that targeting the senescent macrophages presents an avenue to improve age-related revascularization damage.
Collapse
Affiliation(s)
- Minghong Chen
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Junyu Chen
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu Liu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of General and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xuerui Wang
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Meilian Yao
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jing Chen
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jian Zhang
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qun Huang
- Department of Child Health Care, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| |
Collapse
|
9
|
Corsini A, Perticarini L, Palermi S, Bettinsoli P, Marchini A. Re-Evaluating Platelet-Rich Plasma Dosing Strategies in Sports Medicine: The Role of the "10 Billion Platelet Dose" in Optimizing Therapeutic Outcomes-A Narrative Review. J Clin Med 2025; 14:2714. [PMID: 40283544 PMCID: PMC12027823 DOI: 10.3390/jcm14082714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 03/24/2025] [Accepted: 04/01/2025] [Indexed: 04/29/2025] Open
Abstract
Platelet-rich plasma (PRP) therapy is increasingly recognized as a promising treatment for musculoskeletal disorders, including osteoarthritis (OA), tendinopathy, and muscle injuries. This narrative review synthesizes the current literature to evaluate the efficacy of PRP, with a focus on platelet dosing strategies, leukocyte composition, and preparation protocols. Evidence suggests that optimal therapeutic outcomes are achieved when platelet doses exceed 3.5 billion per injection, with cumulative doses of 10-12 billion across multiple treatments. In intra-articular applications, leukocyte-poor PRP (LP-PRP), characterized by reduced neutrophil content, demonstrates superior efficacy compared to leukocyte-rich PRP (LR-PRP). However, its effectiveness in tendon and muscle regeneration remains a subject of debate. Preliminary data suggest that the inclusion of peripheral blood mononuclear cells (PBMNCs) may enhance PRP efficacy, though robust clinical trials are required to confirm these findings. Furthermore, red blood cell contamination and pre-activation have been identified as detrimental to PRP effectiveness, highlighting the need for standardized preparation protocols. This review emphasizes the importance of tailoring PRP formulations to patient-specific factors and musculoskeletal conditions. Future research should focus on refining PRP preparation techniques, identifying optimal leukocyte compositions, and establishing standardized guidelines to enhance clinical outcomes.
Collapse
Affiliation(s)
| | - Loris Perticarini
- Fondazione Poliambulanza Istituti Ospedalieri, 25125 Brescia, Italy;
| | - Stefano Palermi
- Department of Medicine and Surgery, UniCamillus-Saint Camillus International University of Health Sciences, 00187 Rome, Italy;
| | | | | |
Collapse
|
10
|
Borok MJ, Zaidan L, Relaix F. Transposon expression and repression in skeletal muscle. Mob DNA 2025; 16:18. [PMID: 40217332 PMCID: PMC11992895 DOI: 10.1186/s13100-025-00352-1] [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: 12/31/2024] [Accepted: 03/13/2025] [Indexed: 04/14/2025] Open
Abstract
Transposons and their derivatives make up a major proportion of the human genome, but they are not just relics of ancient genomes. They can still be expressed, potentially affecting the transcription of adjacent genes, and can sometimes even contribute to their coding sequence. Active transposons can integrate into new sites in the genome, potentially modifying the expression of nearby loci and leading to genetic disorders. In this review, we highlight work exploring the expression of transposons in skeletal muscles and transcriptional regulation by the KRAB-ZFP/KAP1/SETDB1 complex. We next focus on specific cases of transposon insertion causing phenotypic variation and distinct muscular dystrophies, as well as the implication of transposon expression in immune myopathies. Finally, we discuss the dysregulation of transposons in facioscapulohumeral dystrophy and aging.
Collapse
Affiliation(s)
- Matthew J Borok
- University Paris-Est Créteil, INSERM U955 IMRB, Créteil, 94010, France.
| | - Louai Zaidan
- University Paris-Est Créteil, INSERM U955 IMRB, Créteil, 94010, France
| | - Frederic Relaix
- University Paris-Est Créteil, INSERM U955 IMRB, Créteil, 94010, France.
- École Nationale Vétérinaire d'Alfort U955 IMRB, Maisons-Alfort, 94700, France.
- EFS IMRB, Créteil, 94010, France.
- Assistance Publique-Hôpitaux de Paris, Hôpital Mondor, Service d'Histologie, Créteil, 94010, France.
| |
Collapse
|
11
|
Jiang H, Zhao Y, Lv B, Jiang Z, Cao J. Oxygen-supplying nanotherapeutics for bacterial septic arthritis via hypoxia-relief-enhanced antimicrobial and anti-inflammatory phototherapy. J Control Release 2025; 380:1043-1057. [PMID: 39986475 DOI: 10.1016/j.jconrel.2025.02.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/15/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Bacterial septic arthritis, an inflammatory disease caused by bacterial infections, is often accompanied by the emergence of multidrug-resistant bacteria, making therapeutic treatment a formidable challenge. With the emergence of antibiotic resistance, research on antimicrobial photodynamic therapy (aPDT) has regained attention. However, insufficient singlet oxygen production due to the hypoxia in the infection microenvironment is recognized as a key limiting the efficacy of aPDT. Hence, in this study, we designed a bacteria-targeted liposomal nanoplatform (MCPL) to alleviate hypoxia in the infectious microenvironment and enhance aPDT for bacterial septic arthritis. The nanoplatform was developed by integrating the phototherapeutic agent CyI and small-sized Pt NPs into thermosensitive liposomes, with modification of maltohexose on the liposome surface. In vitro and in vivo studies showed that MCPL could specifically target bacterial cell membranes and be thermally activated to catalytically convert endogenous hydrogen peroxide (H2O2) in the septic joint, supplementing local O2 reservoirs. This, in turn, supplies additional substrate pools for photodynamic conversion into reactive oxygen species (ROS) with bacterial toxicity. Furthermore, the antibacterial mechanism revealed that MCPL can regulate the HIF-1α and NF-κB signaling pathways in immune cells, acting as an effective modulator of antioxidant and anti-inflammatory pathways in both macrophages and neutrophils. This study demonstrates that MCPL could not induce bacterial multidrug resistance and provide as an innovative approach for treating bacterial septic arthritis.
Collapse
Affiliation(s)
- Huimei Jiang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Yifan Zhao
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Bai Lv
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Zijia Jiang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Jie Cao
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266071, China..
| |
Collapse
|
12
|
Zhang X, Zhong Y, Rajabi S. Polyphenols and post-exercise muscle damage: a comprehensive review of literature. Eur J Med Res 2025; 30:260. [PMID: 40205487 PMCID: PMC11983803 DOI: 10.1186/s40001-025-02506-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 03/24/2025] [Indexed: 04/11/2025] Open
Abstract
Recent research highlights the significant role of polyphenols in alleviating post-exercise muscle damage, thus positioning them as a valuable nutritional strategy for athletes and fitness enthusiasts. Polyphenols, naturally occurring bioactive compounds abundant in fruits, vegetables, tea, wine, and other plant-based foods, are recognized for their potent antioxidant and anti-inflammatory properties. This dual mechanism is critical for combating oxidative stress and inflammation-two factors that intensify during vigorous physical activity and contribute to muscle soreness and damage. Among various polyphenols, compounds like quercetin have particularly emerged as effective agents for promoting muscle recovery and enhancing exercise performance. These protective effects are facilitated through several mechanisms, including the modulation of inflammatory pathways, acceleration of muscle repair processes, and enhancement of mitochondrial function, all of which bolster overall muscle health. As ongoing studies yield deeper insights, the potential of polyphenols to enhance athletic performance and overall health will become increasingly substantiated, leading towards their strategic incorporation into exercise nutrition protocols. Therefore, we reviewed relevant studies in order to show how efficient polyphenols can be in reducing muscle fatigue and damage and what are the exact mechanisms.
Collapse
Affiliation(s)
- Xiaofeng Zhang
- Yinchuan University of Energy, Yinchuan, 750105, Ningxia, China
| | - Yuanfu Zhong
- School of Sports and Health Sciences, Xiangsihu College of Guangxi Minzu University, Nanning, 530000, Guangxi, China.
| | - Sogand Rajabi
- Department of Cellular and Molecular Biology, Islamic Azad University, Sirjan Branch, Sirjan, Iran.
| |
Collapse
|
13
|
Lithgow H, Gibson L, Wilson R, Guthrie N, Ingram-Sills L, Clifford T, Ross M. An ultra-endurance event leads to changes in circulating regulatory T-cells, CD4+ naïve and CD8+ effector memory T-cells in the 48 h post-race recovery period. Eur J Appl Physiol 2025; 125:1129-1138. [PMID: 39604770 PMCID: PMC11950060 DOI: 10.1007/s00421-024-05677-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
PURPOSE Exercise is known to acutely affect T-lymphocyte populations in the peripheral blood, which is intensity- and duration-dependent. However, effects of longer duration endurance exercise (>5 h) on T-cells in the days following are unknown. The aim of this study was to investigate the circulating T-cell changes that occur in response to an ultra-endurance event, which may provide insight into the inflammatory response to ultra-endurance exercise. METHODS Ten individuals (m = 7, f = 3) completing an Ironman 70.3 event volunteered for the study. Peripheral blood samples were taken 1-2 days pre-race (PRE-RACE), and 1 day (RACE + 1) and 2 days (RACE + 2) post-race, with circulating T-cells enumerated by flow cytometry (total CD3+, CD4+ and CD8+ T-cells, regulatory T-cells [CD4+CD25+CD127-; TREG], naïve [CD27+CD45RA+; NA], central memory [CD27+CD45RA-; CM], effector memory [CD27-CD45RA-; EM], and effector memory CD45RA+ [CD27-CD45RA+; EMRA]). RESULTS There were no changes in total CD3+, CD4+ and CD8+ T-cells. TREG RACE + 1 was significantly higher compared to PRE-RACE, as were the proportion of CD4+ NA cells and CD8+ CM cells at RACE + 2; CD8+ EM cells fell at RACE + 2 (absolute counts and proportion). CONCLUSION In conclusion, the ultra-endurance event evoked T-cell changes over the 48 h recovery period, with an increase in T-cells that regulate the immune response, and a reduction in circulating EM T-cells, most likely trafficked to sites of tissue damage and inflammation.
Collapse
Affiliation(s)
- Hannah Lithgow
- School of Energy, Geoscience, Infrastructure and Society, Institute of Life and Earth Sciences, Heriot-Watt University, JMF7, Edinburgh, EH14 4AS, UK
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, UK
| | - Laura Gibson
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, UK
| | - Russell Wilson
- School of Energy, Geoscience, Infrastructure and Society, Institute of Life and Earth Sciences, Heriot-Watt University, JMF7, Edinburgh, EH14 4AS, UK
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, UK
| | - Neil Guthrie
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, UK
| | | | - Tom Clifford
- Institute of Cellular Medicine, Newcastle University, Newcastle, UK
- School of Sport, Exercise, and Health Sciences, Loughborough University, Loughborough, UK
| | - Mark Ross
- School of Energy, Geoscience, Infrastructure and Society, Institute of Life and Earth Sciences, Heriot-Watt University, JMF7, Edinburgh, EH14 4AS, UK.
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, UK.
| |
Collapse
|
14
|
TIAN Y, BU H, WANG T, YANG D, ZHANG W, LIU T, ZHANG L, HUO Z. Efficacy of electro-acupuncture at "Weizhong" (BL40) on macrophage polarization in rats with injured lumbar multifidus. J TRADIT CHIN MED 2025; 45:335-347. [PMID: 40151120 PMCID: PMC11955755 DOI: 10.19852/j.cnki.jtcm.20220419.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/17/2022] [Indexed: 03/29/2025]
Abstract
OBJECTIVE To investigate the anti-inflammatory effect of electroacupuncture in rats with bupivacaine-induced lumbar multifidus injury and its underlying regulatory mechanism on macrophage polarization. METHODS A total of seventy-two Sprague-Dawley male rats were randomly divided into control, model, and electroacupuncture groups. Forty-eight rats categorized in model groups were injected 0.5% bupivacaine (BPVC) into the lumbar multifidus at the L4-L5 segment. Rats in the electroacupuncture groups received the intervention for 1, 2, 3 and 5 d, respectively. The degree of macrophage infiltration and change of M1/M2 polarization were observed based on hematoxylin and eosin staining, immunohistochemistry and immunofluorescence to evaluate the anti-inflammatory effect of electroacupuncture. Meanwhile, exosomal miRNA-sequencing and bioinformatics analysis predicted the pathways and biological processes related to inflammatory response and macrophage polarization regulated by electroacupuncture intervention. RESULTS BPVC injection induced the infiltration of local macrophages at the L4-L5 segment of lumbar multifidus. Comparison of mean IOD values with 2 d and 5 d post injury revealed the highest expression of CD68+ macrophages on day 3 post injury by immunohistochemistry. (P < 0.001, P < 0.001, respectively). Compared with the model group, the cell counts of iNOs+ CD68+ M1-macrophages were lower in the electroacupuncture group, while the positive percent of CD163+ CD206+ M2-macrophages was higher in the electroacupuncture group, on day 3 after BPVC injection (P < 0.001, P < 0.001, respectively). Moreover, the results of sequencing and bioinformatic analysis suggested that exosomal miRNAs were involved in the EA regulating macrophage polarization. CONCLUSIONS Electroacupuncture can promote macrophage polarization to reduce inflammation following lumbar multifidus muscular injury.
Collapse
Affiliation(s)
- Yuan TIAN
- 1 Department of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
- 2 Department of Acupuncture-Moxibustion and Tuina, the Second Affiliated Hospital of Baotou Medical College, Inner Mongolia Autonomous Region, Baotou 014030, China
| | - He BU
- 2 Department of Acupuncture-Moxibustion and Tuina, the Second Affiliated Hospital of Baotou Medical College, Inner Mongolia Autonomous Region, Baotou 014030, China
| | - Tieshan WANG
- 3 Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | | | - Wei ZHANG
- 7 Department of Pathology, the First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Inner Mongolia 014010, China
| | - Tong LIU
- 5 the Fifth College of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou 510006, China
- 6 Department of Acupuncture and Rehabilitation, Guangdong Second Hospital of Traditional Chinese Medicine, Guangzhou 510095, China
| | - Li ZHANG
- 1 Department of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zejun HUO
- 8 Department of Chinese Medicine, Peking University 3rd Hospital, Beijing 100191, China
| |
Collapse
|
15
|
Luk H, Levitt DE, Appell CR, Jiwan NC. Resistance exercise-induced circulating factors influence the damaged skeletal muscle proteome in a sex-dependent manner. Physiol Rep 2025; 13:e70291. [PMID: 40223391 PMCID: PMC11994862 DOI: 10.14814/phy2.70291] [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: 08/26/2024] [Revised: 03/13/2025] [Accepted: 03/13/2025] [Indexed: 04/15/2025] Open
Abstract
Muscle recovery after damage is mediated by circulating factors and intracellular signaling pathways. Our previous studies have demonstrated that resistance exercise (RE)-induced circulating factors elicited sex-differential responses in damaged muscle. However, the global effects of these circulating factors on damaged muscle are largely understudied. We examined the differential effects of RE-induced circulating factors and sex on the damaged muscle proteome. Damaged vastus lateralis muscle from 3 men and 3 women from a parent study were analyzed. Participants completed 2 identical bouts of unilateral eccentric knee extensions immediately followed by either upper body RE to induce circulating factors (EXE) or 20-min seated rest (CON). Muscle biopsies collected from the damaged leg at 24 h were used. 900 proteins were identified by LC-MS/MS analysis. Ingenuity Pathway Analysis was used to detect activation prediction using z-scores for functional and pathway analyses. In men, 79 proteins were downregulated and 15 were upregulated in EXE versus CON. These differentially expressed proteins were associated with immunological and inflammatory signaling pathways. Biological functions of the differentially expressed proteins in EXE vs. CON in men include inactivating acute inflammatory signaling, neutrophil extracellular trap signaling, ROS production, and activating IL-12 signaling. These results underline that RE-induced circulating factors have a sex-specific effect on the damaged muscle proteome, where immune signaling is altered in men but not women. Given that the immune response is critical for recovery from muscle damage, these results highlight the potential role of RE-induced circulating factors that could be essential in mediating muscle recovery.
Collapse
Affiliation(s)
- Hui‐Ying Luk
- Department of Kinesiology and Sport ManagementTexas Tech UniversityLubbockTexasUSA
| | - Danielle E. Levitt
- Department of Kinesiology and Sport ManagementTexas Tech UniversityLubbockTexasUSA
| | - Casey R. Appell
- Department of Kinesiology and Sport ManagementTexas Tech UniversityLubbockTexasUSA
| | - Nigel C. Jiwan
- Department of Kinesiology and Sport ManagementTexas Tech UniversityLubbockTexasUSA
| |
Collapse
|
16
|
Wang Y, Wang X, Alabdullatif S, Homma ST, Alekseyev YO, Zhou L. Expansion and pathogenic activation of skeletal muscle-resident macrophages in mdx5cv/Ccr2-/- mice. Proc Natl Acad Sci U S A 2025; 122:e2410095122. [PMID: 40067893 PMCID: PMC11929395 DOI: 10.1073/pnas.2410095122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 02/10/2025] [Indexed: 03/25/2025] Open
Abstract
Infiltrating macrophages contribute to muscle dystrophic changes in Duchenne muscular dystrophy (DMD). In a DMD mouse model, mdx5cv mice, CC chemokine receptor type 2 (CCR2) deficiency diminishes Ly6Chi macrophage infiltration by blocking blood Ly6Chi inflammatory monocyte recruitment. This is accompanied by transient improvement of muscle damage, fibrosis, and regeneration. The benefit, however, is lost after the expansion of intramuscular Ly6Clo macrophages. To address the mechanisms underlying the Ly6Clo macrophage expansion, we compared mdx5cv/Nur77-/- and mdx5cv/Ccr2-/-/Nur7-/- mice with mdx5cv and mdx5cv/Ccr2-/- mice, respectively, and found no evidence to suggest Ly6Clo monocyte recruitment by dystrophic muscles. Single-cell RNA sequencing analysis and Flt3cre/Rosa26LSL-YFP-based lineage tracing of macrophage origins demonstrated the expansion and pathogenic activation of muscle resident macrophages in CCR2-deficient mdx5cv mice. The expansion was associated with increased cell proliferation, which appeared induced by colony-stimulating factor-1 (CSF-1) derived from fibro/adipogenic progenitors (FAPs). Our study establishes a pathogenic role for skeletal muscle resident macrophages and supports a regulatory role of FAPs in stimulating the expansion of resident macrophages in the DMD mouse model when the inflammatory macrophage infiltration is inhibited.
Collapse
MESH Headings
- Animals
- Receptors, CCR2/genetics
- Receptors, CCR2/metabolism
- Receptors, CCR2/deficiency
- Mice
- Muscle, Skeletal/pathology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/immunology
- Macrophages/metabolism
- Macrophages/pathology
- Macrophages/immunology
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/immunology
- Mice, Inbred mdx
- Mice, Knockout
- Disease Models, Animal
- Male
Collapse
Affiliation(s)
- Yinhang Wang
- Department of Neurology, Hospital for Special Surgery, New York, NY10021
| | - Xingyu Wang
- Department of Neurology, Hospital for Special Surgery, New York, NY10021
| | - Salam Alabdullatif
- Department of Medicine, Single Cell Sequencing Core Facility, Boston University Chobanian and Avedisian School of Medicine, Boston, MA02118
| | - Sachiko T. Homma
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA02118
| | - Yuriy O. Alekseyev
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA02118
| | - Lan Zhou
- Department of Neurology, Hospital for Special Surgery, New York, NY10021
| |
Collapse
|
17
|
Basurto IM, Bandara GC, Boudreau RD, Shriver SB, Muhammad SA, Christ GJ, Caliari SR. Freeze-Dried Porous Collagen Scaffolds for the Repair of Volumetric Muscle Loss Injuries. ACS Biomater Sci Eng 2025; 11:1598-1611. [PMID: 39907689 PMCID: PMC11897937 DOI: 10.1021/acsbiomaterials.4c01601] [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: 08/30/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/06/2025]
Abstract
Volumetric muscle loss (VML) injuries are characterized by the traumatic loss of skeletal muscle, resulting in permanent damage to both tissue architecture and electrical excitability. To address this challenge, we previously developed a three-dimensional (3D) aligned collagen-glycosaminoglycan (CG) scaffold platform that supported in vitro myotube alignment and maturation. In this work, we assessed the ability of CG scaffolds to facilitate functional muscle recovery in a rat tibialis anterior (TA) model of VML. Functional muscle recovery was assessed following implantation of either nonconductive CG or electrically conductive CG-polypyrrole (PPy) scaffolds at 4, 8, and 12 weeks postinjury by in vivo electrical stimulation of the peroneal nerve. After 12 weeks, scaffold-treated muscles produced maximum isometric torque that was significantly greater than nontreated tissues. Histological analysis further supported these reparative outcomes with evidence of regenerating muscle fibers at the material-tissue interface in scaffold-treated tissues that were not observed in nonrepaired muscles. Scaffold-treated muscles possessed higher numbers of M1 and M2 macrophages at the injury, while conductive CG-PPy scaffold-treated muscles showed significantly higher levels of neovascularization as indicated by the presence of pericytes and endothelial cells, suggesting a persistent wound repair response not observed in nontreated tissues. Finally, only tissues treated with nonconductive CG scaffolds displayed neurofilament staining similar to native muscle, further corroborating isometric contraction data. Together, these findings show that both conductive and nonconductive CG scaffolds can facilitate improved skeletal muscle function and endogenous cellular repair, highlighting their potential use as therapeutics for VML injuries.
Collapse
Affiliation(s)
- Ivan M. Basurto
- Department
of Biomedical Engineering, Department of Chemical Engineering, Department of Orthopedic
Surgery, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Geshani C. Bandara
- Department
of Biomedical Engineering, Department of Chemical Engineering, Department of Orthopedic
Surgery, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Ryann D. Boudreau
- Department
of Biomedical Engineering, Department of Chemical Engineering, Department of Orthopedic
Surgery, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Sydney B. Shriver
- Department
of Biomedical Engineering, Department of Chemical Engineering, Department of Orthopedic
Surgery, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Samir A. Muhammad
- Department
of Biomedical Engineering, Department of Chemical Engineering, Department of Orthopedic
Surgery, University of Virginia, Charlottesville, Virginia 22903, United States
| | - George J. Christ
- Department
of Biomedical Engineering, Department of Chemical Engineering, Department of Orthopedic
Surgery, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Steven R. Caliari
- Department
of Biomedical Engineering, Department of Chemical Engineering, Department of Orthopedic
Surgery, University of Virginia, Charlottesville, Virginia 22903, United States
| |
Collapse
|
18
|
Day J, Louis C, Swiderski K, Stock A, Wong H, Yao W, Liu B, Nadesapillai S, Lynch GS, Wicks IP. Periarticular myositis and muscle fibrosis are cytokine-dependent complications of inflammatory arthritis. JCI Insight 2025; 10:e179928. [PMID: 40036069 PMCID: PMC11981620 DOI: 10.1172/jci.insight.179928] [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: 02/02/2024] [Accepted: 02/25/2025] [Indexed: 03/06/2025] Open
Abstract
The deleterious consequences of chronic synovitis on cartilage, tendon, and bone in rheumatoid arthritis (RA) are well described. In contrast, its effects on periarticular skeletal muscle are under-studied. Furthermore, while TNF inhibition is an effective therapy for RA synovitis, it exacerbates fibrosis in muscle injury models. We aimed to investigate whether myositis and muscle fibrosis are features of inflammatory arthritis and evaluate whether targeted RA therapies influence these disease features. Periarticular muscle was analyzed in murine models of poly- and monoarticular inflammatory arthritis: serum transfer-induced arthritis, collagen-induced arthritis, K/BxN, and antigen-induced arthritis (AIA). Periarticular myositis and an increase in muscle fibroadipocyte progenitors (FAPs) were observed in all models, despite diverse arthritogenic mechanisms. Periarticular muscle fibrosis was observed from day 15 in AIA. Neither etanercept nor baricitinib suppressed periarticular myositis or subsequent fibrosis compared to vehicle, despite reducing arthritis. Notably, etanercept failed to prevent muscle fibrosis even when initiated early, but this was not linked to increased FAP survival or collagen production. Corroborating these data, radiographic and histological analyses revealed periarticular myositis in patients with RA. We conclude that periarticular myositis and fibrosis are under-recognized features of inflammatory arthritis. Targeted RA therapies may not prevent periarticular muscle sequelae, despite controlling arthritis.
Collapse
MESH Headings
- Animals
- Mice
- Fibrosis
- Myositis/pathology
- Myositis/etiology
- Myositis/drug therapy
- Myositis/immunology
- Muscle, Skeletal/pathology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/immunology
- Etanercept/pharmacology
- Etanercept/therapeutic use
- Arthritis, Experimental/complications
- Arthritis, Experimental/pathology
- Arthritis, Experimental/immunology
- Arthritis, Experimental/drug therapy
- Cytokines/metabolism
- Arthritis, Rheumatoid/complications
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/pathology
- Arthritis, Rheumatoid/immunology
- Male
- Disease Models, Animal
- Humans
- Purines/pharmacology
- Sulfonamides/pharmacology
- Sulfonamides/therapeutic use
- Azetidines/pharmacology
- Azetidines/therapeutic use
- Female
- Pyrazoles/pharmacology
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Jessica Day
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
- Department of Rheumatology, The Royal Melbourne Hospital, Victoria, Australia
| | - Cynthia Louis
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Kristy Swiderski
- Centre for Muscle Research, Department of Anatomy and Physiology, The University of Melbourne, Victoria
| | - Angus Stock
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Huon Wong
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Victoria, Australia
| | - Wentao Yao
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Victoria, Australia
| | - Bonnia Liu
- Department of Rheumatology, The Royal Melbourne Hospital, Victoria, Australia
- Department of Nuclear Medicine, The Royal Melbourne Hospital, Victoria, Australia
| | - Suba Nadesapillai
- Department of Rheumatology, The Royal Melbourne Hospital, Victoria, Australia
- Department of Nuclear Medicine, The Royal Melbourne Hospital, Victoria, Australia
| | - Gordon S. Lynch
- Centre for Muscle Research, Department of Anatomy and Physiology, The University of Melbourne, Victoria
| | - Ian P. Wicks
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
- Department of Rheumatology, The Royal Melbourne Hospital, Victoria, Australia
| |
Collapse
|
19
|
Messing M, Theret M, Hughes MR, Wu J, Syed OH, Li FF, Li Y, Rossi FMV, McNagny KM. Type-2 innate signals are dispensable for skeletal muscle regeneration and pathology linked to Duchenne muscular dystrophy. EMBO Rep 2025; 26:1406-1421. [PMID: 39900735 PMCID: PMC11894123 DOI: 10.1038/s44319-025-00383-y] [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: 04/28/2024] [Revised: 01/12/2025] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
Immune responses play an integral role in skeletal muscle regeneration. In the genetically inherited muscle disease Duchenne muscular dystrophy (DMD), muscle regeneration is disrupted, leading to chronic inflammation, fibrosis, and early mortality. Previously, it has been suggested that type-2 innate immune cells, particularly eosinophils and their production of IL-4, play an essential role in effective muscle regeneration after acute injury. We here re-investigate the role of eosinophils in skeletal muscle repair using mice deficient in eosinophils (ΔdblGATA), or deficient in IL-4R/IL-13R signaling through STAT6 (Stat6-/-). We show that neither deficiency has an impact on skeletal muscle regeneration in response to acute injury as quantified by fiber size, immune cell infiltration, or muscle-resident stem cell proliferation. We also investigate the role of STAT6 signaling in mdx:Stat6-/- mice, a model of DMD and, again, find that ablation of STAT6 signaling has no effect on the rate or severity of fibrotic scar formation or disease progression. In contrast to previous models, our data suggest a negligible role for eosinophils and STAT6 signaling in skeletal muscle regeneration after acute or chronic injury.
Collapse
MESH Headings
- Animals
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/immunology
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Muscle, Skeletal/pathology
- Muscle, Skeletal/physiology
- Muscle, Skeletal/metabolism
- Regeneration
- STAT6 Transcription Factor/genetics
- STAT6 Transcription Factor/metabolism
- Mice
- Signal Transduction
- Eosinophils/metabolism
- Eosinophils/immunology
- Mice, Inbred mdx
- Immunity, Innate
- Disease Models, Animal
- Mice, Knockout
Collapse
Affiliation(s)
- Melina Messing
- School of Biomedical Engineering and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Marine Theret
- School of Biomedical Engineering and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Michael R Hughes
- School of Biomedical Engineering and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Jiaqi Wu
- School of Biomedical Engineering and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Omar Husain Syed
- School of Biomedical Engineering and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Fang Fang Li
- School of Biomedical Engineering and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Yicong Li
- School of Biomedical Engineering and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Fabio M V Rossi
- School of Biomedical Engineering and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.
| | - Kelly M McNagny
- School of Biomedical Engineering and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
20
|
Cai CW, Grey JA, Hubmacher D, Han WM. Biomaterial-Based Regenerative Strategies for Volumetric Muscle Loss: Challenges and Solutions. Adv Wound Care (New Rochelle) 2025; 14:159-175. [PMID: 38775429 PMCID: PMC11971559 DOI: 10.1089/wound.2024.0079] [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: 03/30/2024] [Accepted: 05/15/2024] [Indexed: 07/11/2024] Open
Abstract
Significance: Volumetric muscle loss (VML) is caused by the loss of significant amounts of skeletal muscle tissue. VML cannot be repaired by intrinsic regenerative processes, resulting in permanent loss of muscle function and disability. Current rehabilitative-focused treatment strategies lack efficacy and do not restore muscle function, indicating the need for the development of effective regenerative strategies. Recent Advances: Recent developments implicate biomaterial-based approaches for promoting muscle repair and functional restoration post-VML. Specifically, bioscaffolds transplanted in the injury site have been utilized to mimic endogenous cues of the ablated tissue to promote myogenic pathways, increase neo-myofiber synthesis, and ultimately restore contractile function to the injured unit. Critical Issues: Despite the development and preclinical testing of various biomaterial-based regenerative strategies, effective therapies for patients are not available. The unique challenges posed for biomaterial-based treatments of VML injuries, including its scalability and clinical applicability beyond small-animal models, impede progress. Furthermore, production of tissue-engineered constructs is technically demanding, with reproducibility issues at scale and complexities in achieving vascularization and innervation of large constructs. Future Directions: Biomaterial-based regenerative strategies designed to comprehensively address the pathophysiology of VML are needed. Considerations for clinical translation, including scalability and regulatory compliance, should also be considered when developing such strategies. In addition, an integrated approach that combines regenerative and rehabilitative strategies is essential for ensuring functional improvement.
Collapse
Affiliation(s)
- Charlene W. Cai
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Biology, The College of New Jersey, Ewing, New Jersey, USA
| | - Josh A. Grey
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Institute of Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Dirk Hubmacher
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Woojin M. Han
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Institute of Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
21
|
Fan X, Peng Y, Li B, Wang X, Liu Y, Shen Y, Liu G, Zheng Y, Deng Q, Liu J, Yang L. Liver-Secreted Extracellular Vesicles Promote Cirrhosis-Associated Skeletal Muscle Injury Through mtDNA-cGAS/STING Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410439. [PMID: 39804962 PMCID: PMC11884600 DOI: 10.1002/advs.202410439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/15/2024] [Indexed: 01/16/2025]
Abstract
Skeletal muscle atrophy (sarcopenia) is a serious complication of liver cirrhosis, and chronic muscle inflammation plays a pivotal role in its pathologenesis. However, the detailed mechanism through which injured liver tissues mediate skeletal muscle inflammatory injury remains elusive. Here, it is reported that injured hepatocytes might secrete mtDNA-enriched extracellular vesicles (EVs) to trigger skeletal muscle inflammation by activating the cGAS-STING pathway. Briefly, injured liver secreted increased amounts of EVs into circulation, which are then engulfed primarily by macrophages in skeletal muscle and subsequently induce cGAS-STING signaling and its-mediated inflammatory response in muscles. In contrast, suppression of hepatic EV secretion or STING signaling significantly alleviated cirrhosis-induced skeletal muscle inflammation and muscle atrophy in vivo. Circulating EVs from cirrhotic patients showed higher levels of mtDNA, and the levels of EV-mtDNA positively correlated with the severity of liver injury. In injured hepatocytes, mitochondrial damage promoted the release of cytosolic mtDNA and the subsequent secretion of mtDNA-enriched EVs. This study reveals that injured hepatocyte-derived EVs induce skeletal muscle inflammation via the mtDNA‒STING axis, while targeted blockade of liver EV secretion or STING signaling represents a potential therapeutic approach for preventing cirrhosis-associated skeletal muscle atrophy.
Collapse
Affiliation(s)
- Xiaoli Fan
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver DiseaseWest China HospitalSichuan UniversityChengdu610041China
| | - Yunke Peng
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver DiseaseWest China HospitalSichuan UniversityChengdu610041China
| | - Bo Li
- Department of RadiologyWest China HospitalSichuan UniversityChengdu610041China
| | - Xiaoze Wang
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver DiseaseWest China HospitalSichuan UniversityChengdu610041China
| | - Yifeng Liu
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver DiseaseWest China HospitalSichuan UniversityChengdu610041China
| | - Yi Shen
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver DiseaseWest China HospitalSichuan UniversityChengdu610041China
| | - Guofeng Liu
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver DiseaseWest China HospitalSichuan UniversityChengdu610041China
| | - Yanyi Zheng
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver DiseaseWest China HospitalSichuan UniversityChengdu610041China
| | - Qiaoyu Deng
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver DiseaseWest China HospitalSichuan UniversityChengdu610041China
| | - Jingping Liu
- NHC Key Laboratory of Transplant Engineering and ImmunologyCenter for Disease‐related Molecular NetworkWest China Hospital of Sichuan UniversityChengdu610041China
| | - Li Yang
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver DiseaseWest China HospitalSichuan UniversityChengdu610041China
| |
Collapse
|
22
|
Whitaker R, Sung S, Tylek T, Risser GE, O'Brien EM, Chua PE, Li T, Petrie RJ, Han L, Binder-Markey BI, Spiller KL. Effects of injury size on local and systemic immune cell dynamics in volumetric muscle loss. NPJ Regen Med 2025; 10:9. [PMID: 39939310 PMCID: PMC11822203 DOI: 10.1038/s41536-025-00397-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 01/30/2025] [Indexed: 02/14/2025] Open
Abstract
We took a systems approach to the analysis of macrophage phenotype in regenerative and fibrotic volumetric muscle loss outcomes in mice together with analysis of systemic inflammation and of other leukocytes in the muscle, spleen, and bone marrow. Differences in expression of macrophage phenotype markers occurred as early as day 1, persisted to at least day 28, and were associated with increased numbers of leukocytes in the muscle and bone marrow, increased pro-inflammatory marker expression in splenic macrophages, and changes in the levels of pro-inflammatory cytokines in the blood. The most prominent differences were in muscle neutrophils, which were much more abundant in fibrotic outcomes compared to regenerative outcomes at day 1 after injury. However, neutrophil depletion had little to no effect on macrophage phenotype or on muscle repair outcomes. Together, these results suggest that the entire system of immune cell interactions must be considered to improve muscle repair outcomes.
Collapse
Affiliation(s)
- Ricardo Whitaker
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Samuel Sung
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Tina Tylek
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Gregory E Risser
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Erin M O'Brien
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Phoebe Ellin Chua
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Thomas Li
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Ryan J Petrie
- Department of Biology, College of Arts & Sciences, Drexel University, Philadelphia, PA, USA
| | - Lin Han
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Benjamin I Binder-Markey
- Department of Physical Therapy & Rehabilitation Sciences, College of Nursing and Health Professions, Drexel University, Philadelphia, PA, USA
| | - Kara L Spiller
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA.
| |
Collapse
|
23
|
Zhai Z, Yang C, Yin W, Liu Y, Li S, Ye Z, Xie M, Song X. Engineered Strategies to Interfere with Macrophage Fate in Myocardial Infarction. ACS Biomater Sci Eng 2025; 11:784-805. [PMID: 39884780 DOI: 10.1021/acsbiomaterials.4c02061] [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: 02/01/2025]
Abstract
Myocardial infarction (MI), a severe cardiovascular condition, is typically triggered by coronary artery disease, resulting in ischemic damage and the subsequent necrosis of the myocardium. Macrophages, known for their remarkable plasticity, are capable of exhibiting a range of phenotypes and functions as they react to diverse stimuli within their local microenvironment. In recent years, there has been an increasing number of studies on the regulation of macrophage behavior based on tissue engineering strategies, and its regulatory mechanisms deserve further investigation. This review first summarizes the effects of key regulatory factors of engineered biomaterials (including bioactive molecules, conductivity, and some microenvironmental factors) on macrophage behavior, then explores specific methods for inducing macrophage behavior through tissue engineering materials to promote myocardial repair, and summarizes the role of macrophage-host cell crosstalk in regulating inflammation, vascularization, and tissue remodeling. Finally, we propose some future challenges in regulating macrophage-material interactions and tailoring personalized biomaterials to guide macrophage phenotypes.
Collapse
Affiliation(s)
- Zitong Zhai
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Chang Yang
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Wenming Yin
- Department of Neurology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Yali Liu
- Department of Neurology, Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong 528000, China
| | - Shimin Li
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Ziyi Ye
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Mingxiang Xie
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Xiaoping Song
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
- Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
24
|
Mohapatra A, Howard Z, Ernst JD. CCR2 recruits monocytes to the lung, while CX3CR1 modulates positioning of monocyte-derived CD11c pos cells in the lymph node during pulmonary tuberculosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.637199. [PMID: 39974908 PMCID: PMC11839135 DOI: 10.1101/2025.02.07.637199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Infection by Mycobacterium tuberculosis (Mtb) continues to cause more than 1 million deaths annually, due to pathogen persistence in lung macrophages and dendritic cells derived from blood monocytes. While accumulation of monocyte-derived cells in the Mtb-infected lung partially depends on the chemokine receptor CCR2, the other chemoattractant receptors regulating trafficking remain undefined. We used mice expressing knock-in/knockout reporter alleles of Ccr2 and Cx3cr1 to interrogate their expression and function in monocyte-derived populations of the lungs and draining mediastinal lymph nodes during Mtb infection. CCR2 and CX3CR1 expression varied across monocyte-derived subsets stratified by cell surface Ly6C expression in both organs. We found that expression of CCR2 predicted dependence of monocyte-derived cells on the receptor for lung and lymph node accumulation. CCR2-deficient mice were also observed to have worsened lung and lymph node Mtb burden. While CX3CR1 deficiency, alone or in combination with CCR2 deficiency, did not affect cell frequencies or lung Mtb control, its absence was associated with altered positioning of monocyte-derived dendritic cells in mediastinal lymph nodes. We found that combined loss of Ccr2 and Cx3cr1 also worsened Mtb control in the mediastinal lymph node, suggesting a rationale for the persistent expression of CX3CR1 among monocyte-derived cells in pulmonary tuberculosis. IMPORTANCE Mycobacterium tuberculosis is the respiratory pathogen responsible for the deadliest infectious disease worldwide. Susceptible humans exhibit ineffective immune responses, in which infected phagocytes are not able to eliminate the pathogen. Since recruited monocyte-derived cells serve as reservoirs for persistent infection, understanding how these phagocytes accumulate in the lung and why they are unable to eliminate Mtb can inform development of therapies that can synergize with antimicrobials to achieve faster and more durable Mtb elimination. Monocyte-derived cells express the chemokine receptors CCR2 and CX3CR1, but the role of the latter in Mtb infection remains poorly defined. The significance of our study is in elucidating the roles of these receptors in the trafficking of monocyte-derived cells in the infected lung and mediastinal lymph node. These data shed light on the host response in tuberculosis and in other pulmonary infections.
Collapse
|
25
|
Fennel ZJ, O'Connell RM, Drummond MJ. Macrophage immunometabolism: emerging targets for regrowth in aging muscle. Am J Physiol Endocrinol Metab 2025; 328:E186-E197. [PMID: 39763086 PMCID: PMC12079615 DOI: 10.1152/ajpendo.00403.2024] [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: 10/14/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/31/2025]
Abstract
The recovery from muscle atrophy is impaired with aging as characterized by improper muscle remodeling and sustained functional deficits. Age-related deficits in muscle regrowth are tightly linked with the loss of early pro-inflammatory macrophage responses and subsequent cellular dysregulation within the skeletal muscle niche. Macrophage inflammatory phenotype is regulated at the metabolic level, highlighting immunometabolism as an emerging strategy to enhance macrophage responses and restore functional muscle regrowth. Accordingly, metabolic targets with an emphasis on glycolytic, hypoxia, and redox-related pathways stand out for their role in promoting macrophage inflammation and enhancing muscle regrowth in aging. Here we highlight promising immuno-metabolic targets that could be leveraged to restore optimal pro-inflammatory macrophage function in aging and enhance muscle regrowth following muscular atrophy.
Collapse
Affiliation(s)
- Zachary J Fennel
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, United States
| | - Ryan M O'Connell
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, United States
| | - Micah J Drummond
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, United States
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
26
|
Steele LA, Hernaez Estrada B, Spiller KL. Effects of a Bioengineered Allogeneic Cellularized Construct (BACC) on Primary Human Macrophage Phenotype. Adv Healthc Mater 2025; 14:e2303044. [PMID: 38507713 DOI: 10.1002/adhm.202303044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/01/2024] [Indexed: 03/22/2024]
Abstract
The mechanisms behind the pro-healing effects of multicellular, bioengineered allogeneic cellularized constructs (BACC) are not known. Macrophages are key regulators of every phase of the wound healing process and the primary cells that mediate the response to biomaterials. It is hypothesized that cells within the BACC modulate macrophage behavior, which may contribute to the mechanism by which BACC promotes healing. To probe the influence of cells within the BACC compared to effects of the underlying collagen substrate, primary human macrophages are cultured in direct or indirect contact with BACC or with the same collagen substrate used in the BACC manufacturing. Macrophage phenotype is characterized over time via multiplex gene expression, protein secretion, multidimensional flow cytometry, and functional assays with fibroblasts and endothelial cells. The BACC causes macrophages to exhibit a predominately reparative phenotype over time compared to relevant collagen substrate controls, with multiple subpopulations expressing both pro-inflammatory and reparative markers. Conditioned media from macrophage-BACC co-cultures causes distinct effects on fibroblast and endothelial cell proliferation, migration, and network formation. Given the critical role of the reparative macrophage phenotype in wound healing, these results suggest that modulation of macrophage phenotype may be a critical part of the mechanisms behind BACC's pro-healing effects.
Collapse
Affiliation(s)
- Lindsay A Steele
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Beatriz Hernaez Estrada
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Kara L Spiller
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| |
Collapse
|
27
|
Stocks B, Quesada JP, Mozzicato AM, Jacob C, Jensen S, MacGregor KA, Bangsbo J, Zierath JR, Hostrup M, Deshmukh AS. Temporal dynamics of the interstitial fluid proteome in human skeletal muscle following exhaustive exercise. SCIENCE ADVANCES 2025; 11:eadp8608. [PMID: 39889004 PMCID: PMC11784852 DOI: 10.1126/sciadv.adp8608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 01/02/2025] [Indexed: 02/02/2025]
Abstract
The skeletal muscle interstitial space is the extracellular region around myofibers and mediates cross-talk between resident cell types. We applied a proteomic workflow to characterize the human skeletal muscle interstitial fluid proteome at rest and in response to exercise. Following exhaustive exercise, markers of skeletal muscle damage accumulate in the interstitial space followed by the appearance of immune cell-derived proteins. Among the proteins up-regulated after exercise, we identified cathelicidin-related antimicrobial peptide (CAMP) as a bioactive molecule regulating muscle fiber development. Treatment with the bioactive peptide derivative of CAMP (LL-37) resulted in the growth of larger C2C12 skeletal muscle myotubes. Phosphoproteomics revealed that LL-37 activated pathways central to muscle growth and proliferation, including phosphatidylinositol 3-kinase, AKT serine/threonine kinase 1, mitogen-activated protein kinases, and mammalian target of rapamycin. Our findings provide a proof of concept that the interstitial fluid proteome is quantifiable via microdialysis sampling in vivo. These data highlight the importance of cellular communication in the adaptive response to exercise.
Collapse
Affiliation(s)
- Ben Stocks
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Julia Prats Quesada
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anthony M. Mozzicato
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Carolina Jacob
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Simone Jensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kirstin A. MacGregor
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Jens Bangsbo
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Juleen R. Zierath
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Morten Hostrup
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Atul S. Deshmukh
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
28
|
Acheson J, Joanisse S, Sale C, Hodson N. Recycle, repair, recover: the role of autophagy in modulating skeletal muscle repair and post-exercise recovery. Biosci Rep 2025; 45:1-30. [PMID: 39670455 DOI: 10.1042/bsr20240137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 12/14/2024] Open
Abstract
Skeletal muscle is a highly plastic tissue that can adapt relatively rapidly to a range of stimuli. In response to novel mechanical loading, e.g. unaccustomed resistance exercise, myofibers are disrupted and undergo a period of ultrastructural remodeling to regain full physiological function, normally within 7 days. The mechanisms that underpin this remodeling are believed to be a combination of cellular processes including ubiquitin-proteasome/calpain-mediated degradation, immune cell infiltration, and satellite cell proliferation/differentiation. A relatively understudied system that has the potential to be a significant contributing mechanism to repair and recovery is the autophagolysosomal system, an intracellular process that degrades damaged and redundant cellular components to provide constituent metabolites for the resynthesis of new organelles and cellular structures. This review summarizes our current understanding of the autophagolysosomal system in the context of skeletal muscle repair and recovery. In addition, we also provide hypothetical models of how this system may interact with other processes involved in skeletal muscle remodeling and provide avenues for future research to improve our understanding of autophagy in human skeletal muscle.
Collapse
Affiliation(s)
- Jordan Acheson
- Department of Sport and Exercise Sciences, Manchester Metropolitan University, Institute of Sport, Manchester, U.K
| | - Sophie Joanisse
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, U.K
| | - Craig Sale
- Department of Sport and Exercise Sciences, Manchester Metropolitan University, Institute of Sport, Manchester, U.K
| | - Nathan Hodson
- Department of Sport and Exercise Sciences, Manchester Metropolitan University, Institute of Sport, Manchester, U.K
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
29
|
Hanna M, Akabawy AMA, Khalifa MM, Elbaset MA, Imam RA, Seddiek H. Intracellular iron accumulation throughout the progression of sepsis influences the phenotype and function of activated macrophages in renal tissue damage. Front Physiol 2025; 16:1430946. [PMID: 39949667 PMCID: PMC11821637 DOI: 10.3389/fphys.2025.1430946] [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: 05/10/2024] [Accepted: 01/06/2025] [Indexed: 02/16/2025] Open
Abstract
Sepsis, the most common cause of acute kidney injury, remains a major socioeconomic burden. A dysregulated immune response leads to progressive organ dysfunction. Although numerous inflammatory pathways were described, most are still vague and need to be studied in terms of the mechanisms to improve the therapeutic intervention. We tackled the relationship between intracellular iron overload and macrophage polarization within 6, 24, and 72 h of sepsis induction. In our study, sepsis-induced kidney injury was caused by using the cecal ligation and puncture (CLP) model. Our results indicated severe renal tissue damage with a progressive increase in serum BUN and creatinine with architectural tissue damage and positive PAS staining. There was increased expression of CD8+ CD68+ M1 macrophage markers with upregulation of iNOS and co-expression of CD163+. Alternatively, Arg1+ Fizz1+ M2 macrophage markers were downregulated with increased iNOS/Arg1 ratio. TFR1, cubilin, and DMT1, as iron transport systems, were increased compared to sham but were significant after 72 h, while ZIP8 showed no significant change. There was a correlation between iron overload and M1 macrophage polarization with CD163+ phenotype, together with fibrotic changes. The intracellular iron overload with downregulation of ferritin was strongly related to macrophage polarization that was exaggerated at 72 h. Finally, early introduced therapy to target free iron during sepsis is a proposed novel solution for protecting the renal tissue from acute injury due to macrophage activation that may end up with chronic kidney injury, if not mortality.
Collapse
Affiliation(s)
- Mira Hanna
- Department of Medical Physiology, Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt
| | - Ahmed M. A. Akabawy
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Mohamed Mansour Khalifa
- Department of Medical Physiology, Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt
- Department of Medical Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Marawan Abd Elbaset
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt
| | - Reda Abdelnasser Imam
- Department of Anatomy and Embryology, Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt
| | - Hanan Seddiek
- Department of Medical Physiology, Faculty of Medicine, Kasr Al-Ainy, Cairo University, Cairo, Egypt
| |
Collapse
|
30
|
Jones AE, Rios A, Ibrahimovic N, Chavez C, Bayley NA, Ball AB, Hsieh WY, Sammarco A, Bianchi AR, Cortez AA, Graeber TG, Hoffmann A, Bensinger SJ, Divakaruni AS. The metabolic cofactor Coenzyme A enhances alternative macrophage activation via MyD88-linked signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.03.28.587096. [PMID: 38585887 PMCID: PMC10996702 DOI: 10.1101/2024.03.28.587096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Metabolites and metabolic co-factors can shape the innate immune response, though the pathways by which these molecules adjust inflammation remain incompletely understood. Here we show that the metabolic cofactor Coenzyme A (CoA) enhances IL-4 driven alternative macrophage activation [m(IL-4)] in vitro and in vivo. Unexpectedly, we found that perturbations in intracellular CoA metabolism did not influence m(IL-4) differentiation. Rather, we discovered that exogenous CoA provides a weak TLR4 signal which primes macrophages for increased receptivity to IL-4 signals and resolution of inflammation via MyD88. Mechanistic studies revealed MyD88-linked signals prime for IL-4 responsiveness, in part, by reshaping chromatin accessibility to enhance transcription of IL-4-linked genes. The results identify CoA as a host metabolic co-factor that influences macrophage function through an extrinsic TLR4-dependent mechanism, and suggests that damage-associated molecular patterns (DAMPs) can prime macrophages for alternative activation and resolution of inflammation.
Collapse
Affiliation(s)
- Anthony E Jones
- Departments of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Amy Rios
- Departments of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Neira Ibrahimovic
- Departments of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Carolina Chavez
- Departments of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
- Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Nicholas A Bayley
- Departments of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Andréa B Ball
- Departments of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Wei Yuan Hsieh
- Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alessandro Sammarco
- Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Amber R Bianchi
- Departments of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Angel A Cortez
- Departments of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Thomas G Graeber
- Departments of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Alexander Hoffmann
- Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Steven J Bensinger
- Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ajit S Divakaruni
- Departments of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
- Lead contact
| |
Collapse
|
31
|
Kiesworo K, Agius T, Macarthur MR, Lambelet M, Lyon A, Zhang J, Turiel G, Fan Z, d’Almeida S, Uygun K, Yeh H, Déglise S, de Bock K, Mitchell SJ, Ocampo A, Allagnat F, Longchamp A. Nicotinamide mononucleotide restores impaired metabolism, endothelial cell proliferation and angiogenesis in old sedentary male mice. iScience 2025; 28:111656. [PMID: 39868046 PMCID: PMC11763620 DOI: 10.1016/j.isci.2024.111656] [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: 07/03/2024] [Revised: 10/15/2024] [Accepted: 12/18/2024] [Indexed: 01/28/2025] Open
Abstract
Aging is accompanied by a decline in neovascularization potential and increased susceptibility to ischemic injury. Here, we confirm the age-related impaired neovascularization following ischemic leg injury and impaired angiogenesis. The age-related deficits in angiogenesis arose primarily from diminished EC proliferation capacity, but not migration or VEGF sensitivity. Aged EC harvested from the mouse skeletal muscle displayed a pro-angiogenic gene expression phenotype, along with considerable changes in metabolic genes. Metabolomics analysis and 13C glucose tracing revealed impaired ATP production and blockade in glycolysis and TCA cycle in late passage HUVECs, which occurred at nicotinamide adenine dinucleotide (NAD⁺)-dependent steps, along with NAD+ depletion. Supplementation with nicotinamide mononucleotide (NMN), a precursor of NAD⁺, enhances late-passage EC proliferation and sprouting angiogenesis from aged mice aortas. Taken together, our study illustrates the importance of NAD+-dependent metabolism in the maintenance of EC proliferation capacity with age, and the therapeutic potential of NAD precursors.
Collapse
Affiliation(s)
- Kevin Kiesworo
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Thomas Agius
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Michael R. Macarthur
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Ludwig Princeton Branch, Princeton University, Princeton, NJ, USA
| | - Martine Lambelet
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Arnaud Lyon
- Transplantation Centre and Transplantation Immunopathology Laboratory, Department of Medicine, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Jing Zhang
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Guillermo Turiel
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Zheng Fan
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | | | - Korkut Uygun
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Heidi Yeh
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sébastien Déglise
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Katrien de Bock
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Sarah J. Mitchell
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Ludwig Princeton Branch, Princeton University, Princeton, NJ, USA
| | - Alejandro Ocampo
- Department of Biomedical Sciences, Lausanne University (UNIL), Lausanne, Switzerland
| | - Florent Allagnat
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Alban Longchamp
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
32
|
Runyan CE, Luo L, Welch LC, Lu Z, Chen F, Schleck MJ, Nafikova RA, Grant RA, Aillon RP, Senkow KJ, Bunyan EG, Plodzeen WT, Abdala-Valencia H, Weiss C, Dada LA, Thorp EB, Sznajder JI, Chandel NS, Misharin AV, Budinger GRS. Tissue-resident skeletal muscle macrophages promote recovery from viral pneumonia-induced sarcopenia in normal aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.09.631996. [PMID: 39868236 PMCID: PMC11760773 DOI: 10.1101/2025.01.09.631996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Sarcopenia, which diminishes lifespan and healthspan in the elderly, is commonly exacerbated by viral pneumonia, including influenza and COVID-19. In a study of influenza A pneumonia in mice, young mice fully recovered from sarcopenia, while older mice did not. We identified a population of tissue-resident skeletal muscle macrophages that form a spatial niche with satellite cells and myofibers in young mice but are lost with age. Mice with a gain-of-function mutation in the Mertk receptor maintained this macrophage-myofiber interaction during aging and fully recovered from influenza-induced sarcopenia. In contrast, deletion of Mertk in macrophages or loss of Cx3cr1 disrupted this niche, preventing muscle regeneration. Heterochronic parabiosis did not restore the niche in old mice. These findings suggest that age-related loss of Mertk in muscle tissue-resident macrophages disrupts the cellular signaling necessary for muscle regeneration after viral pneumonia, offering a potential target to mitigate sarcopenia in aging.
Collapse
Affiliation(s)
- Constance E Runyan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Simpson Querrey Lung Institute for Translational Sciences. Northwestern University. Chicago, IL, USA
| | - Lucy Luo
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Simpson Querrey Lung Institute for Translational Sciences. Northwestern University. Chicago, IL, USA
| | - Lynn C Welch
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Simpson Querrey Lung Institute for Translational Sciences. Northwestern University. Chicago, IL, USA
| | - Ziyan Lu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Simpson Querrey Lung Institute for Translational Sciences. Northwestern University. Chicago, IL, USA
| | - Fei Chen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Simpson Querrey Lung Institute for Translational Sciences. Northwestern University. Chicago, IL, USA
| | - Maxwell J Schleck
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Simpson Querrey Lung Institute for Translational Sciences. Northwestern University. Chicago, IL, USA
| | - Radmila A Nafikova
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Simpson Querrey Lung Institute for Translational Sciences. Northwestern University. Chicago, IL, USA
| | - Rogan A Grant
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Simpson Querrey Lung Institute for Translational Sciences. Northwestern University. Chicago, IL, USA
| | - Raul Piseaux Aillon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Simpson Querrey Lung Institute for Translational Sciences. Northwestern University. Chicago, IL, USA
| | - Karolina J Senkow
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Simpson Querrey Lung Institute for Translational Sciences. Northwestern University. Chicago, IL, USA
| | - Elsie G Bunyan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Simpson Querrey Lung Institute for Translational Sciences. Northwestern University. Chicago, IL, USA
| | - William T Plodzeen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Simpson Querrey Lung Institute for Translational Sciences. Northwestern University. Chicago, IL, USA
| | - Hiam Abdala-Valencia
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Simpson Querrey Lung Institute for Translational Sciences. Northwestern University. Chicago, IL, USA
| | - Craig Weiss
- Department of Neuroscience, Northwestern University Feinberg School of Medicine. Chicago, IL, USA
| | - Laura A Dada
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Simpson Querrey Lung Institute for Translational Sciences. Northwestern University. Chicago, IL, USA
| | - Edward B Thorp
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jacob I Sznajder
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Simpson Querrey Lung Institute for Translational Sciences. Northwestern University. Chicago, IL, USA
| | - Navdeep S Chandel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Simpson Querrey Lung Institute for Translational Sciences. Northwestern University. Chicago, IL, USA
| | - Alexander V Misharin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Simpson Querrey Lung Institute for Translational Sciences. Northwestern University. Chicago, IL, USA
| | - G R Scott Budinger
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Simpson Querrey Lung Institute for Translational Sciences. Northwestern University. Chicago, IL, USA
| |
Collapse
|
33
|
Alibhai FJ, Tobin SW. Characterization of Age-Dependent Changes in Skeletal Muscle Repair and Regeneration Using a Mouse Model of Acute Muscle Injury. Methods Mol Biol 2025; 2857:169-180. [PMID: 39348065 DOI: 10.1007/978-1-0716-4128-6_16] [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: 10/01/2024]
Abstract
Acute skeletal muscle injury initiates a process of necrosis, debris clearance, and ultimately tissue regeneration via myogenesis. While skeletal muscle stem cells (MuSCs) are responsible for populating the proliferative myogenic progenitor pool to fuel muscle repair, recruited and resident immune cells have a central role in the regulation of muscle regeneration via the execution of phagocytosis and release of soluble factors that act directly on MuSCs to regulate myogenic differentiation. Therefore, the timing of MuSC proliferation and differentiation is closely linked to the populations and behaviors of immune cells present within skeletal muscle. This has important implications for aging and muscle repair, as systemic changes in immune system function contribute to a decline in muscle regenerative capacity. Here, we present adapted protocols for the isolation of mononuclear cells from skeletal muscles for the quantification of immune cell populations using flow cytometry. We also describe a cardiotoxin skeletal muscle injury protocol and detail the expected outcomes including immune cell infiltration to the injured sites and formation of new myocytes. As immune cell function is substantially influenced by aging, we extend these approaches and outcomes to aged mice.
Collapse
Affiliation(s)
| | - Stephanie W Tobin
- Department of Biology, Trent University, Peterborough, ON, Canada.
- Environmental and Life Sciences Graduate Program, Trent University, Peterborough, ON, Canada.
| |
Collapse
|
34
|
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
|
35
|
Krajewska-Węglewicz L, Felczak P, Dorobek M. Effects of Aging on Orbicularis Oculi Muscle Strength and Ultrastructure in Dermatochalasis: A Pilot Study. J Clin Med 2024; 14:162. [PMID: 39797244 PMCID: PMC11721777 DOI: 10.3390/jcm14010162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/13/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
Background: Age-related changes to the orbicularis oculi muscle include impaired eyelid function, such as lagophthalmos, alterations in tear film dynamics, and aesthetic changes like wrinkles, festoons, and the descent of soft tissue. To date, the structural and functional changes that would comprehensively increase our understanding of orbicularis aging have not been analyzed. This study aims to investigate functional outcomes using surface electromyography and correlate them with ultrastructural changes in orbicularis during aging. Methods: This study enrolled 26 patients aged 37 to 78 years with a clinical diagnosis of dermatochalasis. Patients were divided into two age groups (<60 years; ≥60 years). Ultrastructural and electromyographical examinations were performed, and the electromyographical signals were correlated with the ultrastructural damage in the orbicularis. Results: This study revealed significantly lower values of average voluntary contraction and RMS of the surface electromyography signals in the older age group compared to the younger age group (p = 0.029 and p = 0.045, respectively). There was no statistically significant association between age and muscle damage (χ2(2) = 2.86, p > 0.05). There was no correlation between average voluntary contraction and the degree of ultrastructural damage in both groups (Spearman's coefficient equaled 0.06923 and 0.64366, respectively). Conclusions: sEMG measurements are valuable for monitoring age-related functional changes in the orbicularis. Aging diminishes the functional capacity of the orbicularis, as evidenced by reduced contraction strength. This study, the first to compare ultrastructural and electromyographical changes in the orbicularis among dermatochalasis patients of different ages, finds that ultrastructural damage to muscle fibers is not directly responsible for the contraction strength decline.
Collapse
Affiliation(s)
- Larysa Krajewska-Węglewicz
- Department of Ophthalmology, National Institute of Medicine of the Ministry of Interior and Administration, 02-507 Warsaw, Poland
| | - Paulina Felczak
- Department of Neuropathology, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
| | - Małgorzata Dorobek
- Department of Neurology, National Institute of Medicine of the Ministry of Interior and Administration, 02-507 Warsaw, Poland
| |
Collapse
|
36
|
Yin K, Zhang C, Deng Z, Wei X, Xiang T, Yang C, Chen C, Chen Y, Luo F. FAPs orchestrate homeostasis of muscle physiology and pathophysiology. FASEB J 2024; 38:e70234. [PMID: 39676717 PMCID: PMC11647758 DOI: 10.1096/fj.202400381r] [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: 02/17/2024] [Revised: 10/26/2024] [Accepted: 11/26/2024] [Indexed: 12/17/2024]
Abstract
As a common clinical manifestation, muscle weakness is prevalent in people with mobility disorders. Further studies of muscle weakness have found that patients with muscle weakness present with persistent muscle inflammation, loss of muscle fibers, fat infiltration, and interstitial fibrosis. Therefore, we propose the concept of muscle microenvironment homeostasis, which explains the abnormal pathological changes in muscles through the imbalance of muscle microenvironment homeostasis. And we identified an interstitial progenitor cell FAP during the transition from normal muscle microenvironment homeostasis to muscle microenvironment imbalance caused by muscle damage diseases. As a kind of pluripotent stem cell, FAPs do not participate in myogenic differentiation, but can differentiate into fibroblasts, adipocytes, osteoblasts, and chondrocytes. As a kind of mesenchymal progenitor cell, it is involved in the generation of extracellular matrix, regulate muscle regeneration, and maintain neuromuscular junction. However, the muscle microenvironment is disrupted by the causative factors, and the abnormal activities of FAPs eventually contribute to the complex pathological changes in muscles. Targeting the mechanisms of these muscle pathological changes, we have identified appropriate signaling targets for FAPs to improve and even treat muscle damage diseases. In this review, we propose the construction of muscle microenvironmental homeostasis and find the key cells that cause pathological changes in muscle after homeostasis is broken. By studying the mechanism of abnormal differentiation and apoptosis of FAPs, we found a strategy to inhibit the abnormal pathological changes in muscle damage diseases and improve muscle regeneration.
Collapse
Affiliation(s)
- Kai Yin
- Department of OrthopedicsSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| | - Chengmin Zhang
- Department of OrthopedicsSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| | - Zihan Deng
- Department of OrthopedicsSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| | - Xiaoyu Wei
- Department of OrthopedicsSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| | - Tingwen Xiang
- Department of OrthopedicsSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| | - Chuan Yang
- Department of Biomedical Materials ScienceThird Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| | - Can Chen
- Department for Combat Casualty Care TrainingTraining Base for Army Health Care, Army Medical University (Third Military Medical University)ChongqingPeople's Republic of China
| | - Yueqi Chen
- Department of OrthopedicsSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| | - Fei Luo
- Department of OrthopedicsSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| |
Collapse
|
37
|
Chinvattanachot G, Rivas D, Duque G. Mechanisms of muscle cells alterations and regeneration decline during aging. Ageing Res Rev 2024; 102:102589. [PMID: 39566742 DOI: 10.1016/j.arr.2024.102589] [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: 07/13/2024] [Revised: 10/27/2024] [Accepted: 11/14/2024] [Indexed: 11/22/2024]
Abstract
Skeletal muscles are essential for locomotion and body metabolism regulation. As muscles age, they lose strength, elasticity, and metabolic capability, leading to ineffective motion and metabolic derangement. Both cellular and extracellular alterations significantly influence muscle aging. Satellite cells (SCs), the primary muscle stem cells responsible for muscle regeneration, become exhausted, resulting in diminished population and functionality during aging. This decline in SC function impairs intercellular interactions as well as extracellular matrix production, further hindering muscle regeneration. Other muscle-resident cells, such as fibro-adipogenic progenitors (FAPs), pericytes, and immune cells, also deteriorate with age, reducing local growth factor activities and responsiveness to stress or injury. Systemic signaling, including hormonal changes, contributes to muscle cellular catabolism and disrupts muscle homeostasis. Collectively, these cellular and environmental components interact, disrupting muscle homeostasis and regeneration in advancing age. Understanding these complex interactions offers insights into potential regenerative strategies to mitigate age-related muscle degeneration.
Collapse
Affiliation(s)
- Guntarat Chinvattanachot
- Department of Orthopedics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand; Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
| | - Daniel Rivas
- Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Gustavo Duque
- Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, QC, Canada; Dr. Joseph Kaufmann Chair in Geriatric Medicine, Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
38
|
Saunders AAE, Thomson RE, Goodman CA, Anderson RL, Gregorevic P. Striated muscle: an inadequate soil for cancers. Cancer Metastasis Rev 2024; 43:1511-1527. [PMID: 38995522 PMCID: PMC11554797 DOI: 10.1007/s10555-024-10199-2] [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: 03/18/2024] [Accepted: 07/01/2024] [Indexed: 07/13/2024]
Abstract
Many organs of the body are susceptible to cancer development. However, striated muscles-which include skeletal and cardiac muscles-are rarely the sites of primary cancers. Most deaths from cancer arise due to complications associated with the development of secondary metastatic tumours, for which there are few effective therapies. However, as with primary cancers, the establishment of metastatic tumours in striated muscle accounts for a disproportionately small fraction of secondary tumours, relative to the proportion of body composition. Examining why primary and metastatic cancers are comparatively rare in striated muscle presents an opportunity to better understand mechanisms that can influence cancer cell biology. To gain insights into the incidence and distribution of muscle metastases, this review presents a definitive summary of the 210 case studies of metastasis in muscle published since 2010. To examine why metastases rarely form in muscles, this review considers the mechanisms currently proposed to render muscle an inhospitable environment for cancers. The "seed and soil" hypothesis proposes that tissues' differences in susceptibility to metastatic colonization are due to differing host microenvironments that promote or suppress metastatic growth to varying degrees. As such, the "soil" within muscle may not be conducive to cancer growth. Gaining a greater understanding of the mechanisms that underpin the resistance of muscles to cancer may provide new insights into mechanisms of tumour growth and progression, and offer opportunities to leverage insights into the development of interventions with the potential to inhibit metastasis in susceptible tissues.
Collapse
Affiliation(s)
- Alastair A E Saunders
- Centre for Muscle Research, and Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Rachel E Thomson
- Centre for Muscle Research, and Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Craig A Goodman
- Centre for Muscle Research, and Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Robin L Anderson
- Metastasis Research Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria, Australia
- Peter MacCallum Cancer Centre, Parkville, Victoria, Australia
| | - Paul Gregorevic
- Centre for Muscle Research, and Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, 3010, Australia.
- Department of Neurology, The University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
39
|
Castrogiovanni P, Sanfilippo C, Imbesi R, Lazzarino G, Li Volti G, Tibullo D, Vicario N, Parenti R, Giuseppe L, Barbagallo I, Alanazi AM, Vecchio M, Cappello F, Musumeci G, Di Rosa M. Skeletal muscle of young females under resistance exercise exhibits a unique innate immune cell infiltration profile compared to males and elderly individuals. J Muscle Res Cell Motil 2024; 45:171-190. [PMID: 38578562 DOI: 10.1007/s10974-024-09668-6] [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: 11/23/2023] [Accepted: 03/12/2024] [Indexed: 04/06/2024]
Abstract
Muscle damage resulting from physical activities such as exercise triggers an immune response crucial for tissue repair and recovery. This study investigates the immune cell profiles in muscle biopsies of individuals engaged in resistance exercise (RE) and explores the impact of age and sex on the immune response following exercise-induced muscle damage. Microarray datasets from muscle biopsies of young and old subjects were analyzed, focusing on the gene expression patterns associated with immune cell activation. Genes were compared with immune cell signatures to reveal the cellular landscape during exercise. Results show that the most significant modulated gene after RE was Folliculin Interacting Protein 2 (FNIP2) a crucial regulator in cellular homeostasis. Moreover, the transcriptome was stratified based on the expression of FNIP2 and the 203 genes common to the groups obtained based on sex and age. Gene ontology analysis highlighted the FLCN-FNIP1-FNIP2 complex, which exerts as a negative feedback loop to Pi3k-Akt-mTORC1 pathway. Furthermore, we highlighted that the young females exhibit a distinct innate immune cell activation signature compared to males after a RE session. Specifically, young females demonstrate a notable overlap with dendritic cells (DCs), M1 macrophages, M2 macrophages, and neutrophils, while young males overlap with M1 macrophages, M2 macrophages, and motor neurons. Interestingly, in elderly subjects, both sexes display M1 macrophage activation signatures. Comparison of young and elderly signatures reveals an increased M1 macrophage percentage in young subjects. Additionally, common genes were identified in both sexes across different age groups, elucidating biological functions related to cell remodeling and immune activation. This study underscores the intricate interplay between sex, age, and the immune response in muscle tissue following RE, offering potential directions for future research. Nevertheless, there is a need for further studies to delve deeper and confirm the dynamics of immune cells in response to exercise-induced muscle damage.
Collapse
Affiliation(s)
- Paola Castrogiovanni
- Department of Biomedical and Biotechnological Sciences, Anatomy, Histology and Movement Sciences Section, School of Medicine, University of Catania, Catania, 95125, Italy
| | - Cristina Sanfilippo
- Neurologic Unit, Department of Medical, Surgical Sciences and Advanced Technologies, AOU "Policlinico-San Marco", University of Catania, Via Santa Sofia n.78, Sicily, GF, Ingrassia, Catania, 95100, Italy
| | - Rosa Imbesi
- Department of Biomedical and Biotechnological Sciences, Anatomy, Histology and Movement Sciences Section, School of Medicine, University of Catania, Catania, 95125, Italy
| | - Giacomo Lazzarino
- UniCamillus-Saint Camillus International University of Health Sciences, Via di Sant'Alessandro 8, Rome, 00131, Italy
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, Section of Biochemistry, University of Catania, Catania, 95123, Italy
| | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, Section of Biochemistry, University of Catania, Catania, 95123, Italy
| | - Nunzio Vicario
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 95123, Italy
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 95123, Italy
| | - Lazzarino Giuseppe
- Department of Biomedical and Biotechnological Sciences, Section of Biochemistry, University of Catania, Catania, 95123, Italy
| | - Ignazio Barbagallo
- Department of Biomedical and Biotechnological Sciences, Section of Biochemistry, University of Catania, Catania, 95123, Italy
| | - Amer M Alanazi
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Michele Vecchio
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 95124, Italy
| | - Francesco Cappello
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, Palermo, 90127, Italy
- Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, 90139, Italy
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Anatomy, Histology and Movement Sciences Section, School of Medicine, University of Catania, Catania, 95125, Italy
| | - Michelino Di Rosa
- Department of Biomedical and Biotechnological Sciences, Anatomy, Histology and Movement Sciences Section, School of Medicine, University of Catania, Catania, 95125, Italy.
| |
Collapse
|
40
|
Lee SH, Kim SY, Gwon YG, Lee C, Kim C, Cho IH, Kim TW, Choi &BK. Recombinant ADAMTS1 promotes muscle cell differentiation and alleviates muscle atrophy by repressing NOTCH1. BMB Rep 2024; 57:539-545. [PMID: 39567207 PMCID: PMC11693603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/13/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
A disintegrin and metalloproteinase with thrombospondin motifs 1 (ADAMTS1) plays crucial roles in various biological processes, including myogenesis, by modulating the neurogenic locus notch homolog protein 1 (NOTCH1) signaling pathway. However, the mechanisms through which ADAMTS1 regulates myogenesis remain unclear. In this study, we generated recombinant ADAMTS1 mutants and determined their effects on muscle cell differentiation, focusing on the regulation of NOTCH1 signaling. Treatment of C2C12 cells with recombinant ADAMTS1 protein enhanced muscle cell differentiation. Meanwhile, ADAM10 treatment inhibited muscle differentiation through the activation of NOTCH1 cleavage. Recombinant ADAMTS1 reversed ADAM10-induced muscle cell atrophy by suppressing NOTCH1 activation and downregulating its target gene. Recombinant ADAMTS1 also alleviated dexamethasoneinduced muscle atrophy in a mouse model. In summary, our findings suggest that recombinant ADAMTS1 promotes muscle regeneration by suppressing NOTCH1 and highlight the potential of recombinant ADAMTS1 proteins in the treatment of muscle wasting disease. [BMB Reports 2024; 57(12): 539-545].
Collapse
Affiliation(s)
| | | | | | | | | | | | - Tae-Won Kim
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, Daejeon 34131, Korea
| | | |
Collapse
|
41
|
Rehak L, Giurato L, Monami M, Meloni M, Scatena A, Panunzi A, Manti GM, Caravaggi CMF, Uccioli L. The Immune-Centric Revolution Translated into Clinical Application: Peripheral Blood Mononuclear Cell (PBMNC) Therapy in Diabetic Patients with No-Option Critical Limb-Threatening Ischemia (NO-CLTI)-Rationale and Meta-Analysis of Observational Studies. J Clin Med 2024; 13:7230. [PMID: 39685690 DOI: 10.3390/jcm13237230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/04/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Chronic limb-threatening ischemia (CLTI), the most advanced form of peripheral arterial disease (PAD), is the comorbidity primarily responsible for major lower-limb amputations, particularly for diabetic patients. Autologous cell therapy has been the focus of efforts over the past 20 years to create non-interventional therapeutic options for no-option CLTI to improve limb perfusion and wound healing. Among the different available techniques, peripheral blood mononuclear cells (PBMNC) appear to be the most promising autologous cell therapy due to physio-pathological considerations and clinical evidence, which will be discussed in this review. A meta-analysis of six clinical studies, including 256 diabetic patients treated with naive, fresh PBMNC produced via a selective filtration point-of-care device, was conducted. PBMNC was associated with a mean yearly amputation rate of 15.7%, a mean healing rate of 62%, and a time to healing of 208.6 ± 136.5 days. Moreover, an increase in TcPO2 and a reduction in pain were observed. All-cause mortality, with a mean rate of 22.2% and a yearly mortality rate of 18.8%, was reported. No serious adverse events were reported. Finally, some practical and financial considerations are provided, which point to the therapy's recommendation as the first line of treatment for this particular and crucial patient group.
Collapse
Affiliation(s)
- Laura Rehak
- Athena Cell Therapy Technologies, 50126 Florence, Italy
| | - Laura Giurato
- Department of Biomedicine and Prevention, Diabetes-Endocrine Section CTO Hospital, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Matteo Monami
- Department of Diabetology Azienda Ospedaliera Universitaria Careggi, University of Florence, 50134 Florence, Italy
| | - Marco Meloni
- Diabetic Foot Unit, Department of Systems Medicine, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Alessia Scatena
- Diabetology Unit, San Donato Hospital Arezzo, Local Health Authorities Southeast Tuscany, 52100 Arezzo, Italy
| | - Andrea Panunzi
- Department of Biomedicine and Prevention, Diabetes-Endocrine Section CTO Hospital, Tor Vergata University of Rome, 00133 Rome, Italy
- PhD School of Applied Medical and Surgical Sciences, University of Rome Tor Vergata Italy, 00133 Rome, Italy
| | | | | | - Luigi Uccioli
- Department of Biomedicine and Prevention, Diabetes-Endocrine Section CTO Hospital, Tor Vergata University of Rome, 00133 Rome, Italy
| |
Collapse
|
42
|
Sousa NS, Bica M, Brás MF, Sousa AC, Antunes IB, Encarnação IA, Costa TM, Martins IB, Barbosa-Morais NL, Sousa-Victor P, Neves J. The immune landscape of murine skeletal muscle regeneration and aging. Cell Rep 2024; 43:114975. [PMID: 39541212 DOI: 10.1016/j.celrep.2024.114975] [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: 12/04/2023] [Revised: 09/16/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Age-related alterations in the immune system are starting to emerge as key contributors to impairments found in aged organs. A decline in regenerative capacity is a hallmark of tissue aging; however, the contribution of immune aging to regenerative failure is just starting to be explored. Here, we apply a strategy combining single-cell RNA sequencing with flow cytometry, histological analysis, and functional assays to perform a complete analysis of the immune environment of the aged regenerating skeletal muscle on a time course following injury with single-cell resolution. Our results reveal an unanticipated complexity and functional heterogeneity in immune populations within the skeletal muscle that have been regarded as homogeneous. Furthermore, we uncover a profound remodeling of both myeloid and lymphoid compartments in aging. These discoveries challenge established notions on immune regulation of skeletal muscle regeneration, providing a set of potential targets to improve skeletal muscle health and regenerative capacity in aging.
Collapse
Affiliation(s)
- Neuza S Sousa
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal; Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Marta Bica
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal; Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Margarida F Brás
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal
| | - Ana C Sousa
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal
| | - Inês B Antunes
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal
| | - Isabel A Encarnação
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal
| | - Tiago M Costa
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal; Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Inês B Martins
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal; Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | | | - Pedro Sousa-Victor
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal.
| | - Joana Neves
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal.
| |
Collapse
|
43
|
Pryce BR, Oles A, Talbert EE, Romeo MJ, Vaena S, Sharma S, Spadafora V, Tolliver L, Mahvi DA, Morgan KA, Lancaster WP, Beal E, Koren N, Watts B, Overstreet M, Berto S, Subramanian S, Calisir K, Crawford A, Neelon B, Ostrowski MC, Zimmers TA, Tidball JG, Wang DJ, Guttridge DC. Muscle inflammation is regulated by NF-κB from multiple cells to control distinct states of wasting in cancer cachexia. Cell Rep 2024; 43:114925. [PMID: 39475511 PMCID: PMC11774514 DOI: 10.1016/j.celrep.2024.114925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 07/01/2024] [Accepted: 10/14/2024] [Indexed: 12/01/2024] Open
Abstract
Although cancer cachexia is classically characterized as a systemic inflammatory disorder, emerging evidence indicates that weight loss also associates with local tissue inflammation. We queried the regulation of this inflammation and its causality to cachexia by exploring skeletal muscle, whose atrophy strongly associates with poor outcomes. Using multiple mouse models and patient samples, we show that cachectic muscle is marked by enhanced innate immunity. Nuclear factor κB (NF-κB) activity in multiple cells, including satellite cells, myofibers, and fibro-adipogenic progenitors, promotes macrophage expansion equally derived from infiltrating monocytes and resident cells. Moreover, NF-κB-activated cells and macrophages undergo crosstalk; NF-κB+ cells recruit macrophages to inhibit regeneration and promote atrophy but, interestingly, also protect myofibers, while macrophages stimulate NF-κB+ cells to sustain an inflammatory feedforward loop. Together, we propose that NF-κB functions in multiple cells in the muscle microenvironment to stimulate macrophages that both promote and protect against muscle wasting in cancer.
Collapse
Affiliation(s)
- Benjamin R Pryce
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Alexander Oles
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Erin E Talbert
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Health and Human Physiology, and the Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA
| | - Martin J Romeo
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Silvia Vaena
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Sudarshana Sharma
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Victoria Spadafora
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Lauren Tolliver
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC 29425, USA
| | - David A Mahvi
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29403, USA
| | - Katherine A Morgan
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29403, USA
| | - William P Lancaster
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29403, USA
| | - Eryn Beal
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29403, USA
| | - Natlie Koren
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29403, USA
| | - Bailey Watts
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29403, USA
| | - Morgan Overstreet
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29403, USA
| | - Stefano Berto
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Suganya Subramanian
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Kubra Calisir
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Anna Crawford
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Brian Neelon
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michael C Ostrowski
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Teresa A Zimmers
- Department of Cell, Developmental, and Cancer Biology, Knight Cancer Institute, Portland, Oregon Health Science University, Portland, OR 97239, USA
| | - James G Tidball
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - David J Wang
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Denis C Guttridge
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
44
|
Zhao Q, Mertová I, Wróblová A, Žabková S, Tlapáková T, Krylov V. Immunomodulatory role of Xenopus tropicalis immature Sertoli cells in tadpole muscle regeneration via macrophage response modulation. Stem Cell Res Ther 2024; 15:421. [PMID: 39533333 PMCID: PMC11558833 DOI: 10.1186/s13287-024-04050-2] [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: 04/24/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Regenerative medicine and transplantation science continuously seek methods to circumvent immune-mediated rejection and promote tissue regeneration. Sertoli cells, with their inherent immunoprotective properties, emerge as pivotal players in this quest. However, whether Sertoli cells can play immunomodulatory role in tadpole tail regeneration and can thus benefit the regeneration process are needed to be discovered. METHODS Immature Sertoli cells from Xenopus tropicalis (XtiSCs) were transplanted into X. tropicalis tadpoles, followed by the amputation of the final third of their tails. We assessed the migration of XtiSCs, tail regeneration length, muscle degradation and growth, and macrophage counts across various regions including the entire tail, tail trunk, injection site, and regeneration site. The interactions between XtiSCs and macrophages were examined using a confocal microscope. To deplete macrophages, clodronate liposomes were administered prior to the transplantation of XtiSCs, while the administration of control liposomes acted as a negative control. Student's t-test was used to compare the effects of XtiSCs injection to those of a 2/3PBS injection across groups with no liposomes, control liposomes, and clodronate liposomes. RESULTS XtiSCs have excellent viability after transplantation to tadpole tail and remarkable homing capabilities to the regeneration site after tail amputation. XtiSCs injection increased macrophage numbers at 3 days post-amputation and 5 days post-amputation in the tail trunk, specifically at the injection site and at the regenerated tail, in a macrophage depleted environment (clodronate-liposome injection). What's more, XtiSCs injection decreased muscle fibers degradation significantly at 1 day post-amputation and facilitated new muscle growth significantly at 3 days post-amputation. In addition, whole-mount immunostaining showed that some XtiSCs co-localized with macrophages. And we observed potential mitochondria transport from XtiSCs to macrophages using MitoTracker staining in tadpole tail. CONCLUSIONS Our study delineates the novel role of XtiSCs in facilitating muscle regeneration post tadpole tail amputation, underscoring a unique interaction with macrophages that is crucial for regenerative success. This study not only highlights the therapeutic potential of Sertoli cells in regenerative medicine but also opens avenues for clinical translation, offering insights into immunoregulatory strategies that could enhance tissue regeneration and transplant acceptance.
Collapse
Affiliation(s)
- Qing Zhao
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, Prague 2, 128 00, Czech Republic
| | - Irem Mertová
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, Prague 2, 128 00, Czech Republic
| | - Aneta Wróblová
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, Prague 2, 128 00, Czech Republic
| | - Světlana Žabková
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, Prague 2, 128 00, Czech Republic
| | - Tereza Tlapáková
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, Prague 2, 128 00, Czech Republic
| | - Vladimir Krylov
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, Prague 2, 128 00, Czech Republic.
| |
Collapse
|
45
|
Chu Y, Yuan X, Tao Y, Yang B, Luo J. Autophagy in Muscle Regeneration: Mechanisms, Targets, and Therapeutic Perspective. Int J Mol Sci 2024; 25:11901. [PMID: 39595972 PMCID: PMC11593790 DOI: 10.3390/ijms252211901] [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: 09/30/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Autophagy maintains the stability of eukaryotic cells by degrading unwanted components and recycling nutrients and plays a pivotal role in muscle regeneration by regulating the quiescence, activation, and differentiation of satellite cells. Effective muscle regeneration is vital for maintaining muscle health and homeostasis. However, under certain disease conditions, such as aging, muscle regeneration can fail due to dysfunctional satellite cells. Dysregulated autophagy may limit satellite cell self-renewal, hinder differentiation, and increase susceptibility to apoptosis, thereby impeding muscle regeneration. This review explores the critical role of autophagy in muscle regeneration, emphasizing its interplay with apoptosis and recent advances in autophagy research related to diseases characterized by impaired muscle regeneration. Additionally, we discuss new approaches involving autophagy regulation to promote macrophage polarization, enhancing muscle regeneration. We suggest that utilizing cell therapy and biomaterials to modulate autophagy could be a promising strategy for supporting muscle regeneration. We hope that this review will provide new insights into the treatment of muscle diseases and promote muscle regeneration.
Collapse
Affiliation(s)
- Yun Chu
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.C.); (Y.T.); (B.Y.)
| | - Xinrun Yuan
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| | - Yiming Tao
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.C.); (Y.T.); (B.Y.)
| | - Bin Yang
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.C.); (Y.T.); (B.Y.)
| | - Jinlong Luo
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| |
Collapse
|
46
|
Choi J, Chudziak J, Lee JH. Bi-directional regulation between inflammation and stem cells in the respiratory tract. J Cell Sci 2024; 137:jcs263413. [PMID: 39508347 PMCID: PMC11574357 DOI: 10.1242/jcs.263413] [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: 11/15/2024] Open
Abstract
Inflammation plays a crucial role in tissue injury, repair and disease, orchestrating a complex interplay of immune responses and cellular processes. Recent studies have uncovered the intricate connection between inflammation and stem cell dynamics, shedding light on the central role of stem cells in tissue regeneration. This Review highlights the significance of inflammation in shaping epithelial stem cell dynamics and its implications for tissue repair, regeneration and aging. We explore the multifaceted interactions between inflammation and stem cells, focusing on how inflammatory signals affect stem cell behavior and fate as well as the remodeling of their niche in the respiratory tract. We also discuss the concept of 'inflammatory memory' in epithelial stem cells, where prior inflammatory stimuli endow these cells with enhanced regenerative potential and confer long-lasting protective mechanisms for maintaining tissue integrity and function. Furthermore, we review the impact of cell senescence induced by inflammation on tissue regeneration and aging, delving into the molecular mechanisms underlying the modulation of signaling pathways, epigenetic modifications and cellular crosstalk. Understanding these dynamic processes not only deepens our knowledge of tissue homeostasis and repair but also holds profound implications for regenerative medicine strategies aimed at preventing pulmonary diseases.
Collapse
Affiliation(s)
- Jinwook Choi
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Jakub Chudziak
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB1 0AW, UK
| | - Joo-Hyeon Lee
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB1 0AW, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EL, UK
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York NY 10065, USA
| |
Collapse
|
47
|
Saugues A, Kneppers A, Mounier R. [Muscle stem cells and metabolism in Duchenne muscular dystrophy, focus on AMPK]. Med Sci (Paris) 2024; 40 Hors série n° 1:60-63. [PMID: 39555881 DOI: 10.1051/medsci/2024133] [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: 11/19/2024] Open
Abstract
Through their myogenic activity, adult muscle stem cells (MuSCs) are crucial for the regeneration of striated skeletal muscle. Once activated, they proliferate, differentiate and then fuse to repair or form new muscle fibers (myofibers). Their progression through myogenesis requires a complex regulation involving multiple players such as metabolism, in particular via AMPK. This protein kinase regulates the self-renewal and myonuclear accretion of MuSCs after acute skeletal muscle injury or skeletal muscle contraction. However, in a context of dystrophy such as Duchenne muscular dystrophy (DMD), the regenerative capacity of MuSCs is reduced, presumably due to an increase of the proliferation that is detrimental to differentiation. We are interested here in the potential of metabolism to regulate the myogenic activity of MuSCs in DMD via AMPK.
Collapse
Affiliation(s)
- Audrey Saugues
- Institut NeuroMyoGène, PGNM, CNRS UMR5261/Inserm U1315/ Université Claude Bernard Lyon 1, France
| | - Anita Kneppers
- Institut NeuroMyoGène, PGNM, CNRS UMR5261/Inserm U1315/ Université Claude Bernard Lyon 1, France
| | - Rémi Mounier
- Institut NeuroMyoGène, PGNM, CNRS UMR5261/Inserm U1315/ Université Claude Bernard Lyon 1, France
| |
Collapse
|
48
|
Alharbi A, Li J, Womack E, Farrow M, Yarar-Fisher C. The Effect of Lower Limb Combined Neuromuscular Electrical Stimulation on Skeletal Muscle Cross-Sectional Area and Inflammatory Signaling. Int J Mol Sci 2024; 25:11095. [PMID: 39456876 PMCID: PMC11507577 DOI: 10.3390/ijms252011095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
In individuals with a spinal cord injury (SCI), rapid skeletal muscle atrophy and metabolic dysfunction pose profound rehabilitation challenges, often resulting in substantial loss of muscle mass and function. This study evaluates the effect of combined neuromuscular electrical stimulation (Comb-NMES) on skeletal muscle cross-sectional area (CSA) and inflammatory signaling within the acute phase of SCI. We applied a novel Comb-NMES regimen, integrating both high-frequency resistance and low-frequency aerobic protocols on the vastus lateralis muscle, to participants early post-SCI. Muscle biopsies were analyzed for CSA and inflammatory markers pre- and post-intervention. The results suggest a potential preservation of muscle CSA in the Comb-NMES group compared to a control group. Inflammatory signaling proteins such as TLR4 and Atrogin-1 were downregulated, whereas markers associated with muscle repair and growth were modulated beneficially in the Comb-NMES group. The study's findings suggest that early application of Comb-NMES post-SCI may attenuate inflammatory pathways linked to muscle atrophy and promote muscle repair. However, the small sample size and variability in injury characteristics emphasize the need for further research to corroborate these results across a more diverse and extensive SCI population.
Collapse
Affiliation(s)
- Amal Alharbi
- Department of Physical Therapy, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
- Department of Physical Therapy, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Jia Li
- Department of Physical Medicine and Rehabilitation, Ohio State University, Columbus, OH 43210, USA; (J.L.); (M.F.)
| | - Erika Womack
- Department of Biochemistry, Molecular Biology, Entomology, and Plant Pathology, Mississippi State University, Starkville, MS 39762, USA;
| | - Matthew Farrow
- Department of Physical Medicine and Rehabilitation, Ohio State University, Columbus, OH 43210, USA; (J.L.); (M.F.)
| | - Ceren Yarar-Fisher
- Department of Physical Medicine and Rehabilitation, Ohio State University, Columbus, OH 43210, USA; (J.L.); (M.F.)
- Department of Neuroscience, Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
49
|
Huang D, Jiao X, Huang S, Liu J, Si H, Qi D, Pei X, Lu D, Wang Y, Li Z. Analysis of the heterogeneity and complexity of murine extraorbital lacrimal gland via single-cell RNA sequencing. Ocul Surf 2024; 34:60-95. [PMID: 38945476 DOI: 10.1016/j.jtos.2024.06.005] [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: 08/26/2022] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/02/2024]
Abstract
PURPOSE The lacrimal gland is essential for maintaining ocular surface health and avoiding external damage by secreting an aqueous layer of the tear film. However, a healthy lacrimal gland's inventory of cell types and heterogeneity remains understudied. METHODS Here, 10X Genome-based single-cell RNA sequencing was used to generate an unbiased classification of cellular diversity in the extraorbital lacrimal gland (ELG) of C57BL/6J mice. From 43,850 high-quality cells, we produced an atlas of cell heterogeneity and defined cell types using classic marker genes. The possible functions of these cells were analyzed through bioinformatics analysis. Additionally, the CellChat was employed for a preliminary analysis of the cell-cell communication network in the ELG. RESULTS Over 37 subclasses of cells were identified, including seven types of glandular epithelial cells, three types of fibroblasts, ten types of myeloid-derived immune cells, at least eleven types of lymphoid-derived immune cells, and five types of vascular-associated cell subsets. The cell-cell communication network analysis revealed that fibroblasts and immune cells play a pivotal role in the dense intercellular communication network within the mouse ELG. CONCLUSIONS This study provides a comprehensive transcriptome atlas and related database of the mouse ELG.
Collapse
Affiliation(s)
- Duliurui Huang
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Xinwei Jiao
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Shenzhen Huang
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Jiangman Liu
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Hongli Si
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Di Qi
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Xiaoting Pei
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Dingli Lu
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Yimian Wang
- Division of Medicine, Faculty of Medical Sciences, University College London, Gower Street, London, WC1E 6BT, UK
| | - Zhijie Li
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China; Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China.
| |
Collapse
|
50
|
Chen C, Feng C, Luo Q, Zeng Y, Yuan W, Cui Y, Tang Z, Zhang H, Li T, Peng J, Peng L, Xie X, Guo Y, Peng F, Jiang X, Bai P, Qi Z, Dai H. CD5L up-regulates the TGF-β signaling pathway and promotes renal fibrosis. Life Sci 2024; 354:122945. [PMID: 39127319 DOI: 10.1016/j.lfs.2024.122945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/25/2024] [Accepted: 08/04/2024] [Indexed: 08/12/2024]
Abstract
Renal fibrosis is the common final pathway of progressive renal diseases, in which the macrophages play an important role. ELISA was used to detect CD5 antigen-like (CD5L) in serum samples from end-stage renal disease (ESRD), as well as in mice serum with unilateral ureteral occlusion (UUO). Recombinant CD5L was injected into UUO mice to assess renal injury, fibrosis, and macrophage infiltration. The expression of CD5L was significantly upregulated in the serum of patients with ESRD and UUO mice. Histological analysis showed that rCD5L-treated UUO mice had more severe renal injury and fibrosis. Furthermore, rCD5L promoted the phenotypic transfer of monocytes from Ly6Chigh to LyC6low. RCD5L promoted TGF-β signaling pathway activation by promoting Smad2/3 phosphorylation. We used Co-IP to identify HSPA5 interact with CD5L on cell membrane could inhibit the formation of the Cripto/HSPA5 complex, and promote the activation of the TGF-β signaling pathway. The CD5L antibody could reduce the degree of renal fibrosis in UUO mice.
Collapse
Affiliation(s)
- Chao Chen
- Medical College, Guangxi University, Nanning 530004, China; Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Chen Feng
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Qiulin Luo
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Yingqi Zeng
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Wenjia Yuan
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Yan Cui
- Medical College, Guangxi University, Nanning 530004, China; Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Zhouqi Tang
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Hedong Zhang
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Tengfang Li
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Jiawei Peng
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Longkai Peng
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xubiao Xie
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Yong Guo
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Fenghua Peng
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xin Jiang
- Department of Organ Transplantation, The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), Zhengzhou, Henan 450000, China
| | - Peiming Bai
- Medical College, Guangxi University, Nanning 530004, China; Department of Urology, Zhongshan Hospital Xiamen University, Xiamen 361000, China.
| | - Zhongquan Qi
- Medical College, Guangxi University, Nanning 530004, China; Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian 350001, China.
| | - Helong Dai
- Medical College, Guangxi University, Nanning 530004, China; Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|