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
|
Jablonka-Shariff A, Broberg C, Snyder-Warwick AK. Absence of T-box transcription factor 21 limits neuromuscular junction recovery after nerve injury in T-bet-knockout mice. Front Cell Dev Biol 2025; 13:1535323. [PMID: 40162097 PMCID: PMC11949913 DOI: 10.3389/fcell.2025.1535323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/20/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction Terminal Schwann cells (tSCs), at the neuromuscular junction (NMJ), play critical roles in the repair of motor axon terminals at muscle, and rebuild neuronal signaling following nerve injury. Knowledge of mediators impacting tSCs post-nerve injury and in disease may guide beneficial therapies to improve motor outcomes. We previously found T-box transcription factor 21 (TBX21/TBET), classically associated with T-helper1 cells and immune cell recruitment, is expressed in tSCs at the mouse NMJ. The purpose of this study was to examine effects of Tbx21 absence during NMJ regeneration following peripheral nerve injury. Methods Wildtype (WT) and Tbet-knockout (Tbet-KO) mice underwent sciatic nerve transection and immediate repair. Functional muscle recovery assessment was performed with muscle force testing on mice at 2-, 3-, 4-, and 6-week (wks) and 6 months after nerve injury repair. Morphometric analyses of NMJ reinnervation, tSC number, and tSC processes were evaluated. Full NMJ reinnervation was defined as ≥75% coverage of endplates by axons. A minimum of three mice were evaluated in each group, and 50-100 NMJs were evaluated per mouse. Results Tbet-KO mice had significantly diminished muscle function compared to WT mice at every time point beyond 3 weeks. Tbet-KO mice showed just over half of the muscle force generated by WT mice at 4 weeks and 6 weeks post-injury and repair. By 6 months, Tbet-KO mice generated only 84.1% the muscle force of WT mice. Tbet-KO mice showed significantly decreased levels of fully reinnervated NMJs compared to WT mice at each time point tested. Tbet-KO mice also showed a lower number of tSCs with reduced cytoplasmic processes beyond NMJ area and lower number of immune cells during process of NMJ regeneration. Discussion Our findings show that the Tbx21 transcription factor promotes NMJ reinnervation to regain muscle function following nerve injury.
Collapse
Affiliation(s)
- Albina Jablonka-Shariff
- Research Scientist, Division of Plastic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States
| | - Curtis Broberg
- Research Student, Washington University School of Medicine, St. Louis, MO, United States
| | - Alison K. Snyder-Warwick
- Division of Plastic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
3
|
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
|
4
|
Klawitter F, Laukien F, Fischer DC, Rahn A, Porath K, Danckert L, Bajorat R, Walter U, Patejdl R, Ehler J. Longitudinal Assessment of Blood-Based Inflammatory, Neuromuscular, and Neurovascular Biomarker Profiles in Intensive Care Unit-Acquired Weakness: A Prospective Single-Center Cohort Study. Neurocrit Care 2025; 42:118-130. [PMID: 38982001 PMCID: PMC11811256 DOI: 10.1007/s12028-024-02050-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/13/2024] [Indexed: 07/11/2024]
Abstract
BACKGROUND The diagnosis of intensive care unit (ICU)-acquired weakness (ICUAW) and critical illness neuromyopathy (CINM) is frequently hampered in the clinical routine. We evaluated a novel panel of blood-based inflammatory, neuromuscular, and neurovascular biomarkers as an alternative diagnostic approach for ICUAW and CINM. METHODS Patients admitted to the ICU with a Sequential Organ Failure Assessment score of ≥ 8 on 3 consecutive days within the first 5 days as well as healthy controls were enrolled. The Medical Research Council Sum Score (MRCSS) was calculated, and motor and sensory electroneurography (ENG) for assessment of peripheral nerve function were performed at days 3 and 10. ICUAW was defined by an MRCSS < 48 and CINM by pathological ENG alterations, both at day 10. Blood samples were taken at days 3, 10, and 17 for quantitative analysis of 18 different biomarkers (white blood cell count, C-reactive protein, procalcitonin, C-terminal agrin filament, fatty-acid-binding protein 3, growth and differentiation factor 15, syndecan 1, troponin I, interferon-γ, tumor necrosis factor-α, interleukin-1α [IL-1α], IL-1β, IL-4, IL-6, IL-8, IL-10, IL-13, and monocyte chemoattractant protein 1). Results of the biomarker analysis were categorized according to the ICUAW and CINM status. Clinical outcome was assessed after 3 months. RESULTS Between October 2016 and December 2018, 38 critically ill patients, grouped into ICUAW (18 with and 20 without) and CINM (18 with and 17 without), as well as ten healthy volunteers were included. Biomarkers were significantly elevated in critically ill patients compared to healthy controls and correlated with disease severity and 3-month outcome parameters. However, none of the biomarkers enabled discrimination of patients with and without neuromuscular impairment, irrespective of applied classification. CONCLUSIONS Blood-based biomarkers are generally elevated in ICU patients but do not identify patients with ICUAW or CINM. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT02706314.
Collapse
Affiliation(s)
- Felix Klawitter
- Department of Anesthesiology, Intensive Care Medicine, and Pain Therapy, Rostock University Medical Center, Rostock, 18057, Germany.
| | - Friederike Laukien
- Department of Anesthesiology, Intensive Care Medicine, and Pain Therapy, Rostock University Medical Center, Rostock, 18057, Germany
| | - Dagmar-C Fischer
- Department of Pediatrics, Rostock University Medical Center, Rostock, Germany
| | - Anja Rahn
- Department of Pediatrics, Rostock University Medical Center, Rostock, Germany
| | - Katrin Porath
- Oscar Langendorff Institute of Physiology, Rostock University Medical Center, Rostock, Germany
| | - Lena Danckert
- Department of Anesthesiology, Intensive Care Medicine, and Pain Therapy, Rostock University Medical Center, Rostock, 18057, Germany
| | - Rika Bajorat
- Department of Anesthesiology, Intensive Care Medicine, and Pain Therapy, Rostock University Medical Center, Rostock, 18057, Germany
| | - Uwe Walter
- Department of Neurology, Rostock University Medical Center, Rostock, Germany
| | - Robert Patejdl
- Oscar Langendorff Institute of Physiology, Rostock University Medical Center, Rostock, Germany
- Department of Medicine, Health and Medical University Erfurt, Erfurt, Germany
| | - Johannes Ehler
- Department of Anesthesiology, Intensive Care Medicine, and Pain Therapy, Rostock University Medical Center, Rostock, 18057, Germany
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Jena, Jena, Germany
| |
Collapse
|
5
|
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
|
6
|
Willis AB, Zelikovich AS, Sufit R, Ajroud-Driss S, Vandenborne K, Demonbreun AR, Batra A, Walter GA, McNally EM. Serum protein and imaging biomarkers after intermittent steroid treatment in muscular dystrophy. Sci Rep 2024; 14:28745. [PMID: 39567576 PMCID: PMC11579281 DOI: 10.1038/s41598-024-79024-8] [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: 04/15/2024] [Accepted: 11/04/2024] [Indexed: 11/22/2024] Open
Abstract
Weekly Steroids in Muscular Dystrophy (WSiMD) was a pilot study to evaluate once weekly prednisone in patients with Limb Girdle and Becker muscular dystrophy (LGMD and BMD, respectively). At study endpoint, there were trends towards increased lean mass, reduced fat mass, reduced creatine kinase and improved motor function. The investigation was motivated by studies in mouse muscular dystrophy models in which once weekly glucocorticoid exposure enhanced muscle strength and reduced fibrosis. WSiMD participants provided blood samples for aptamer serum profiling at baseline and after 6 months of weekly steroids. A subset completed magnetic resonance (MR) evaluation of muscle at study onset and endpoint. At baseline compared to age and sex-matched healthy controls, the aggregate serum protein profile in the WSiMD cohort was dominated by muscle proteins, reflecting leak of muscle proteins into serum. Disease status produced more proteins differentially present in serum compared to steroid-treatment effect. Nonetheless, a response to prednisone was discernable in the WSiMD cohort, even at this low dose. Glucocorticoids decreased muscle proteins and increased certain immune process- and matrix-associated proteins. Muscle MR fat fraction showed trends with functional status. The prednisone-responsive markers could be used in larger trial of prednisone efficacy.
Collapse
Affiliation(s)
- Alexander B Willis
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, 303 E Superior SQ 5-516, Chicago, IL, 60611, USA
| | - Aaron S Zelikovich
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, 303 E Superior SQ 5-516, Chicago, IL, 60611, USA
| | - Robert Sufit
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Senda Ajroud-Driss
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Alexis R Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, 303 E Superior SQ 5-516, Chicago, IL, 60611, USA
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Abhinandan Batra
- Department of Physical Therapy, University of Louisiana at Monroe, Monroe, LA, USA
| | - Glenn A Walter
- Department of Physiology and Aging, University of Florida, Gainesville, FL, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, 303 E Superior SQ 5-516, Chicago, IL, 60611, USA.
| |
Collapse
|
7
|
Russo C, Surdo S, Valle MS, Malaguarnera L. The Gut Microbiota Involvement in the Panorama of Muscular Dystrophy Pathogenesis. Int J Mol Sci 2024; 25:11310. [PMID: 39457092 PMCID: PMC11508360 DOI: 10.3390/ijms252011310] [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: 09/10/2024] [Revised: 10/16/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024] Open
Abstract
Muscular dystrophies (MDs) are genetically heterogeneous diseases characterized by primary skeletal muscle atrophy. The collapse of muscle structure and irreversible degeneration of tissues promote the occurrence of comorbidities, including cardiomyopathy and respiratory failure. Mitochondrial dysfunction leads to inflammation, fibrosis, and adipogenic cellular infiltrates that exacerbate the symptomatology of MD patients. Gastrointestinal disorders and metabolic anomalies are common in MD patients and may be determined by the interaction between the intestine and its microbiota. Therefore, the gut-muscle axis is one of the actors involved in the spread of inflammatory signals to all muscles. In this review, we aim to examine in depth how intestinal dysbiosis can modulate the metabolic state, the immune response, and mitochondrial biogenesis in the course and progression of the most investigated MDs such as Duchenne Muscular Dystrophy (DMD) and Myotonic Dystrophy (MD1), to better identify gut microbiota metabolites working as therapeutic adjuvants to improve symptoms of MD.
Collapse
Affiliation(s)
- Cristina Russo
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Sofia Surdo
- Italian Center for the Study of Osteopathy (CSDOI), 95124 Catania, Italy;
| | - Maria Stella Valle
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
| | - Lucia Malaguarnera
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| |
Collapse
|
8
|
Patsalos A, Halasz L, Oleksak D, Wei X, Nagy G, Tzerpos P, Conrad T, Hammers DW, Sweeney HL, Nagy L. Spatiotemporal transcriptomic mapping of regenerative inflammation in skeletal muscle reveals a dynamic multilayered tissue architecture. J Clin Invest 2024; 134:e173858. [PMID: 39190487 PMCID: PMC11473166 DOI: 10.1172/jci173858] [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: 07/12/2023] [Accepted: 08/21/2024] [Indexed: 08/29/2024] Open
Abstract
Tissue regeneration is orchestrated by macrophages that clear damaged cells and promote regenerative inflammation. How macrophages spatially adapt and diversify their functions to support the architectural requirements of actively regenerating tissue remains unknown. In this study, we reconstructed the dynamic trajectories of myeloid cells isolated from acutely injured and early stage dystrophic muscles. We identified divergent subsets of monocytes/macrophages and DCs and validated markers (e.g., glycoprotein NMB [GPNMB]) and transcriptional regulators associated with defined functional states. In dystrophic muscle, specialized repair-associated subsets exhibited distinct macrophage diversity and reduced DC heterogeneity. Integrating spatial transcriptomics analyses with immunofluorescence uncovered the ordered distribution of subpopulations and multilayered regenerative inflammation zones (RIZs) where distinct macrophage subsets are organized in functional zones around damaged myofibers supporting all phases of regeneration. Importantly, intermittent glucocorticoid treatment disrupted the RIZs. Our findings suggest that macrophage subtypes mediated the development of the highly ordered architecture of regenerative tissues, unveiling the principles of the structured yet dynamic nature of regenerative inflammation supporting effective tissue repair.
Collapse
Affiliation(s)
- Andreas Patsalos
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, Florida, USA
| | - Laszlo Halasz
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, Florida, USA
| | - Darby Oleksak
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, Florida, USA
| | - Xiaoyan Wei
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, Florida, USA
| | - Gergely Nagy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Petros Tzerpos
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Thomas Conrad
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - David W. Hammers
- Myology Institute and Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, USA
| | - H. Lee Sweeney
- Myology Institute and Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, USA
| | - Laszlo Nagy
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, Florida, USA
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
9
|
Correia JC, Jannig PR, Gosztyla ML, Cervenka I, Ducommun S, Præstholm SM, Dias JM, Dumont KD, Liu Z, Liang Q, Edsgärd D, Emanuelsson O, Gregorevic P, Westerblad H, Venckunas T, Brazaitis M, Kamandulis S, Lanner JT, Teixeira AI, Yeo GW, Ruas JL. Zfp697 is an RNA-binding protein that regulates skeletal muscle inflammation and remodeling. Proc Natl Acad Sci U S A 2024; 121:e2319724121. [PMID: 39141348 PMCID: PMC11348326 DOI: 10.1073/pnas.2319724121] [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: 11/17/2023] [Accepted: 06/27/2024] [Indexed: 08/15/2024] Open
Abstract
Skeletal muscle atrophy is a morbidity and mortality risk factor that happens with disuse, chronic disease, and aging. The tissue remodeling that happens during recovery from atrophy or injury involves changes in different cell types such as muscle fibers, and satellite and immune cells. Here, we show that the previously uncharacterized gene and protein Zfp697 is a damage-induced regulator of muscle remodeling. Zfp697/ZNF697 expression is transiently elevated during recovery from muscle atrophy or injury in mice and humans. Sustained Zfp697 expression in mouse muscle leads to a gene expression signature of chemokine secretion, immune cell recruitment, and extracellular matrix remodeling. Notably, although Zfp697 is expressed in several cell types in skeletal muscle, myofiber-specific Zfp697 genetic ablation in mice is sufficient to hinder the inflammatory and regenerative response to muscle injury, compromising functional recovery. We show that Zfp697 is an essential mediator of the interferon gamma response in muscle cells and that it functions primarily as an RNA-interacting protein, with a very high number of miRNA targets. This work identifies Zfp697 as an integrator of cell-cell communication necessary for tissue remodeling and regeneration.
Collapse
Affiliation(s)
- Jorge C. Correia
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
| | - Paulo R. Jannig
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
| | - Maya L. Gosztyla
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA92093
- Sanford Stem Cell Institute Innovation Center and Stem Cell Program, University of California San Diego, La Jolla, CA92093
- Center for RNA Technologies and Therapeutics, University of California San Diego, La Jolla, CA92093
| | - Igor Cervenka
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
| | - Serge Ducommun
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
| | - Stine M. Præstholm
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
| | - José M. Dias
- Nanomedicine and Spatial Biology, Department of Physiology and Pharmacology, Biomedicum, Karolinska, StockholmSE-171 77, Sweden
- Department of Cell and Molecular Biology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
| | - Kyle D. Dumont
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
| | - Zhengye Liu
- Molecular Muscle Physiology and Pathophysiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
| | - Qishan Liang
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA92093
- Center for RNA Technologies and Therapeutics, University of California San Diego, La Jolla, CA92093
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA92093
| | - Daniel Edsgärd
- Science for Life Laboratory, Department of Gene Technology, School of Engineering Sciences in Biotechnology, Chemistry and Health, KTH Royal Institute of Technology, Stockholm SE-100 44, Sweden
| | - Olof Emanuelsson
- Science for Life Laboratory, Department of Gene Technology, School of Engineering Sciences in Biotechnology, Chemistry and Health, KTH Royal Institute of Technology, Stockholm SE-100 44, Sweden
| | - Paul Gregorevic
- Centre for Muscle Research, Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Håkan Westerblad
- Muscle Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
| | - Tomas Venckunas
- Institute of Sports Science and Innovations, Lithuanian Sports University, Kaunas44221, Lithuania
| | - Marius Brazaitis
- Institute of Sports Science and Innovations, Lithuanian Sports University, Kaunas44221, Lithuania
| | - Sigitas Kamandulis
- Institute of Sports Science and Innovations, Lithuanian Sports University, Kaunas44221, Lithuania
| | - Johanna T. Lanner
- Molecular Muscle Physiology and Pathophysiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
| | - Ana I. Teixeira
- Nanomedicine and Spatial Biology, Department of Physiology and Pharmacology, Biomedicum, Karolinska, StockholmSE-171 77, Sweden
| | - Gene W. Yeo
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA92093
- Sanford Stem Cell Institute Innovation Center and Stem Cell Program, University of California San Diego, La Jolla, CA92093
- Center for RNA Technologies and Therapeutics, University of California San Diego, La Jolla, CA92093
| | - Jorge L. Ruas
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
- Department of Pharmacology and Stanley & Judith Frankel Institute for Heart & Brain Health, University of Michigan Medical School, Ann Arbor, MI48109
| |
Collapse
|
10
|
Willis AB, Zelikovich AS, Sufit R, Ajroud-Driss S, Vandenborne K, Demonbreun AR, Batra A, Walter GA, McNally EM. Serum protein and imaging biomarkers after intermittent steroid treatment in muscular dystrophy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.14.24308858. [PMID: 38947030 PMCID: PMC11213068 DOI: 10.1101/2024.06.14.24308858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Background Weekly Steroids in Muscular Dystrophy (WSiMD) was a pilot study to evaluate once weekly prednisone in patients with Limb Girdle and Becker muscular dystrophy (LGMD and BMD, respectively). At study endpoint, there were trends towards increased lean mass, reduced fat mass, reduced creatine kinase and improved motor function. The investigation was motivated by studies in mouse muscular dystrophy models in which once weekly glucocorticoid exposure enhanced muscle strength and reduced fibrosis. Methods WSiMD participants provided blood samples for aptamer serum profiling at baseline and after 6 months of weekly steroids. A subset completed magnetic resonance (MR) evaluation of muscle at study onset and endpoint. Results/Conclusions At baseline compared to age and sex-matched healthy controls, the aggregate serum protein profile in the WSiMD cohort was dominated by muscle proteins, reflecting leak of muscle proteins into serum. Disease status produced more proteins differentially present in serum compared to steroid-treatment effect. Nonetheless, a response to prednisone was discernable in the WSiMD cohort, even at this low dose. Glucocorticoids downregulated muscle proteins and upregulated certain immune process- and matrix-associated proteins. Muscle MR fat fraction showed trends with functional status. The prednisone-responsive markers could be used in larger trial of prednisone efficacy.
Collapse
Affiliation(s)
- Alexander B. Willis
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Aaron S. Zelikovich
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Robert Sufit
- Dept of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Senda Ajroud-Driss
- Dept of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | - Alexis R. Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Abhinandan Batra
- Department of Physical Therapy, University of Louisiana at Monroe, Monroe, LA
| | - Glenn A. Walter
- Department of Physiology and Aging, University of Florida, Gainesville, FL
| | - Elizabeth M. McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
11
|
Espino-Gonzalez E, Dalbram E, Mounier R, Gondin J, Farup J, Jessen N, Treebak JT. Impaired skeletal muscle regeneration in diabetes: From cellular and molecular mechanisms to novel treatments. Cell Metab 2024; 36:1204-1236. [PMID: 38490209 DOI: 10.1016/j.cmet.2024.02.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 01/10/2024] [Accepted: 02/22/2024] [Indexed: 03/17/2024]
Abstract
Diabetes represents a major public health concern with a considerable impact on human life and healthcare expenditures. It is now well established that diabetes is characterized by a severe skeletal muscle pathology that limits functional capacity and quality of life. Increasing evidence indicates that diabetes is also one of the most prevalent disorders characterized by impaired skeletal muscle regeneration, yet underlying mechanisms and therapeutic treatments remain poorly established. In this review, we describe the cellular and molecular alterations currently known to occur during skeletal muscle regeneration in people with diabetes and animal models of diabetes, including its associated comorbidities, e.g., obesity, hyperinsulinemia, and insulin resistance. We describe the role of myogenic and non-myogenic cell types on muscle regeneration in conditions with or without diabetes. Therapies for skeletal muscle regeneration and gaps in our knowledge are also discussed, while proposing future directions for the field.
Collapse
Affiliation(s)
- Ever Espino-Gonzalez
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Emilie Dalbram
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Rémi Mounier
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Julien Gondin
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Jean Farup
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Niels Jessen
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark; Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark.
| |
Collapse
|
12
|
Lynch CA, Acosta SA, Anderson DM, Rogers GE, Wilson-Rawls J, Rawls A. The Transcription Factor Mohawk Facilitates Skeletal Muscle Repair via Modulation of the Inflammatory Environment. Int J Mol Sci 2024; 25:5019. [PMID: 38732238 PMCID: PMC11084535 DOI: 10.3390/ijms25095019] [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: 03/23/2024] [Revised: 05/02/2024] [Accepted: 05/02/2024] [Indexed: 05/13/2024] Open
Abstract
Efficient repair of skeletal muscle relies upon the precise coordination of cells between the satellite cell niche and innate immune cells that are recruited to the site of injury. The expression of pro-inflammatory cytokines and chemokines such as TNFα, IFNγ, CXCL1, and CCL2, by muscle and tissue resident immune cells recruits neutrophils and M1 macrophages to the injury and activates satellite cells. These signal cascades lead to highly integrated temporal and spatial control of muscle repair. Despite the therapeutic potential of these factors for improving tissue regeneration after traumatic and chronic injuries, their transcriptional regulation is not well understood. The transcription factor Mohawk (Mkx) functions as a repressor of myogenic differentiation and regulates fiber type specification. Embryonically, Mkx is expressed in all progenitor cells of the musculoskeletal system and is expressed in human and mouse myeloid lineage cells. An analysis of mice deficient for Mkx revealed a delay in postnatal muscle repair characterized by impaired clearance of necrotic fibers and smaller newly regenerated fibers. Further, there was a delay in the expression of inflammatory signals such as Ccl2, Ifnγ, and Tgfß. This was coupled with impaired recruitment of pro-inflammatory macrophages to the site of muscle damage. These studies demonstrate that Mkx plays a critical role in adult skeletal muscle repair that is mediated through the initial activation of the inflammatory response.
Collapse
Affiliation(s)
- Cherie Alissa Lynch
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85287, USA; (C.A.L.); (S.A.A.); (D.M.A.); (G.E.R.); (J.W.-R.)
- Molecular and Cellular Biology Graduate Program, Arizona State University, Tempe, AZ 85287, USA
| | - Sofia A. Acosta
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85287, USA; (C.A.L.); (S.A.A.); (D.M.A.); (G.E.R.); (J.W.-R.)
- Molecular and Cellular Biology Graduate Program, Arizona State University, Tempe, AZ 85287, USA
| | - Douglas M. Anderson
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85287, USA; (C.A.L.); (S.A.A.); (D.M.A.); (G.E.R.); (J.W.-R.)
- Molecular and Cellular Biology Graduate Program, Arizona State University, Tempe, AZ 85287, USA
| | - Gavin E. Rogers
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85287, USA; (C.A.L.); (S.A.A.); (D.M.A.); (G.E.R.); (J.W.-R.)
- Molecular and Cellular Biology Graduate Program, Arizona State University, Tempe, AZ 85287, USA
| | - Jeanne Wilson-Rawls
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85287, USA; (C.A.L.); (S.A.A.); (D.M.A.); (G.E.R.); (J.W.-R.)
| | - Alan Rawls
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85287, USA; (C.A.L.); (S.A.A.); (D.M.A.); (G.E.R.); (J.W.-R.)
| |
Collapse
|
13
|
Carey CM, Hollins HL, Schmid AV, Gagnon JA. Distinct features of the regenerating heart uncovered through comparative single-cell profiling. Biol Open 2024; 13:bio060156. [PMID: 38526188 PMCID: PMC11007736 DOI: 10.1242/bio.060156] [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: 09/18/2023] [Accepted: 03/14/2024] [Indexed: 03/26/2024] Open
Abstract
Adult humans respond to heart injury by forming a permanent scar, yet other vertebrates are capable of robust and complete cardiac regeneration. Despite progress towards characterizing the mechanisms of cardiac regeneration in fish and amphibians, the large evolutionary gulf between mammals and regenerating vertebrates complicates deciphering which cellular and molecular features truly enable regeneration. To better define these features, we compared cardiac injury responses in zebrafish and medaka, two fish species that share similar heart anatomy and common teleost ancestry but differ in regenerative capability. We used single-cell transcriptional profiling to create a time-resolved comparative cell atlas of injury responses in all major cardiac cell types across both species. With this approach, we identified several key features that distinguish cardiac injury response in the non-regenerating medaka heart. By comparing immune responses to injury, we found altered cell recruitment and a distinct pro-inflammatory gene program in medaka leukocytes, and an absence of the injury-induced interferon response seen in zebrafish. In addition, we found a lack of pro-regenerative signals, including nrg1 and retinoic acid, from medaka endothelial and epicardial cells. Finally, we identified alterations in the myocardial structure in medaka, where they lack primordial layer cardiomyocytes and fail to employ a cardioprotective gene program shared by regenerating vertebrates. Our findings reveal notable variation in injury response across nearly all major cardiac cell types in zebrafish and medaka, demonstrating how evolutionary divergence influences the hidden cellular features underpinning regenerative potential in these seemingly similar vertebrates.
Collapse
Affiliation(s)
- Clayton M. Carey
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - Hailey L. Hollins
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - Alexis V. Schmid
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - James A. Gagnon
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
- Henry Eyring Center for Cell & Genome Science, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
14
|
Hu P. Effects of the immune system on muscle regeneration. Curr Top Dev Biol 2024; 158:239-251. [PMID: 38670708 DOI: 10.1016/bs.ctdb.2024.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Muscle regeneration is a complex process orchestrated by multiple steps. Recent findings indicate that inflammatory responses could play central roles in bridging initial muscle injury responses and timely muscle injury reparation. The various types of immune cells and cytokines have crucial roles in muscle regeneration process. In this review, we provide an overview of the functions of acute inflammation in muscle regeneration.
Collapse
Affiliation(s)
- Ping Hu
- The 10th People's Hospital affiliated to Tongji University, Shanghai, P. R. China.
| |
Collapse
|
15
|
Kim JR, Hong BK, Pham THN, Kim WU, Kim HA. Interferon-gamma signaling promotes cartilage regeneration after injury. Sci Rep 2024; 14:8046. [PMID: 38580748 PMCID: PMC10997668 DOI: 10.1038/s41598-024-58779-0] [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: 12/05/2023] [Accepted: 04/03/2024] [Indexed: 04/07/2024] Open
Abstract
Osteoarthritis is a common chronic disease and major cause of disability and chronic pain in ageing populations. In this pathology, the entire joint is involved, and the regeneration of articular cartilage still remains one of the main challenges. Here, we investigated the molecular mechanisms underlying cartilage regeneration in young mice using a full-thickness cartilage injury (FTCI) model. FTCI-induced cartilage defects were created in the femoral trochlea of young and adult C57BL/6 mice. To identify key molecules and pathways involved in the early response to cartilage injury, we performed RNA sequencing (RNA-seq) analysis of cartilage RNA at 3 days after injury. Young mice showed superior cartilage regeneration compared to adult mice after cartilage injury. RNA-seq analysis revealed significant upregulation of genes associated with the immune response, particularly in the IFN-γ signaling pathway and qRT-PCR analysis showed macrophage polarization in the early phase of cartilage regeneration (3 days) in young mice after injury, which might promote the removal of damaged or necrotic cells and initiate cartilage regeneration in response to injury. IFN-γR1- and IFN-γ-deficient mice exhibited impaired cartilage regeneration following cartilage injury. DMM-induced and spontaneous OA phenotypes were exacerbated in IFN-γR1-/- mice than in wild-type mice. Our data support the hypothesis that IFN-γ signaling is necessary for cartilage regeneration, as well as for the amelioration of post-traumatic and age-induced OA.
Collapse
Affiliation(s)
- Ju-Ryoung Kim
- Division of Rheumatology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, 896, Pyungchon, Anyang, Kyunggi, 14068, Korea
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Center for Intergrative Rheumatoid Transcriptomics and Dynamics, School of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Bong-Ki Hong
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Center for Intergrative Rheumatoid Transcriptomics and Dynamics, School of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Thi Hong Nhung Pham
- Division of Rheumatology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, 896, Pyungchon, Anyang, Kyunggi, 14068, Korea
- Institute for Skeletal Aging, Hallym University, Chuncheon, Gangwon-do, 24252, Korea
| | - Wan-Uk Kim
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Center for Intergrative Rheumatoid Transcriptomics and Dynamics, School of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Hyun Ah Kim
- Division of Rheumatology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, 896, Pyungchon, Anyang, Kyunggi, 14068, Korea.
- Institute for Skeletal Aging, Hallym University, Chuncheon, Gangwon-do, 24252, Korea.
| |
Collapse
|
16
|
Kobayashi AJ, Sesillo FB, Do E, Alperin M. Effect of nonsteroidal anti-inflammatory drugs on pelvic floor muscle regeneration in a preclinical birth injury rat model. Am J Obstet Gynecol 2024; 230:432.e1-432.e14. [PMID: 38065378 PMCID: PMC10990831 DOI: 10.1016/j.ajog.2023.12.001] [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: 08/25/2023] [Revised: 11/22/2023] [Accepted: 12/03/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Pelvic floor muscle injury is a common consequence of vaginal childbirth. Nonsteroidal anti-inflammatory drugs are widely used postpartum analgesics. Multiple studies have reported negative effects of these drugs on limb muscle regeneration, but their impact on pelvic floor muscle recovery following birth injury has not been explored. OBJECTIVE Using a validated rat model, we assessed the effects of nonsteroidal anti-inflammatory drug on acute and longer-term pelvic floor muscle recovery following simulated birth injury. STUDY DESIGN Three-month old Sprague Dawley rats were randomly assigned to the following groups: (1) controls, (2) simulated birth injury, (3) simulated birth injury+nonsteroidal anti-inflammatory drug, or (4) nonsteroidal anti-inflammatory drug. Simulated birth injury was induced using a well-established vaginal balloon distension protocol. Ibuprofen was administered in drinking water (0.2 mg/mL), which was consumed by the animals ad libitum. Animals were euthanized at 1, 3, 5, 7, 10, and 28 days after birth injury/ibuprofen administration. The pubocaudalis portion of the rat levator ani, which, like the human pubococcygeus, undergoes greater parturition-associated strains, was harvested (N=3-9/time point/group). The cross-sectional areas of regenerating (embryonic myosin heavy chain+) and mature myofibers were assessed at the acute and 28-day time points, respectively. The intramuscular collagen content was assessed at the 28-day time point. Myogenesis was evaluated using anti-Pax7 and anti-myogenin antibodies to identify activated and differentiated muscle stem cells, respectively. The overall immune infiltrate was assessed using anti-CD45 antibody. Expression of genes coding for pro- and anti-inflammatory cytokines was assessed by quantitative reverse transcriptase polymerase chain reaction at 3, 5, and 10 days after injury. RESULTS The pubocaudalis fiber size was significantly smaller in the simulated birth injury+nonsteroidal anti-inflammatory drug compared with the simulated birth injury group at 28 days after injury (P<.0001). The median size of embryonic myosin heavy chain+ fibers was also significantly reduced, with the fiber area distribution enriched with smaller fibers in the simulated birth injury+nonsteroidal anti-inflammatory drug group relative to the simulated birth injury group at 3 days after injury (P<.0001), suggesting a delay in the onset of regeneration in the presence of nonsteroidal anti-inflammatory drugs. By 10 days after injury, the median embryonic myosin heavy chain+ fiber size in the simulated birth injury group decreased from 7 days after injury (P<.0001) with a tight cross-sectional area distribution, indicating nearing completion of this state of regeneration. However, in the simulated birth injury+nonsteroidal anti-inflammatory drug group, the size of embryonic myosin heavy chain+ fibers continued to increase (P<.0001) with expansion of the cross-sectional area distribution, signifying a delay in regeneration in these animals. Nonsteroidal anti-inflammatory drugs decreased the muscle stem cell pool at 7 days after injury (P<.0001) and delayed muscle stem cell differentiation, as indicated by persistently elevated number of myogenin+ cells 7 days after injury (P<.05). In contrast, a proportion of myogenin+ cells returned to baseline by 5 days after injury in the simulated birth injury group. The analysis of expression of genes coding for pro- and anti-inflammatory cytokines demonstrated only transient elevation of Tgfb1 in the simulated birth injury+nonsteroidal anti-inflammatory drug group at 5 but not at 10 days after injury. Consistently with previous studies, nonsteroidal anti-inflammatory drug administration following simulated birth injury resulted in increased deposition of intramuscular collagen relative to uninjured animals. There were no significant differences in any outcomes of interest between the nonsteroidal anti-inflammatory drug group and the unperturbed controls. CONCLUSION Nonsteroidal anti-inflammatory drugs negatively impacted pelvic floor muscle regeneration in a preclinical simulated birth injury model. This appears to be driven by the negative impact of these drugs on pelvic muscle stem cell function, resulting in delayed temporal progression of pelvic floor muscle regeneration following birth injury. These findings provide impetus to investigate the impact of postpartum nonsteroidal anti-inflammatory drug administration on muscle regeneration in women at high risk for pelvic floor muscle injury.
Collapse
Affiliation(s)
- Alyssa J Kobayashi
- Division of Biological Sciences, University of California San Diego, La Jolla, CA
| | - Francesca Boscolo Sesillo
- Division of Urogynecology and Reconstructive Pelvic Surgery, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California San Diego, San Diego, CA
| | - Emmy Do
- Division of Biological Sciences, University of California San Diego, La Jolla, CA
| | - Marianna Alperin
- Division of Urogynecology and Reconstructive Pelvic Surgery, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California San Diego, San Diego, CA; Sanford Consortium for Regenerative Medicine, La Jolla, CA.
| |
Collapse
|
17
|
Yin Y, He GJ, Hu S, Tse EHY, Cheung TH. Muscle stem cell niche dynamics during muscle homeostasis and regeneration. Curr Top Dev Biol 2024; 158:151-177. [PMID: 38670704 DOI: 10.1016/bs.ctdb.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
The process of skeletal muscle regeneration involves a coordinated interplay of specific cellular and molecular interactions within the injury site. This review provides an overview of the cellular and molecular components in regenerating skeletal muscle, focusing on how these cells or molecules in the niche regulate muscle stem cell functions. Dysfunctions of muscle stem cell-to-niche cell communications during aging and disease will also be discussed. A better understanding of how niche cells coordinate with muscle stem cells for muscle repair will greatly aid the development of therapeutic strategies for treating muscle-related disorders.
Collapse
Affiliation(s)
- Yishu Yin
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, P.R. China
| | - Gary J He
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, P.R. China
| | - Shenyuan Hu
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, P.R. China
| | - Erin H Y Tse
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, P.R. China; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, P.R. China
| | - Tom H Cheung
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, P.R. China; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, P.R. China; Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, Shenzhen-Hong Kong Institute of Brain Science, HKUST Shenzhen Research Institute, Shenzhen, P.R. China.
| |
Collapse
|
18
|
Geara P, Dilworth FJ. Epigenetic integration of signaling from the regenerative environment. Curr Top Dev Biol 2024; 158:341-374. [PMID: 38670712 DOI: 10.1016/bs.ctdb.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Skeletal muscle has an extraordinary capacity to regenerate itself after injury due to the presence of tissue-resident muscle stem cells. While these muscle stem cells are the primary contributor to the regenerated myofibers, the process occurs in a regenerative microenvironment where multiple different cell types act in a coordinated manner to clear the damaged myofibers and restore tissue homeostasis. In this regenerative environment, immune cells play a well-characterized role in initiating repair by establishing an inflammatory state that permits the removal of dead cells and necrotic muscle tissue at the injury site. More recently, it has come to be appreciated that the immune cells also play a crucial role in communicating with the stem cells within the regenerative environment to help coordinate the timing of repair events through the secretion of cytokines, chemokines, and growth factors. Evidence also suggests that stem cells can help modulate the extent of the inflammatory response by signaling to the immune cells, demonstrating a cross-talk between the different cells in the regenerative environment. Here, we review the current knowledge on the innate immune response to sterile muscle injury and provide insight into the epigenetic mechanisms used by the cells in the regenerative niche to integrate the cellular cross-talk required for efficient muscle repair.
Collapse
Affiliation(s)
- Perla Geara
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI, United States
| | - F Jeffrey Dilworth
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI, United States.
| |
Collapse
|
19
|
Xu HR, Le VV, Oprescu SN, Kuang S. Muscle stem cells as immunomodulator during regeneration. Curr Top Dev Biol 2024; 158:221-238. [PMID: 38670707 PMCID: PMC11801201 DOI: 10.1016/bs.ctdb.2024.01.010] [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: 04/28/2024]
Abstract
The skeletal muscle is well known for its remarkable ability to regenerate after injuries. The regeneration is a complex and dynamic process that involves muscle stem cells (also called muscle satellite cells, MuSCs), fibro-adipogenic progenitors (FAPs), immune cells, and other muscle-resident cell populations. The MuSCs are the myogenic cell populaiton that contribute nuclei directly to the regenerated myofibers, while the other cell types collaboratively establish a microenvironment that facilitates myogenesis of MuSCs. The myogenic process includes activation, proliferation and differentiationof MuSCs, and subsequent fusion their descendent mononuclear myocytes into multinuclear myotubes. While the contributions of FAPs and immune cells to this microenvironment have been well studied, the influence of MuSCs on other cell types remains poorly understood. This review explores recent evidence supporting the potential role of MuSCs as immunomodulators during muscle regeneration, either through cytokine production or ligand-receptor interactions.
Collapse
Affiliation(s)
- H Rex Xu
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - Victor V Le
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Stephanie N Oprescu
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States; Purdue University Institute for Cancer Research, West Lafayette, IN, United States.
| |
Collapse
|
20
|
Delacher M, Schmidleithner L, Simon M, Stüve P, Sanderink L, Hotz-Wagenblatt A, Wuttke M, Schambeck K, Ruhland B, Hofmann V, Bittner S, Ritter U, Pant A, Helbich SS, Voss M, Lemmermann NA, Bessiri-Schake L, Bohn T, Eigenberger A, Menevse AN, Gebhard C, Strieder N, Abken H, Rehli M, Huehn J, Beckhove P, Hehlgans T, Junger H, Geissler EK, Prantl L, Werner JM, Schmidl C, Brors B, Imbusch CD, Feuerer M. The effector program of human CD8 T cells supports tissue remodeling. J Exp Med 2024; 221:e20230488. [PMID: 38226976 PMCID: PMC10791561 DOI: 10.1084/jem.20230488] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 10/19/2023] [Accepted: 12/06/2023] [Indexed: 01/17/2024] Open
Abstract
CD8 T lymphocytes are classically viewed as cytotoxic T cells. Whether human CD8 T cells can, in parallel, induce a tissue regeneration program is poorly understood. Here, antigen-specific assay systems revealed that human CD8 T cells not only mediated cytotoxicity but also promoted tissue remodeling. Activated CD8 T cells could produce the epidermal growth factor receptor (EGFR)-ligand amphiregulin (AREG) and sensitize epithelial cells for enhanced regeneration potential. Blocking the EGFR or the effector cytokines IFN-γ and TNF could inhibit tissue remodeling. This regenerative program enhanced tumor spheroid and stem cell-mediated organoid growth. Using single-cell gene expression analysis, we identified an AREG+, tissue-resident CD8 T cell population in skin and adipose tissue from patients undergoing abdominal wall or abdominoplasty surgery. These tissue-resident CD8 T cells showed a strong TCR clonal relation to blood PD1+TIGIT+ CD8 T cells with tissue remodeling abilities. These findings may help to understand the complex CD8 biology in tumors and could become relevant for the design of therapeutic T cell products.
Collapse
Affiliation(s)
- Michael Delacher
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, Mainz, Germany
| | - Lisa Schmidleithner
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Malte Simon
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Division of Applied Bioinformatics, German Cancer Research Center, Heidelberg, Germany
| | - Philipp Stüve
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Lieke Sanderink
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Agnes Hotz-Wagenblatt
- Core Facility Omics IT and Data Management, German Cancer Research Center, Heidelberg, Germany
| | - Marina Wuttke
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Kathrin Schambeck
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Brigitte Ruhland
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Veronika Hofmann
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Sebastian Bittner
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Uwe Ritter
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Asmita Pant
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Sara Salome Helbich
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, Mainz, Germany
| | - Morten Voss
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, Mainz, Germany
| | - Niels A. Lemmermann
- Research Center for Immunotherapy, University Medical Center Mainz, Mainz, Germany
- Institute of Virology, University Medical Center Mainz, Mainz, Germany
- Institute of Virology, University of Bonn, Bonn, Germany
| | - Lisa Bessiri-Schake
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, Mainz, Germany
| | - Toszka Bohn
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, Mainz, Germany
| | - Andreas Eigenberger
- Department of Plastic, Hand- and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Ayse Nur Menevse
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Interventional Immunology, University Regensburg, Regensburg, Germany
| | | | | | - Hinrich Abken
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Genetic Immunotherapy, University Regensburg, Regensburg, Germany
| | - Michael Rehli
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Jochen Huehn
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Hannover Medical School, Hannover, Germany
- RESIST, Cluster of Excellence 2155, Hannover Medical School, Hannover, Germany
| | - Philipp Beckhove
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Interventional Immunology, University Regensburg, Regensburg, Germany
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Thomas Hehlgans
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Henrik Junger
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Edward K. Geissler
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Lukas Prantl
- Department of Plastic, Hand- and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Jens M. Werner
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | | | - Benedikt Brors
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Faculty of Medicine Heidelberg, Heidelberg University, Heidelberg, Germany
- Division of Applied Bioinformatics, German Cancer Research Center, Heidelberg, Germany
- National Center for Tumor Diseases, Heidelberg, Germany
- German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany
| | - Charles D. Imbusch
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, Mainz, Germany
- Division of Applied Bioinformatics, German Cancer Research Center, Heidelberg, Germany
| | - Markus Feuerer
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| |
Collapse
|
21
|
Fioretti D, Ledda M, Iurescia S, Carletti R, Di Gioia C, Lolli MG, Marchese R, Lisi A, Rinaldi M. Severely Damaged Freeze-Injured Skeletal Muscle Reveals Functional Impairment, Inadequate Repair, and Opportunity for Human Stem Cell Application. Biomedicines 2023; 12:30. [PMID: 38275391 PMCID: PMC10813063 DOI: 10.3390/biomedicines12010030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND The regeneration of severe traumatic muscle injuries is an unsolved medical need that is relevant for civilian and military medicine. In this work, we produced a critically sized nonhealing muscle defect in a mouse model to investigate muscle degeneration/healing phases. MATERIALS AND METHODS We caused a freeze injury (FI) in the biceps femoris of C57BL/6N mice. From day 1 to day 25 post-injury, we conducted histological/morphometric examinations, an analysis of the expression of genes involved in inflammation/regeneration, and an in vivo functional evaluation. RESULTS We found that FI activates cytosolic DNA sensing and inflammatory responses. Persistent macrophage infiltration, the prolonged expression of eMHC, the presence of centrally nucleated myofibers, and the presence of PAX7+ satellite cells at late time points and with chronic physical impairment indicated inadequate repair. By looking at stem-cell-based therapeutic protocols of muscle repair, we investigated the crosstalk between M1-biased macrophages and human amniotic mesenchymal stem cells (hAMSCs) in vitro. We demonstrated their reciprocal paracrine effects where hAMSCs induced a shift of M1 macrophages into an anti-inflammatory phenotype, and M1 macrophages promoted an increase in the expression of hAMSC immunomodulatory factors. CONCLUSIONS Our findings support the rationale for the future use of our injury model to exploit the full potential of in vivo hAMSC transplantation following severe traumatic injuries.
Collapse
Affiliation(s)
- Daniela Fioretti
- Department Biomedical Sciences, Institute of Translational Pharmacology, National Research Council, Area di Ricerca Roma2 Tor Vergata, 00133 Rome, Italy; (M.L.); (S.I.); (M.G.L.); (A.L.)
| | - Mario Ledda
- Department Biomedical Sciences, Institute of Translational Pharmacology, National Research Council, Area di Ricerca Roma2 Tor Vergata, 00133 Rome, Italy; (M.L.); (S.I.); (M.G.L.); (A.L.)
| | - Sandra Iurescia
- Department Biomedical Sciences, Institute of Translational Pharmacology, National Research Council, Area di Ricerca Roma2 Tor Vergata, 00133 Rome, Italy; (M.L.); (S.I.); (M.G.L.); (A.L.)
| | - Raffaella Carletti
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy;
| | - Cira Di Gioia
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, 00161 Rome, Italy;
| | - Maria Grazia Lolli
- Department Biomedical Sciences, Institute of Translational Pharmacology, National Research Council, Area di Ricerca Roma2 Tor Vergata, 00133 Rome, Italy; (M.L.); (S.I.); (M.G.L.); (A.L.)
| | - Rodolfo Marchese
- Department of Clinical Pathology, FBF S. Peter Hospital, 00189 Rome, Italy;
| | - Antonella Lisi
- Department Biomedical Sciences, Institute of Translational Pharmacology, National Research Council, Area di Ricerca Roma2 Tor Vergata, 00133 Rome, Italy; (M.L.); (S.I.); (M.G.L.); (A.L.)
| | - Monica Rinaldi
- Department Biomedical Sciences, Institute of Translational Pharmacology, National Research Council, Area di Ricerca Roma2 Tor Vergata, 00133 Rome, Italy; (M.L.); (S.I.); (M.G.L.); (A.L.)
| |
Collapse
|
22
|
Andreoletti G, Romano O, Chou HJ, Sefid-Dashti MJ, Grilli A, Chen C, Lakshman N, Purushothaman P, Varfaj F, Mavilio F, Bicciato S, Urbinati F. High-throughput transcriptome analyses from ASPIRO, a phase 1/2/3 study of gene replacement therapy for X-linked myotubular myopathy. Am J Hum Genet 2023; 110:1648-1660. [PMID: 37673065 PMCID: PMC10577074 DOI: 10.1016/j.ajhg.2023.08.008] [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: 03/28/2023] [Revised: 08/09/2023] [Accepted: 08/11/2023] [Indexed: 09/08/2023] Open
Abstract
X-linked myotubular myopathy (XLMTM) is a severe congenital disease characterized by profound muscle weakness, respiratory failure, and early death. No approved therapy for XLMTM is currently available. Adeno-associated virus (AAV)-mediated gene replacement therapy has shown promise as an investigational therapeutic strategy. We aimed to characterize the transcriptomic changes in muscle biopsies of individuals with XLMTM who received resamirigene bilparvovec (AT132; rAAV8-Des-hMTM1) in the ASPIRO clinical trial and to identify potential biomarkers that correlate with therapeutic outcome. We leveraged RNA-sequencing data from the muscle biopsies of 15 study participants and applied differential expression analysis, gene co-expression analysis, and machine learning to characterize the transcriptomic changes at baseline (pre-dose) and at 24 and 48 weeks after resamirigene bilparvovec dosing. As expected, MTM1 expression levels were significantly increased after dosing (p < 0.0001). Differential expression analysis identified upregulated genes after dosing that were enriched in several pathways, including lipid metabolism and inflammatory response pathways, and downregulated genes were enriched in cell-cell adhesion and muscle development pathways. Genes involved in inflammatory and immune pathways were differentially expressed between participants exhibiting ventilator support reduction of either greater or less than 6 h/day after gene therapy compared to pre-dosing. Co-expression analysis identified similarly regulated genes, which were grouped into modules. Finally, the machine learning model identified five genes, including MTM1, as potential RNA biomarkers to monitor the progress of AAV gene replacement therapy. These findings further extend our understanding of AAV-mediated gene therapy in individuals with XLMTM at the transcriptomic level.
Collapse
Affiliation(s)
- Gaia Andreoletti
- Astellas Gene Therapies (formerly Audentes Therapeutics), San Francisco, CA, USA.
| | - Oriana Romano
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Hsin-Jung Chou
- Astellas Gene Therapies (formerly Audentes Therapeutics), San Francisco, CA, USA
| | | | - Andrea Grilli
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Clarice Chen
- Astellas Gene Therapies (formerly Audentes Therapeutics), San Francisco, CA, USA; Tox and Text Solutions, LLC, Anaheim, CA 92807, USA
| | - Neema Lakshman
- Astellas Gene Therapies (formerly Audentes Therapeutics), San Francisco, CA, USA
| | - Pravin Purushothaman
- Astellas Gene Therapies (formerly Audentes Therapeutics), San Francisco, CA, USA
| | - Fatbardha Varfaj
- Astellas Gene Therapies (formerly Audentes Therapeutics), San Francisco, CA, USA
| | - Fulvio Mavilio
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Fabrizia Urbinati
- Astellas Gene Therapies (formerly Audentes Therapeutics), San Francisco, CA, USA.
| |
Collapse
|
23
|
Thuner J, Coutant F. IFN-γ: An overlooked cytokine in dermatomyositis with anti-MDA5 antibodies. Autoimmun Rev 2023; 22:103420. [PMID: 37625674 DOI: 10.1016/j.autrev.2023.103420] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 08/22/2023] [Indexed: 08/27/2023]
Abstract
Dermatomyositis with anti-melanoma differentiation-associated gene 5 antibody (anti-MDA5 DM) is a rare autoimmune disease, often complicated by life-threatening, rapidly progressive interstitial lung disease. Additional manifestations of the disease include skin lesions, vascular abnormalities, joints and muscles pain. Despite its clinical significance, the pathogenesis of anti-MDA5 DM remains largely unknown. Currently, the disease is perceived as driven by type I interferon (IFN) whose expression is increased in most of the patients. Importantly, the regulation of IFN-γ is also altered in anti-MDA5 DM as evidenced by the presence of IFN-γ positive histiocytes in the lungs of patients, and the identification of autoantibodies that directly stimulate the production of IFN-γ by mononuclear cells. This review critically examines the pathogenesis of the disease, shedding light on recent findings that emphasize a potential role of IFN-γ. A novel conceptual framework is proposed, which integrates the molecular mechanisms altering IFN-γ regulation in anti-MDA5 DM with the known functional effects of IFN-γ on key tissues affected during the disease, such as the lungs, skin, and vessels. Understanding the precise role and relevance of IFN-γ in the pathogenesis of the disease will not only enhance the selection of available therapies for anti-MDA5 DM patients but also pave the way for the development of new therapeutic approaches targeting the altered molecular pathways.
Collapse
Affiliation(s)
- Jonathan Thuner
- Immunogenomics and Inflammation Research Team, University of Lyon, Edouard Herriot Hospital, Lyon, France; Internal medicine Department, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Frédéric Coutant
- Immunogenomics and Inflammation Research Team, University of Lyon, Edouard Herriot Hospital, Lyon, France; Immunology Department, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre-Bénite, France.
| |
Collapse
|
24
|
Gandolfi S, Pileyre B, Drouot L, Dubus I, Auquit-Auckbur I, Martinet J. Stromal vascular fraction in the treatment of myositis. Cell Death Discov 2023; 9:346. [PMID: 37726262 PMCID: PMC10509179 DOI: 10.1038/s41420-023-01605-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 08/01/2023] [Accepted: 08/14/2023] [Indexed: 09/21/2023] Open
Abstract
Muscle regeneration is a physiological process that converts satellite cells into mature myotubes under the influence of an inflammatory environment progressively replaced by an anti-inflammatory environment, with precise crosstalk between immune and muscular cells. If the succession of these phases is disturbed, the immune system can sometimes become auto-reactive, leading to chronic muscular inflammatory diseases, such as myositis. The triggers of these autoimmune myopathies remain mostly unknown, but the main mechanisms of pathogenesis are partially understood. They involve chronic inflammation, which could be associated with an auto-reactive immune response, and gradually with a decrease in the regenerative capacities of the muscle, leading to its degeneration, fibrosis and vascular architecture deterioration. Immunosuppressive treatments can block the first part of the process, but sometimes muscle remains weakened, or even still deteriorates, due to the exhaustion of its capacities. For patients refractory to immunosuppressive therapies, mesenchymal stem cells have shown interesting effects but their use is limited by their availability. Stromal vascular fraction, which can easily be extracted from adipose tissue, has shown good tolerance and possible therapeutic benefits in several degenerative and autoimmune diseases. However, despite the increasing use of stromal vascular fraction, the therapeutically active components within this heterogeneous cellular product are ill-defined and the mechanisms by which this therapy might be active remain insufficiently understood. We review herein the current knowledge on the mechanisms of action of stromal vascular fraction and hypothesise on how it could potentially respond to some of the unmet treatment needs of refractory myositis.
Collapse
Affiliation(s)
- S Gandolfi
- Univ Rouen Normandie, INSERM U1234, FOCIS Center of Excellence PAn'THER, F-76000, Rouen, France
- Toulouse University Hospital, Department of Plastic and Reconstructive Surgery, F-31000, Toulouse, France
| | - B Pileyre
- Univ Rouen Normandie, INSERM U1234, FOCIS Center of Excellence PAn'THER, F-76000, Rouen, France.
- Centre Henri Becquerel, Department of Pharmacy, F-76000, Rouen, France.
| | - L Drouot
- Univ Rouen Normandie, INSERM U1234, FOCIS Center of Excellence PAn'THER, F-76000, Rouen, France
| | - I Dubus
- Univ Rouen Normandie, INSERM U1234, FOCIS Center of Excellence PAn'THER, F-76000, Rouen, France
| | - I Auquit-Auckbur
- Univ Rouen Normandie, INSERM U1234, FOCIS Center of Excellence PAn'THER, CHU Rouen, Department of Plastic, Reconstructive and Hand Surgery, F-76000, Rouen, France
| | - J Martinet
- Univ Rouen Normandie, INSERM U1234, FOCIS Center of Excellence PAn'THER, CHU Rouen, Department of Immunology and Biotherapy, F-76000, Rouen, France
| |
Collapse
|
25
|
Carey CM, Hollins HL, Schmid AV, Gagnon JA. Distinct features of the regenerating heart uncovered through comparative single-cell profiling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.04.547574. [PMID: 37461520 PMCID: PMC10349989 DOI: 10.1101/2023.07.04.547574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Adult humans respond to heart injury by forming a permanent scar, yet other vertebrates are capable of robust and complete cardiac regeneration. Despite progress towards characterizing the mechanisms of cardiac regeneration in fish and amphibians, the large evolutionary gulf between mammals and regenerating vertebrates complicates deciphering which cellular and molecular features truly enable regeneration. To better define these features, we compared cardiac injury responses in zebrafish and medaka, two fish species that share similar heart anatomy and common teleost ancestry but differ in regenerative capability. We used single-cell transcriptional profiling to create a time-resolved comparative cell atlas of injury responses in all major cardiac cell types across both species. With this approach, we identified several key features that distinguish cardiac injury response in the non-regenerating medaka heart. By comparing immune responses to injury, we found altered cell recruitment and a distinct pro-inflammatory gene program in medaka leukocytes, and an absence of the injury-induced interferon response seen in zebrafish. In addition, we found a lack of pro-regenerative signals, including nrg1 and retinoic acid, from medaka endothelial and epicardial cells. Finally, we identified alterations in the myocardial structure in medaka, where they lack embryonic-like primordial layer cardiomyocytes, and fail to employ a cardioprotective gene program shared by regenerating vertebrates. Our findings reveal notable variation in injury response across nearly all major cardiac cell types in zebrafish and medaka, demonstrating how evolutionary divergence influences the hidden cellular features underpinning regenerative potential in these seemingly similar vertebrates.
Collapse
Affiliation(s)
- Clayton M. Carey
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - Hailey L. Hollins
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - Alexis V. Schmid
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - James A. Gagnon
- School of Biological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
- Henry Eyring Center for Cell & Genome Science, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
26
|
Morena da Silva F, Lim S, Cabrera AR, Schrems ER, Jones RG, Rosa-Caldwell ME, Washington TA, Murach KA, Greene NP. The time-course of cancer cachexia onset reveals biphasic transcriptional disruptions in female skeletal muscle distinct from males. BMC Genomics 2023; 24:374. [PMID: 37403010 DOI: 10.1186/s12864-023-09462-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 06/17/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND Cancer-cachexia (CC) is a debilitating condition affecting up to 80% of cancer patients and contributing to 40% of cancer-related deaths. While evidence suggests biological sex differences in the development of CC, assessments of the female transcriptome in CC are lacking, and direct comparisons between sexes are scarce. This study aimed to define the time course of Lewis lung carcinoma (LLC)-induced CC in females using transcriptomics, while directly comparing biological sex differences. RESULTS We found the global gene expression of the gastrocnemius muscle of female mice revealed biphasic transcriptomic alterations, with one at 1 week following tumor allograft and another during the later stages of cachexia development. The early phase was associated with the upregulation of extracellular-matrix pathways, while the later phase was characterized by the downregulation of oxidative phosphorylation, electron transport chain, and TCA cycle. When DEGs were compared to a known list of mitochondrial genes (MitoCarta), ~ 47% of these genes were differently expressed in females exhibiting global cachexia, suggesting transcriptional changes to mitochondrial gene expression happens concomitantly to functional impairments previously published. In contrast, the JAK-STAT pathway was upregulated in both the early and late stages of CC. Additionally, we observed a consistent downregulation of Type-II Interferon signaling genes in females, which was associated with protection in skeletal muscle atrophy despite systemic cachexia. Upregulation of Interferon signaling was noted in the gastrocnemius muscle of cachectic and atrophic male mice. Comparison of female tumor-bearing mice with males revealed ~ 70% of DEGs were distinct between sexes in cachectic animals, demonstrating dimorphic mechanisms of CC. CONCLUSION Our findings suggest biphasic disruptions in the transcriptome of female LLC tumor-bearing mice: an early phase associated with ECM remodeling and a late phase, accompanied by the onset of systemic cachexia, affecting overall muscle energy metabolism. Notably, ~ 2/3 of DEGs in CC are biologically sex-specific, providing evidence of dimorphic mechanisms of cachexia between sexes. Downregulation of Type-II Interferon signaling genes appears specific to CC development in females, suggesting a new biological sex-specific marker of CC not reliant on the loss of muscle mass, that might represent a protective mechanism against muscle loss in CC in female mice.
Collapse
Affiliation(s)
- Francielly Morena da Silva
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Seongkyun Lim
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Ana Regina Cabrera
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Eleanor R Schrems
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Ronald G Jones
- Molecular Muscle Mass Regulation Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Megan E Rosa-Caldwell
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Tyrone A Washington
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Kevin A Murach
- Molecular Muscle Mass Regulation Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA.
| | - Nicholas P Greene
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA.
| |
Collapse
|
27
|
Correia JC, Jannig PR, Gosztyla ML, Cervenka I, Ducommun S, Præstholm SM, Dumont K, Liu Z, Liang Q, Edsgärd D, Emanuelsson O, Gregorevic P, Westerblad H, Venckunas T, Brazaitis M, Kamandulis S, Lanner JT, Yeo GW, Ruas JL. Zfp697 is an RNA-binding protein that regulates skeletal muscle inflammation and regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.544338. [PMID: 37398033 PMCID: PMC10312635 DOI: 10.1101/2023.06.12.544338] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Muscular atrophy is a mortality risk factor that happens with disuse, chronic disease, and aging. Recovery from atrophy requires changes in several cell types including muscle fibers, and satellite and immune cells. Here we show that Zfp697/ZNF697 is a damage-induced regulator of muscle regeneration, during which its expression is transiently elevated. Conversely, sustained Zfp697 expression in mouse muscle leads to a gene expression signature of chemokine secretion, immune cell recruitment, and extracellular matrix remodeling. Myofiber-specific Zfp697 ablation hinders the inflammatory and regenerative response to muscle injury, compromising functional recovery. We uncover Zfp697 as an essential interferon gamma mediator in muscle cells, interacting primarily with ncRNAs such as the pro-regenerative miR-206. In sum, we identify Zfp697 as an integrator of cell-cell communication necessary for tissue regeneration.
Collapse
Affiliation(s)
- Jorge C. Correia
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum. Karolinska. SE-171 77, Stockholm, Sweden
| | - Paulo R. Jannig
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum. Karolinska. SE-171 77, Stockholm, Sweden
| | - Maya L. Gosztyla
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA; Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Igor Cervenka
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum. Karolinska. SE-171 77, Stockholm, Sweden
| | - Serge Ducommun
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum. Karolinska. SE-171 77, Stockholm, Sweden
| | - Stine M. Præstholm
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum. Karolinska. SE-171 77, Stockholm, Sweden
| | - Kyle Dumont
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum. Karolinska. SE-171 77, Stockholm, Sweden
| | - Zhengye Liu
- Molecular Muscle Physiology and Pathophysiology. Department of Physiology and Pharmacology, Biomedicum. Karolinska Institutet. SE-171 77, Stockholm. Sweden
| | - Qishan Liang
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA; Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Daniel Edsgärd
- Science for Life Laboratory, Department of Gene Technology, School of Engineering Sciences in Biotechnology, Chemistry and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Olof Emanuelsson
- Science for Life Laboratory, Department of Gene Technology, School of Engineering Sciences in Biotechnology, Chemistry and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Paul Gregorevic
- Centre for Muscle Research, Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Håkan Westerblad
- Muscle Physiology, Department of Physiology and Pharmacology, Biomedicum. Karolinska. SE-171 77, Stockholm, Sweden
| | - Tomas Venckunas
- Institute of Sports Science and Innovations, Lithuanian Sports University, 44221 Kaunas, Lithuania
| | - Marius Brazaitis
- Institute of Sports Science and Innovations, Lithuanian Sports University, 44221 Kaunas, Lithuania
| | - Sigitas Kamandulis
- Institute of Sports Science and Innovations, Lithuanian Sports University, 44221 Kaunas, Lithuania
| | - Johanna T. Lanner
- Molecular Muscle Physiology and Pathophysiology. Department of Physiology and Pharmacology, Biomedicum. Karolinska Institutet. SE-171 77, Stockholm. Sweden
| | - Gene W. Yeo
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA; Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jorge L. Ruas
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum. Karolinska. SE-171 77, Stockholm, Sweden
| |
Collapse
|
28
|
Scala P, Manzo P, Longo R, Giudice V, Ciardulli MC, Serio B, Selleri C, Guadagno L, Rehak L, Maffulli N, Della Porta G. Contribution of peripheral blood mononuclear cells isolated by advanced filtration system to myogenesis of human bone marrow mesenchymal stem cells co-cultured with myoblasts. Heliyon 2023; 9:e17141. [PMID: 37484299 PMCID: PMC10361327 DOI: 10.1016/j.heliyon.2023.e17141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 07/25/2023] Open
Abstract
Background Contribution of peripheral blood mononuclear cells (PBMCs) in myogenesis is still under debate, even though blood filtration systems are commonly used in clinical practice for successfully management of critic limb ischemia. Objectives A commercial blood filter used for autologous human PBMC transplantation procedures is characterized and used to collect PBMCs, that are then added to well-established 2D in vitro myogenic models assembled with a co-culture of human bone marrow-derived mesenchymal stem cells (hBM-MSCs) and skeletal myoblasts (hSkMs) whit the aim of investigating their potential contribution to stem cell myogenic commitment. Methods A commercial blood filter was physically and chemically studied to understand its morphological characteristics and composition. PBMCs were concentrated using this system, further isolated by Ficoll-Paque density gradient centrifugation, and then added in an upper transwell chamber to a 2D co-culture of hBM-MSCs and hSkMs. Myogenic commitment was investigated by RT-PCR, immunofluorescence, and flow cytometry immunophenotyping. Cytokine levels were monitored by ELISA assay in culture media. Results The blood filtration system was disassembled and appeared to be formed by twelve membranes of poly-butylene terephthalate fibers (diameters, 0.9-4.0 μm) with pore size distribution of 1-20 μm. Filter functional characterization was achieved by characterizing collected cells by flow cytometry. Subsequently, collected PBMCs fraction was added to an in-vitro model of hBM-MSC myogenic commitment. In the presence of PBMCs, stem cells significantly upregulated myogenic genes, such as Desmin and MYH2, as confirmed by qRT-PCR and expressed related proteins by immunofluorescence (IF) assay, while downregulated pro-inflammatory cytokines (IL12A at day 14) along the 21 days of culture. Novelty Our work highlights chemical-physical properties of commercial blood filter and suggests that blood filtrated fraction of PBMC might modulate cytokine expression in response to muscle injury and promote myogenic events, supporting their clinical use in autologous transplantation.
Collapse
Affiliation(s)
- Pasqualina Scala
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 43, 84081 Baronissi SA, Italy
| | - Paola Manzo
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D'Aragona”, Largo Città d'Ippocrate, 1, 84131 Salerno SA, Italy
| | - Raffaele Longo
- Department of Industrial Engineering, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano SA, Italy
| | - Valentina Giudice
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 43, 84081 Baronissi SA, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D'Aragona”, Largo Città d'Ippocrate, 1, 84131 Salerno SA, Italy
| | - Maria Camilla Ciardulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 43, 84081 Baronissi SA, Italy
| | - Bianca Serio
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D'Aragona”, Largo Città d'Ippocrate, 1, 84131 Salerno SA, Italy
| | - Carmine Selleri
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 43, 84081 Baronissi SA, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D'Aragona”, Largo Città d'Ippocrate, 1, 84131 Salerno SA, Italy
| | - Liberata Guadagno
- Department of Industrial Engineering, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano SA, Italy
| | - Laura Rehak
- Athena Biomedical Innovations, Viale Europa 139, Florence, 50126, Italy
| | - Nicola Maffulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 43, 84081 Baronissi SA, Italy
- Centre for Sports and Exercise Medicine, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 275 Bancroft Road, London E1 4DG, UK
| | - Giovanna Della Porta
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 43, 84081 Baronissi SA, Italy
- Interdepartment Centre BIONAM, Università di Salerno, via Giovanni Paolo II, 132, 84084 Fisciano SA, Italy
| |
Collapse
|
29
|
Hanna BS, Wang G, Galván-Peña S, Mann AO, Ramirez RN, Muñoz-Rojas AR, Smith K, Wan M, Benoist C, Mathis D. The gut microbiota promotes distal tissue regeneration via RORγ + regulatory T cell emissaries. Immunity 2023; 56:829-846.e8. [PMID: 36822206 PMCID: PMC10101925 DOI: 10.1016/j.immuni.2023.01.033] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 12/22/2022] [Accepted: 01/30/2023] [Indexed: 02/24/2023]
Abstract
Specific microbial signals induce the differentiation of a distinct pool of RORγ+ regulatory T (Treg) cells crucial for intestinal homeostasis. We discovered highly analogous populations of microbiota-dependent Treg cells that promoted tissue regeneration at extra-gut sites, notably acutely injured skeletal muscle and fatty liver. Inflammatory meditators elicited by tissue damage combined with MHC-class-II-dependent T cell activation to drive the accumulation of gut-derived RORγ+ Treg cells in injured muscle, wherein they regulated the dynamics and tenor of early inflammation and helped balance the proliferation vs. differentiation of local stem cells. Reining in IL-17A-producing T cells was a major mechanism underlying the rheostatic functions of RORγ+ Treg cells in compromised tissues. Our findings highlight the importance of gut-trained Treg cell emissaries in controlling the response to sterile injury of non-mucosal tissues.
Collapse
Affiliation(s)
- Bola S Hanna
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Gang Wang
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Silvia Galván-Peña
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Alexander O Mann
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Ricardo N Ramirez
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Andrés R Muñoz-Rojas
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Kathleen Smith
- Internal Medicine Research Unit, Worldwide Research, Development & Medical, Pfizer Inc., Cambridge, MA, USA
| | - Min Wan
- Internal Medicine Research Unit, Worldwide Research, Development & Medical, Pfizer Inc., Cambridge, MA, USA
| | - Christophe Benoist
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
30
|
Zhuang S, Russell A, Guo Y, Xu Y, Xiao W. IFN-γ blockade after genetic inhibition of PD-1 aggravates skeletal muscle damage and impairs skeletal muscle regeneration. Cell Mol Biol Lett 2023; 28:27. [PMID: 37016287 PMCID: PMC10071770 DOI: 10.1186/s11658-023-00439-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/15/2023] [Indexed: 04/06/2023] Open
Abstract
BACKGROUND Innate immune responses play essential roles in skeletal muscle recovery after injury. Programmed cell death protein 1 (PD-1) contributes to skeletal muscle regeneration by promoting macrophage proinflammatory to anti-inflammatory phenotype transition. Interferon (IFN)-γ induces proinflammatory macrophages that appear to hinder myogenesis in vitro. Therefore, we tested the hypothesis that blocking IFN-γ in PD-1 knockout mice may dampen inflammation and promote skeletal muscle regeneration via regulating the macrophage phenotype and neutrophils. METHODS Anti-IFN-γ antibody was administered in PD-1 knockout mice, and cardiotoxin (CTX) injection was performed to induce acute skeletal muscle injury. Hematoxylin and eosin (HE) staining was used to view morphological changes of injured and regenerated skeletal muscle. Masson's trichrome staining was used to assess the degree of fibrosis. Gene expressions of proinflammatory and anti-inflammatory factors, fibrosis-related factors, and myogenic regulator factors were determined by real-time polymerase chain reaction (PCR). Changes in macrophage phenotype were examined by western blot and real-time PCR. Immunofluorescence was used to detect the accumulation of proinflammatory macrophages, anti-inflammatory macrophages, and neutrophils. RESULTS IFN-γ blockade in PD-1 knockout mice did not alleviate skeletal muscle damage or improve regeneration following acute cardiotoxin-induced injury. Instead, it exacerbated skeletal muscle inflammation and fibrosis, and impaired regeneration via inhibiting macrophage accumulation, blocking macrophage proinflammatory to anti-inflammatory transition, and enhancing infiltration of neutrophils. CONCLUSION IFN-γ is crucial for efficient skeletal muscle regeneration in the absence of PD-1.
Collapse
Affiliation(s)
- Shuzhao Zhuang
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences, Shanghai University of Sport, Ministry of Education, Shanghai, China
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Aaron Russell
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Yifan Guo
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences, Shanghai University of Sport, Ministry of Education, Shanghai, China
| | - Yingying Xu
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences, Shanghai University of Sport, Ministry of Education, Shanghai, China
| | - Weihua Xiao
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China.
- Key Laboratory of Exercise and Health Sciences, Shanghai University of Sport, Ministry of Education, Shanghai, China.
| |
Collapse
|
31
|
Ferrara PJ, Reidy PT, Petrocelli JJ, Yee EM, Fix DK, Mahmassani ZS, Montgomery JA, McKenzie AI, de Hart NMMP, Drummond MJ. Global deletion of CCL2 has adverse impacts on recovery of skeletal muscle fiber size and function and is muscle specific. J Appl Physiol (1985) 2023; 134:923-932. [PMID: 36861669 PMCID: PMC10069960 DOI: 10.1152/japplphysiol.00444.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 02/17/2023] [Accepted: 02/25/2023] [Indexed: 03/03/2023] Open
Abstract
Timely and complete recovery of muscle mass and function following a bout of physical disuse are critical components of returning to normal activities of daily living and lifestyle. Proper cross talk between the muscle tissue and myeloid cells (e.g., macrophages) throughout the recovery period from disuse atrophy plays a significant role in the complete resolution of muscle size and function. Chemokine C-C motif ligand 2 (CCL2) has a critical function of recruiting macrophages during the early phase of muscle damage. However, the importance of CCL2 has not been defined in the context of disuse and recovery. Here, we utilized a mouse model of whole body CCL2 deletion (CCL2KO) and subjected them to a period of hindlimb unloading followed by reloading to investigate the importance of CCL2 on the regrowth of muscle following disuse atrophy using ex vivo muscle tests, immunohistochemistry, and fluorescence-activated cell sorting approaches. We show mice that lack CCL2 display an incomplete recovery of gastrocnemius muscle mass, myofiber cross-sectional area, and EDL muscle contractile characteristics during the recovery from disuse atrophy. The soleus and plantaris had limited impact as a result of CCL2 deficiency suggesting a muscle-specific effect. Mice that lack CCL2 have decreased skeletal muscle collagen turnover, which may be related to defects in muscle function and stiffness. In addition, we show that the recruitment of macrophages to gastrocnemius muscle was dramatically reduced in CCL2KO mice during the recovery from disuse atrophy, which likely precipitated poor recovery of muscle size and function and aberrant collagen remodeling.NEW & NOTEWORTHY We provide evidence that the whole body loss of CCL2 in mice has adverse impacts on whole body function and skeletal muscle-specific contractile characteristics and collagen content. These defects in muscle function worsened during the recovery from disuse atrophy and corresponded with decreased recovery of muscle mass. We conclude that the absence of CCL2 decreased recruitment of proinflammatory macrophages to the muscle during the regrowth phase following disuse atrophy resulting in impaired collagen remodeling events and full resolution of muscle morphology and function.
Collapse
Affiliation(s)
- Patrick J Ferrara
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States
| | - Paul T Reidy
- Department of Kinesiology, Nutrition and Health, Miami University, Oxford, Ohio, United States
| | - Jonathan J Petrocelli
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, United States
| | - Elena M Yee
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, United States
| | - Dennis K Fix
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States
| | - Ziad S Mahmassani
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, United States
| | - Jessie A Montgomery
- Department of Chemistry, University of Utah, Salt Lake City, Utah, United States
| | - Alec I McKenzie
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, United States
| | - Naomi M M P de Hart
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| | - Micah J Drummond
- 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
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
32
|
Huang W, Kraynak CA, Bender EC, Farrar RP, Suggs LJ. Soluble components from mesenchymal stromal cell processing exert anti-inflammatory effects and facilitate ischemic muscle regeneration. Cytotherapy 2023; 25:375-386. [PMID: 36543717 PMCID: PMC10006307 DOI: 10.1016/j.jcyt.2022.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/16/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND AIMS Skeletal muscle regeneration after severe damage is reliant on local stem cell proliferation and differentiation, processes that are tightly regulated by macrophages. Peripheral artery disease is a globally prevalent cardiovascular disease affecting millions of people. Progression of the disease leads to intermittent claudication, subsequent critical limb ischemia and muscle injury. Tissue-derived and ex vivo-expanded mesenchymal stromal cells (MSCs) for skeletal muscle regeneration have been studied, but pre-clinical and clinical results have not been consistent. As a result, the potential therapeutic efficacy and associated repair mechanisms of MSCs remain unclear. Numerous studies have demonstrated the vulnerability of delivered MSCs, with a precipitous drop in cell viability upon transplantation. This has prompted investigation into the therapeutic benefit of apoptotic cells, microvesicles, exosomes and soluble signals that are released upon cell death. METHODS In this study, we characterized various components produced by MSCs after cell death induction under different conditions. We discovered anti-inflammatory and pro-regenerative effects produced by cell components following a freeze and thaw (F&T) process on macrophage polarization in vitro. We further investigated the underlying mechanisms of macrophage polarization by those components resulting from severe cell death induction. RESULTS We found potent therapeutic effects from F&T-induced cell debris are dependent on the externalization of phosphatidylserine on the plasma membrane. In contrast, effects from the supernatant of F&T-induced cell death primarily depends on the released protein content. We then applied the F&T-induced cell supernatant to an animal model of peripheral artery disease to treat muscle injury caused by severe ischemia. Treatment with the F&T supernatant but not the vulnerable MSCs resulted in significantly improved recovery of muscle function, blood flow and morphology and inflammation resolution in the affected muscles 2 weeks after injury. CONCLUSIONS This study validates the therapeutic potential of F&T-induced supernatant obviating the need for a viable population from vulnerable MSCs to treat injury, thus providing a roadmap for cell-free therapeutic approaches for tissue regeneration.
Collapse
Affiliation(s)
- Wenbai Huang
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA; Department of Kinesiology and Health Education, University of Texas at Austin, Austin, Texas, USA
| | - Chelsea A Kraynak
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Elizabeth C Bender
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Roger P Farrar
- Department of Kinesiology and Health Education, University of Texas at Austin, Austin, Texas, USA
| | - Laura J Suggs
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA.
| |
Collapse
|
33
|
Andre AB, Rees KP, O’Connor S, Severson GW, Newbern JM, Wilson-Rawls J, Plaisier CL, Rawls A. Single cell analysis reveals satellite cell heterogeneity for proinflammatory chemokine expression. Front Cell Dev Biol 2023; 11:1084068. [PMID: 37051469 PMCID: PMC10083252 DOI: 10.3389/fcell.2023.1084068] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 03/16/2023] [Indexed: 03/28/2023] Open
Abstract
Background: The expression of proinflammatory signals at the site of muscle injury are essential for efficient tissue repair and their dysregulation can lead to inflammatory myopathies. Macrophages, neutrophils, and fibroadipogenic progenitor cells residing in the muscle are significant sources of proinflammatory cytokines and chemokines. However, the inducibility of the myogenic satellite cell population and their contribution to proinflammatory signaling is less understood.Methods: Mouse satellite cells were isolated and exposed to lipopolysaccharide (LPS) to mimic sterile skeletal muscle injury and changes in the expression of proinflammatory genes was examined by RT-qPCR and single cell RNA sequencing. Expression patterns were validated in skeletal muscle injured with cardiotoxin by RT-qPCR and immunofluorescence.Results: Satellite cells in culture were able to express Tnfa, Ccl2, and Il6, within 2 h of treatment with LPS. Single cell RNA-Seq revealed seven cell clusters representing the continuum from activation to differentiation. LPS treatment led to a heterogeneous pattern of induction of C-C and C-X-C chemokines (e.g., Ccl2, Ccl5, and Cxcl0) and cytokines (e.g., Tgfb1, Bmp2, Il18, and Il33) associated with innate immune cell recruitment and satellite cell proliferation. One cell cluster was enriched for expression of the antiviral interferon pathway genes under control conditions and LPS treatment. Activation of this pathway in satellite cells was also detectable at the site of cardiotoxin induced muscle injury.Conclusion: These data demonstrate that satellite cells respond to inflammatory signals and secrete chemokines and cytokines. Further, we identified a previously unrecognized subset of satellite cells that may act as sensors for muscle infection or injury using the antiviral interferon pathway.
Collapse
Affiliation(s)
- Alexander B. Andre
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
- Molecular and Cellular Biology Graduate Program, Arizona State University, Tempe, AZ, United States
| | - Katherina P. Rees
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
- Molecular and Cellular Biology Graduate Program, Arizona State University, Tempe, AZ, United States
| | - Samantha O’Connor
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
- Biomedical Engineering Graduate Program, Arizona State University, Tempe, AZ, United States
| | - Grant W. Severson
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
- Molecular and Cellular Biology Graduate Program, Arizona State University, Tempe, AZ, United States
| | - Jason M. Newbern
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | | | - Christopher L. Plaisier
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
| | - Alan Rawls
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
- *Correspondence: Alan Rawls,
| |
Collapse
|
34
|
Hanna BS, Yaghi OK, Langston PK, Mathis D. The potential for Treg-enhancing therapies in tissue, in particular skeletal muscle, regeneration. Clin Exp Immunol 2023; 211:138-148. [PMID: 35972909 PMCID: PMC10019136 DOI: 10.1093/cei/uxac076] [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: 05/11/2022] [Revised: 06/29/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Foxp3+CD4+ regulatory T cells (Tregs) are famous for their role in maintaining immunological tolerance. With their distinct transcriptomes, growth-factor dependencies and T-cell receptor (TCR) repertoires, Tregs in nonlymphoid tissues, termed "tissue-Tregs," also perform a variety of functions to help assure tissue homeostasis. For example, they are important for tissue repair and regeneration after various types of injury, both acute and chronic. They exert this influence by controlling both the inflammatory tenor and the dynamics of the parenchymal progenitor-cell pool in injured tissues, thereby promoting efficient repair and limiting fibrosis. Thus, tissue-Tregs are seemingly attractive targets for immunotherapy in the context of tissue regeneration, offering several advantages over existing therapies. Using skeletal muscle as a model system, we discuss the existing literature on Tregs' role in tissue regeneration in acute and chronic injuries, and various approaches for their therapeutic modulation in such contexts, including exercise as a natural Treg modulator.
Collapse
Affiliation(s)
- Bola S Hanna
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital; Boston, USA
| | - Omar K Yaghi
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital; Boston, USA
| | - P Kent Langston
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital; Boston, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital; Boston, USA
| |
Collapse
|
35
|
Benítez S, Figueroa Á, Lagos NA, Silva AX, Duarte C, Vargas CA, Lardies MA, Cárdenas L. Differential gene expression analysis in the scallop Argopecten purpuratus exposed to altered pH and temperature conditions in an upwelling-influenced farming area. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2023; 45:101046. [PMID: 36495831 DOI: 10.1016/j.cbd.2022.101046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 11/17/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022]
Abstract
Increased carbon dioxide in the atmosphere and its absorption across the ocean surface will alter natural variations in pH and temperature levels, occurring in coastal upwelling ecosystems. The scallop Argopecten purpuratus, one of the most economically important species farmed in northern Chile, has been shown to be vulnerable to these environmental drivers. However, the regulatory responses at the gene-level of scallops to these climate stressors remain almost unknown. Consequently, we used an orthogonal experimental design and RNAseq approach to analyze the acute effects of variability in pH and temperature on gene expression in the muscle tissue of A. purpuratus. In respect to control conditions (pH ~ 8.0/ 14 °C), the influence of low pH (~ 7.7) and temperature (14 °C) induced the activation of several genes associated with apoptotic signaling pathways and protein localization to plasma membrane. Elevated temperature (18 °C) and pH (~8.0) conditions increased the expression of transcripts associated with the activation of muscle contraction, regulation, and sarcomere organization effects on muscle tissue. In scallops exposed to low pH and elevated temperature, the genes expressed were differentially associated with the oxidation-reduction process, signal translation, and positive regulation of GTPase activity. These results indicated that the differentially expressed genes under the experimental conditions tested are mainly related to the mitigation of cellular damage and homeostasis control. Our results add knowledge about the function of the adductor muscle in response to stressors in scallops. Furthermore, these results could help in the identification of molecular biomarkers of stress necessary to be integrated into the aquaculture programs for the mitigation of climate change.
Collapse
Affiliation(s)
- Samanta Benítez
- Programa de Doctorado en Biología Marina, Instituto de Ciencias Marinas y Limnológicas, Universidad Austral de Chile, Valdivia, Chile; Centro de Investigación e Innovación para el Cambio Climático, Facultad de Ciencias, Universidad Santo Tomás, Santiago, Chile
| | - Álvaro Figueroa
- Instituto de Ciencias Ambientales y Evolutivas, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Nelson A Lagos
- Centro de Investigación e Innovación para el Cambio Climático, Facultad de Ciencias, Universidad Santo Tomás, Santiago, Chile; Instituto Milenio de Socio-Ecología Costera (SECOS), Santiago, Chile
| | - Andrea X Silva
- AUSTRAL-omics, Vicerrectoría de Investigación, Desarrollo y Creación Artística, Universidad Austral de Chile, Chile
| | - Cristian Duarte
- Departamento de Ecología y Biodiversidad, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Cristian A Vargas
- Instituto Milenio de Socio-Ecología Costera (SECOS), Santiago, Chile; Laboratorio de Ecosistemas Costeros y Cambio Ambiental Global (ECCALab), Facultad de Ciencias Ambientales & Centro EULA Chile, Universidad de Concepción, Concepción, Chile
| | - Marco A Lardies
- Instituto Milenio de Socio-Ecología Costera (SECOS), Santiago, Chile; Departamento de Ciencias, Facultad de Artes Liberales, Universidad Adolfo Ibañez, Santiago, Chile
| | - Leyla Cárdenas
- Centro FONDAP de Investigación en Dinámica de Ecosistemas Marinos de Altas Latitudes (IDEAL), Valdivia, Chile.
| |
Collapse
|
36
|
Scala P, Manzo P, Lamparelli EP, Lovecchio J, Ciardulli MC, Giudice V, Selleri C, Giordano E, Rehak L, Maffulli N, Della Porta G. Peripheral blood mononuclear cells contribute to myogenesis in a 3D bioengineered system of bone marrow mesenchymal stem cells and myoblasts. Front Bioeng Biotechnol 2023; 10:1075715. [PMID: 36704300 PMCID: PMC9871311 DOI: 10.3389/fbioe.2022.1075715] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/28/2022] [Indexed: 01/12/2023] Open
Abstract
In this work, a 3D environment obtained using fibrin scaffold and two cell populations, such as bone marrow-derived mesenchymal stem cells (BM-MSCs), and primary skeletal muscle cells (SkMs), was assembled. Peripheral blood mononuclear cells (PBMCs) fraction obtained after blood filtration with HemaTrate® filter was then added to the 3D culture system to explore their influence on myogenesis. The best cell ratio into a 3D fibrin hydrogel was 1:1 (BM-MSCs plus SkMs:PBMCs) when cultured in a perfusion bioreactor; indeed, excellent viability and myogenic event induction were observed. Myogenic genes were significantly overexpressed when cultured with PBMCs, such as MyoD1 of 118-fold at day 14 and Desmin 6-fold at day 21. Desmin and Myosin Heavy Chain were also detected at protein level by immunostaining along the culture. Moreover, the presence of PBMCs in 3D culture induced a significant downregulation of pro-inflammatory cytokine gene expression, such as IL6. This smart biomimetic environment can be an excellent tool for investigation of cellular crosstalk and PBMC influence on myogenic processes.
Collapse
Affiliation(s)
- Pasqualina Scala
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy
| | - Paola Manzo
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy,Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, Salerno, Italy
| | | | - Joseph Lovecchio
- Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi” (DEI), University of Bologna, Bologna, Italy
| | | | - Valentina Giudice
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy,Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, Salerno, Italy
| | - Carmine Selleri
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy,Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, Salerno, Italy
| | - Emanuele Giordano
- Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi” (DEI), University of Bologna, Bologna, Italy
| | - Laura Rehak
- Athena Biomedical innovations, Florence, Italy
| | - Nicola Maffulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy,Centre for Sports and Exercise Medicine, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, England
| | - Giovanna Della Porta
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy,Interdepartment Centre BIONAM, University of Salerno, Fisciano, Italy,*Correspondence: Giovanna Della Porta,
| |
Collapse
|
37
|
Wang Y, Lu J, Liu Y. Skeletal Muscle Regeneration in Cardiotoxin-Induced Muscle Injury Models. Int J Mol Sci 2022; 23:ijms232113380. [PMID: 36362166 PMCID: PMC9657523 DOI: 10.3390/ijms232113380] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Skeletal muscle injuries occur frequently in daily life and exercise. Understanding the mechanisms of regeneration is critical for accelerating the repair and regeneration of muscle. Therefore, this article reviews knowledge on the mechanisms of skeletal muscle regeneration after cardiotoxin-induced injury. The process of regeneration is similar in different mouse strains and is inhibited by aging, obesity, and diabetes. Exercise, microcurrent electrical neuromuscular stimulation, and mechanical loading improve regeneration. The mechanisms of regeneration are complex and strain-dependent, and changes in functional proteins involved in the processes of necrotic fiber debris clearance, M1 to M2 macrophage conversion, SC activation, myoblast proliferation, differentiation and fusion, and fibrosis and calcification influence the final outcome of the regenerative activity.
Collapse
|
38
|
TNFα and IFNγ cooperate for efficient pro- to anti-inflammatory transition of macrophages during muscle regeneration. Proc Natl Acad Sci U S A 2022; 119:e2209976119. [PMID: 36279473 PMCID: PMC9636974 DOI: 10.1073/pnas.2209976119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
IFNγ is traditionally known as a proinflammatory cytokine with diverse roles in antimicrobial and antitumor immunity. Yet, findings regarding its sources and functions during the regeneration process following a sterile injury are conflicting. Here, we show that natural killer (NK) cells are the main source of IFNγ in regenerating muscle. Beyond this cell population, IFNγ production is limited to a small population of T cells. We further show that NK cells do not play a major role in muscle regeneration following an acute injury or in dystrophic mice. Surprisingly, the absence of IFNγ per se also has no effect on muscle regeneration following an acute injury. However, the role of IFNγ is partially unmasked when TNFα is also neutralized, suggesting a compensatory mechanism. Using transgenic mice, we showed that conditional inhibition of IFNGR1 signaling in muscle stem cells or fibro-adipogenic progenitors does not play a major role in muscle regeneration. In contrast to common belief, we found that IFNγ is not present in the early inflammatory phase of the regeneration process but rather peaks when macrophages are acquiring an anti-inflammatory phenotype. Further transcriptomic analysis suggests that IFNγ cooperates with TNFα to regulate the transition of macrophages from pro- to anti-inflammatory states. The absence of the cooperative effect of these cytokines on macrophages, however, does not result in significant regeneration impairment likely due to the presence of other compensatory mechanisms. Our findings support the arising view of IFNγ as a pleiotropic inflammatory regulator rather than an inducer of the inflammatory response.
Collapse
|
39
|
Waldemer-Streyer RJ, Kim D, Chen J. Muscle cell-derived cytokines in skeletal muscle regeneration. FEBS J 2022; 289:6463-6483. [PMID: 35073461 PMCID: PMC9308828 DOI: 10.1111/febs.16372] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 01/13/2022] [Accepted: 01/21/2022] [Indexed: 12/14/2022]
Abstract
Regeneration of the mammalian adult skeletal muscle is a well-orchestrated process regulated by multiple proteins and signalling pathways. Cytokines constitute a major class of regulators of skeletal myogenesis. It is well established that infiltrating immune cells at the site of muscle injury secrete cytokines, which play critical roles in the myofibre repair and regeneration process. In the past 10-15 years, skeletal muscle itself has emerged as a prolific producer of cytokines. Much attention in the field has been focused on the endocrine effects of muscle-secreted cytokines (myokines) on metabolic regulation. However, ample evidence suggests that muscle-derived cytokines also regulate myogenic differentiation and muscle regeneration in an autocrine manner. In this review, we survey cytokines that meet two criteria: (a) evidence of expression by muscle cells; (b) evidence demonstrating a myogenic function. Dozens of cytokines representing several major classes make up this group, and together they regulate all steps of the myogenic process. How such a large array of cytokines coordinate their signalling to form a regulatory network is a fascinating, pressing question. Functional studies that can distinguish the source of the cytokines in vivo are also much needed in order to facilitate exploration of their full therapeutic potential.
Collapse
Affiliation(s)
| | | | - Jie Chen
- Department of Cell & Developmental Biology, University of Illinois at Urbana-Champaign, 601 S. Goodwin Ave., Urbana, IL 61801
| |
Collapse
|
40
|
Nakanishi N, Ono Y, Miyazaki Y, Moriyama N, Fujioka K, Yamashita K, Inoue S, Kotani J. Sepsis causes neutrophil infiltration in muscle leading to muscle atrophy and weakness in mice. Front Immunol 2022; 13:950646. [PMID: 36389802 PMCID: PMC9659852 DOI: 10.3389/fimmu.2022.950646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 10/19/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Sepsis-induced muscle atrophy leads to prolonged physical dysfunction. Although the interaction of muscle atrophy and macrophage has been reported in sepsis, the role of neutrophils in muscle atrophy has not been thoroughly investigated. This study sought to investigate the long-term changes in muscle-localized neutrophils after sepsis induction and their possible role in sepsis. METHODS Sepsis was induced in seven-week-old male C57BL/6J mice 8-12 (cecal slurry [CS] model) via intraperitoneal injection of 1 mg/g cecal slurry. The percentage change in body weight and grip strength was evaluated. The tibialis anterior muscles were dissected for microscopic examination of the cross-sectional area of myofibers or Fluorescence-activated cell sorting (FACS) analysis of immune cells. These changes were evaluated in the following conditions: (1) Longitudinal change until day 61, (2) CS concentration-dependent change on day 14 at the low (0.3 mg/g), middle (1.0 mg/g), and high (2.0 mg/g) concentrations, and (3) CS mice on day 14 treated with an anti-Ly6G antibody that depletes neutrophils. RESULTS Body weight and grip strength were significantly lower in the CS model until day 61 (body weight: 123.1% ± 1.8% vs. 130.3% ± 2.5%, p = 0.04; grip strength: 104.5% ± 3.8% vs. 119.3% ± 5.3%, p = 0.04). Likewise, cross-sectional muscle area gradually decreased until day 61 from the CS induction (895.6 [606.0-1304.9] μm2 vs. 718.8 [536.2-937.0] μm2, p < 0.01). The number of muscle-localized neutrophils increased from 2.3 ± 0.6 cell/mg on day 0 to 22.2 ± 13.0 cell/mg on day 14, and decreased thereafter. In terms of CS concentration-dependent change, cross-sectional area was smaller (484.4 ± 221.2 vs. 825.8 ± 436.2 μm2 [p < 0.001]) and grip strength was lower (71.4% ± 12.8% vs. 116.3% ± 7.4%, p = 0.01) in the CS High group compared with the control, with increased neutrophils (p = 0.03). Ly6G-depleted mice demonstrated significant increase of muscle cross-sectional area and grip strength compared with control mice (p < 0.01). CONCLUSIONS Sepsis causes infiltration of neutrophils in muscles, leading to muscle atrophy and weakness. Depletion of neutrophils in muscle reverses sepsis-induced muscle atrophy and weakness. These results suggest that neutrophils may play a critical role in sepsis-induced muscle atrophy and weakness.
Collapse
Affiliation(s)
- Nobuto Nakanishi
- Division of Disaster and Emergency Medicine, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuko Ono
- Division of Disaster and Emergency Medicine, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yusuke Miyazaki
- Division of Disaster and Emergency Medicine, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naoki Moriyama
- Division of Disaster and Emergency Medicine, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kazumichi Fujioka
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kimihiro Yamashita
- Division of Gastrointestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shigeaki Inoue
- Division of Disaster and Emergency Medicine, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Joji Kotani
- Division of Disaster and Emergency Medicine, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
41
|
Bernard C, Zavoriti A, Pucelle Q, Chazaud B, Gondin J. Role of macrophages during skeletal muscle regeneration and hypertrophy-Implications for immunomodulatory strategies. Physiol Rep 2022; 10:e15480. [PMID: 36200266 PMCID: PMC9535344 DOI: 10.14814/phy2.15480] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/12/2022] [Indexed: 06/16/2023] Open
Abstract
Skeletal muscle is a plastic tissue that regenerates ad integrum after injury and adapts to raise mechanical loading/contractile activity by increasing its mass and/or myofiber size, a phenomenon commonly refers to as skeletal muscle hypertrophy. Both muscle regeneration and hypertrophy rely on the interactions between muscle stem cells and their neighborhood, which include inflammatory cells, and particularly macrophages. This review first summarizes the role of macrophages in muscle regeneration in various animal models of injury and in response to exercise-induced muscle damage in humans. Then, the potential contribution of macrophages to skeletal muscle hypertrophy is discussed on the basis of both animal and human experiments. We also present a brief comparative analysis of the role of macrophages during muscle regeneration versus hypertrophy. Finally, we summarize the current knowledge on the impact of different immunomodulatory strategies, such as heat therapy, cooling, massage, nonsteroidal anti-inflammatory drugs and resolvins, on skeletal muscle regeneration and their potential impact on muscle hypertrophy.
Collapse
Affiliation(s)
- Clara Bernard
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du MuscleUniversité Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Université LyonLyonFrance
| | - Aliki Zavoriti
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du MuscleUniversité Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Université LyonLyonFrance
| | - Quentin Pucelle
- Université de Versailles Saint‐Quentin‐En‐YvelinesVersaillesFrance
| | - Bénédicte Chazaud
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du MuscleUniversité Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Université LyonLyonFrance
| | - Julien Gondin
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du MuscleUniversité Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Université LyonLyonFrance
| |
Collapse
|
42
|
Gabellini D. A regenerative niche for stem cells. Science 2022; 377:578-579. [PMID: 35926040 DOI: 10.1126/science.add6804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Production of hyaluronic acid allows regenerative signaling in muscle stem cells after injury.
Collapse
Affiliation(s)
- Davide Gabellini
- Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milano, Italy
| |
Collapse
|
43
|
Nakka K, Hachmer S, Mokhtari Z, Kovac R, Bandukwala H, Bernard C, Li Y, Xie G, Liu C, Fallahi M, Megeney LA, Gondin J, Chazaud B, Brand M, Zha X, Ge K, Dilworth FJ. JMJD3 activated hyaluronan synthesis drives muscle regeneration in an inflammatory environment. Science 2022; 377:666-669. [PMID: 35926054 DOI: 10.1126/science.abm9735] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Muscle stem cells (MuSCs) reside in a specialized niche that ensures their regenerative capacity. Although we know that innate immune cells infiltrate the niche in response to injury, it remains unclear how MuSCs adapt to this altered environment for initiating repair. Here, we demonstrate that inflammatory cytokine signaling from the regenerative niche impairs the ability of quiescent MuSCs to reenter the cell cycle. The histone H3 lysine 27 (H3K27) demethylase JMJD3, but not UTX, allowed MuSCs to overcome inhibitory inflammation signaling by removing trimethylated H3K27 (H3K27me3) marks at the Has2 locus to initiate production of hyaluronic acid, which in turn established an extracellular matrix competent for integrating signals that direct MuSCs to exit quiescence. Thus, JMJD3-driven hyaluronic acid synthesis plays a proregenerative role that allows MuSC adaptation to inflammation and the initiation of muscle repair.
Collapse
Affiliation(s)
- Kiran Nakka
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Sarah Hachmer
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Zeinab Mokhtari
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Radmila Kovac
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Hina Bandukwala
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Clara Bernard
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS 5261, INSERM U1315, Université de Lyon, Lyon, France
| | - Yuefeng Li
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Guojia Xie
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Chengyu Liu
- Transgenic Core, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Magid Fallahi
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Lynn A Megeney
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Julien Gondin
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS 5261, INSERM U1315, Université de Lyon, Lyon, France
| | - Bénédicte Chazaud
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS 5261, INSERM U1315, Université de Lyon, Lyon, France
| | - Marjorie Brand
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.,LIFE Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Xiaohui Zha
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Kai Ge
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - F Jeffrey Dilworth
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.,LIFE Research Institute, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
44
|
Lee JS, Ghang B, Choi W, Hong S, Kim YG, Lee CK, Nam SJ, Yoo B. Expression of Inflammatory Markers in the Muscles of Patients with Idiopathic Inflammatory Myopathy According to the Presence of Interstitial Lung Disease. J Clin Med 2022; 11:3021. [PMID: 35683408 PMCID: PMC9181768 DOI: 10.3390/jcm11113021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/06/2022] [Accepted: 05/25/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Several studies have identified factors associated with the development of interstitial lung disease (ILD) in patients with idiopathic inflammatory myopathies (IIMs). However, few have assessed the association between ILD and muscle biopsy findings, including inflammatory marker expressions analyzed using immunohistochemistry (IHC). METHODS Muscle biopsies from patients who were newly diagnosed with IIMs between 2000 and 2017 were reviewed. ILD was diagnosed based on chest computed tomography findings at the time of diagnosis of IIMs. IHC staining was performed for CD3, CD4, CD8, CD20, CD68, CD163, MX1, MHC class I, and HLA-DR. The factors associated with the presence of ILD were evaluated by logistic regression analysis. RESULTS Of the 129 patients with IIM, 49 (38%) had ILD. In the muscle biopsy findings, CD4 expression, MX1 expression on immune cells, and expression of MHC class I and HLA-DR on myofibers were more common in patients with ILD than those without. In the logistic regression analysis, the HLA-DR expression on myofibers was significantly associated with the risk of ILD (OR, 2.39; 95% CI, 1.24-4.90, p = 0.012) after adjusting for pathologic findings, clinical features, and autoantibodies. CONCLUSION The expression of HLA-DR on myofibers was associated with the presence of ILD in patients with IIM.
Collapse
Affiliation(s)
- Jung Sun Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul Veterans Hospital, Seoul 05368, Korea;
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (W.C.); (S.H.); (Y.-G.K.); (C.-K.L.)
| | - Byeongzu Ghang
- Division of Rheumatology, Department of Internal Medicine, Jeju National University School of Medicine, Jeju National University Hospital, Jeju 63241, Korea;
| | - Wonho Choi
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (W.C.); (S.H.); (Y.-G.K.); (C.-K.L.)
| | - Seokchan Hong
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (W.C.); (S.H.); (Y.-G.K.); (C.-K.L.)
| | - Yong-Gil Kim
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (W.C.); (S.H.); (Y.-G.K.); (C.-K.L.)
| | - Chang-Keun Lee
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (W.C.); (S.H.); (Y.-G.K.); (C.-K.L.)
| | - Soo Jeong Nam
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Bin Yoo
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (W.C.); (S.H.); (Y.-G.K.); (C.-K.L.)
| |
Collapse
|
45
|
Myxomavirus Serp-1 Protein Ameliorates Inflammation in a Mouse Model of Duchenne Muscular Dystrophy. Biomedicines 2022; 10:biomedicines10051154. [PMID: 35625891 PMCID: PMC9138346 DOI: 10.3390/biomedicines10051154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 01/27/2023] Open
Abstract
Duchenne muscular dystrophy is an X-linked disease afflicting 1 in 3500 males that is characterized by muscle weakness and wasting during early childhood, and loss of ambulation and death by early adulthood. Chronic inflammation due to myofiber instability leads to fibrosis, which is a primary cause of loss of ambulation and cardiorespiratory insufficiency. Current standard of care focuses on reducing inflammation with corticosteroids, which have serious adverse effects. It is imperative to identify alternate immunosuppressants as treatments to reduce fibrosis and mortality. Serp-1, a Myxoma virus-derived 55 kDa secreted glycoprotein, has proven efficacy in a range of animal models of acute inflammation, and its safety and efficacy has been shown in a clinical trial. In this initial study, we examined whether pegylated Serp-1 (PEGSerp-1) treatment would ameliorate chronic inflammation in a mouse model for Duchenne muscular dystrophy. Our data revealed a significant reduction in diaphragm fibrosis and increased myofiber diameter, and significantly decreased pro-inflammatory M1 macrophage infiltration. The M2a macrophage and overall T cell populations showed no change. These data demonstrate that treatment with this new class of poxvirus-derived immune-modulating serpin has potential as a therapeutic approach designed to ameliorate DMD pathology and facilitate muscle regeneration.
Collapse
|
46
|
Mann AO, Hanna BS, Muñoz-Rojas AR, Sandrock I, Prinz I, Benoist C, Mathis D. IL-17A-producing γδT cells promote muscle regeneration in a microbiota-dependent manner. J Exp Med 2022; 219:213111. [PMID: 35380608 PMCID: PMC8989184 DOI: 10.1084/jem.20211504] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 02/23/2022] [Accepted: 03/16/2022] [Indexed: 01/07/2023] Open
Abstract
Subsequent to acute injury, skeletal muscle undergoes a stereotypic regenerative process that reestablishes homeostasis. Various types of innate and adaptive immunocytes exert positive or negative influences at specific stages along the course of muscle regeneration. We describe an unanticipated role for γδT cells in promoting healthy tissue recovery after injection of cardiotoxin into murine hindlimb muscle. Within a few days of injury, IL-17A-producing γδT cells displaying primarily Vγ6+ antigen receptors accumulated at the wound site. Punctual ablation experiments showed that these cells boosted early inflammatory events, notably recruitment of neutrophils; fostered the proliferation of muscle stem and progenitor cells; and thereby promoted tissue regeneration. Supplementation of mice harboring low numbers of IL-17A+ γδT cells with recombinant IL-17A largely reversed their inflammatory and reparative defects. Unexpectedly, the accumulation and influences of γδT cells in this experimental context were microbiota dependent, unveiling an orthogonal perspective on the treatment of skeletal muscle pathologies such as catastrophic wounds, wasting, muscular dystrophies, and myositides.
Collapse
Affiliation(s)
- Alexander O. Mann
- Department of Immunology, Harvard Medical School, Boston, MA,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA
| | - Bola S. Hanna
- Department of Immunology, Harvard Medical School, Boston, MA,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA
| | - Andrés R. Muñoz-Rojas
- Department of Immunology, Harvard Medical School, Boston, MA,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA
| | - Inga Sandrock
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany,Institute of Systems Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christophe Benoist
- Department of Immunology, Harvard Medical School, Boston, MA,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA,Correspondence to Diane Mathis:
| |
Collapse
|
47
|
Single-cell transcriptome analysis reveals three sequential phases of gene expression during zebrafish sensory hair cell regeneration. Dev Cell 2022; 57:799-819.e6. [PMID: 35316618 PMCID: PMC9188816 DOI: 10.1016/j.devcel.2022.03.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/19/2021] [Accepted: 02/28/2022] [Indexed: 12/20/2022]
Abstract
Loss of sensory hair cells (HCs) in the mammalian inner ear leads to permanent hearing and vestibular defects, whereas loss of HCs in zebrafish results in their regeneration. We used single-cell RNA sequencing (scRNA-seq) to characterize the transcriptional dynamics of HC regeneration in zebrafish at unprecedented spatiotemporal resolution. We uncovered three sequentially activated modules: first, an injury/inflammatory response and downregulation of progenitor cell maintenance genes within minutes after HC loss; second, the transient activation of regeneration-specific genes; and third, a robust re-activation of developmental gene programs, including HC specification, cell-cycle activation, ribosome biogenesis, and a metabolic switch to oxidative phosphorylation. The results are relevant not only for our understanding of HC regeneration and how we might be able to trigger it in mammals but also for regenerative processes in general. The data are searchable and publicly accessible via a web-based interface.
Collapse
|
48
|
Samandari M, Quint J, Rodríguez-delaRosa A, Sinha I, Pourquié O, Tamayol A. Bioinks and Bioprinting Strategies for Skeletal Muscle Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2105883. [PMID: 34773667 PMCID: PMC8957559 DOI: 10.1002/adma.202105883] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/28/2021] [Indexed: 05/16/2023]
Abstract
Skeletal muscles play important roles in critical body functions and their injury or disease can lead to limitation of mobility and loss of independence. Current treatments result in variable functional recovery, while reconstructive surgery, as the gold-standard approach, is limited due to donor shortage, donor-site morbidity, and limited functional recovery. Skeletal muscle tissue engineering (SMTE) has generated enthusiasm as an alternative solution for treatment of injured tissue and serves as a functional disease model. Recently, bioprinting has emerged as a promising tool for recapitulating the complex and highly organized architecture of skeletal muscles at clinically relevant sizes. Here, skeletal muscle physiology, muscle regeneration following injury, and current treatments following muscle loss are discussed, and then bioprinting strategies implemented for SMTE are critically reviewed. Subsequently, recent advancements that have led to improvement of bioprinting strategies to construct large muscle structures, boost myogenesis in vitro and in vivo, and enhance tissue integration are discussed. Bioinks for muscle bioprinting, as an essential part of any bioprinting strategy, are discussed, and their benefits, limitations, and areas to be improved are highlighted. Finally, the directions the field should expand to make bioprinting strategies more translational and overcome the clinical unmet needs are discussed.
Collapse
Affiliation(s)
- Mohamadmahdi Samandari
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Jacob Quint
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | - Indranil Sinha
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, USA
| | - Olivier Pourquié
- Department of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
| |
Collapse
|
49
|
Sun Y, Liang C, Zheng L, Liu L, Li Z, Yang G, Li Y. Anti-fatigue effect of hypericin in a chronic forced exercise mouse model. JOURNAL OF ETHNOPHARMACOLOGY 2022; 284:114767. [PMID: 34710555 DOI: 10.1016/j.jep.2021.114767] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hypericum perforatum L. is a traditional Chinese medicine used to sooth the liver, relieve depression, reduce body temperature, reduce sweating, and stimulate lactation. HP was extracted from Hypericum perforatum L. AIM OF STUDY The antifatigue effects of hypericin were assessed in a series of experiments. MATERIALS AND METHODS Six-to eight-week-old male ICR mice were raised in our lab. Mice were subjected to swimming training for 2 h, 6 days/week for 6 weeks. One hour prior to each swimming session, intraperitoneal injection of saline or HP (2 or 4 mg/kg) was performed. RESULTS Compared with the fatigue model control group, HP was found to significantly increase the swimming time in forced swimming tests. The molecular mechanisms underlying the antifatigue effects were further revealed by analysing energy metabolism, the oxidant-antioxidant system and the inflammatory response. HP normalized changes in BLA, LDH, BUN, and CK, LG in the liver. In addition, multiple assays have confirmed that HP improved the MDA, T-AOC, GSH-PX and SOD activity, and the relevant signalling pathways involved in the antifatigue effects were clarified. Furthermore, HP improves the expression of pro- and anti-inflammatory cytokines in skeletal muscle. CONCLUSION These results suggested that the anti-chronic fatigue effects of HP are likely achieved by normalizing energy metabolism and attenuating oxidative and inflammatory responses. Consequently, this study supports HP use in the clinic to alleviate chronic fatigue.
Collapse
Affiliation(s)
- Yang Sun
- School of Physical Education, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Chen Liang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Lihua Zheng
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Lei Liu
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, Jilin, 130024, China.
| | - Zhijin Li
- Xiamen Health and Medical Big Data Center (Xiamen Medicine Research Institute), Xiamen Key Laboratory of Natural Medicine Research and Development, Xiamen, Fujian, 361008, China.
| | - Guang Yang
- School of Physical Education, Northeast Normal University, Changchun, Jilin, 130024, China.
| | - Yuxin Li
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, Jilin, 130024, China
| |
Collapse
|
50
|
Staunton CA, Owen ED, Hemmings K, Vasilaki A, McArdle A, Barrett-Jolley R, Jackson MJ. Skeletal muscle transcriptomics identifies common pathways in nerve crush injury and ageing. Skelet Muscle 2022; 12:3. [PMID: 35093178 PMCID: PMC8800362 DOI: 10.1186/s13395-021-00283-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 11/24/2021] [Indexed: 12/16/2022] Open
Abstract
Motor unit remodelling involving repeated denervation and re-innervation occurs throughout life. The efficiency of this process declines with age contributing to neuromuscular deficits. This study investigated differentially expressed genes (DEG) in muscle following peroneal nerve crush to model motor unit remodelling in C57BL/6 J mice. Muscle RNA was isolated at 3 days post-crush, RNA libraries were generated using poly-A selection, sequenced and analysed using gene ontology and pathway tools. Three hundred thirty-four DEG were found in quiescent muscle from (26mnth) old compared with (4-6mnth) adult mice and these same DEG were present in muscle from adult mice following nerve crush. Peroneal crush induced 7133 DEG in muscles of adult and 699 DEG in muscles from old mice, although only one DEG (ZCCHC17) was found when directly comparing nerve-crushed muscles from old and adult mice. This analysis revealed key differences in muscle responses which may underlie the diminished ability of old mice to repair following nerve injury.
Collapse
Affiliation(s)
- C A Staunton
- MRC- Versus Arthritis Research Centre for Integrated research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| | - E D Owen
- MRC- Versus Arthritis Research Centre for Integrated research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| | - K Hemmings
- MRC- Versus Arthritis Research Centre for Integrated research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| | - A Vasilaki
- MRC- Versus Arthritis Research Centre for Integrated research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| | - A McArdle
- MRC- Versus Arthritis Research Centre for Integrated research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| | - R Barrett-Jolley
- MRC- Versus Arthritis Research Centre for Integrated research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| | - M J Jackson
- MRC- Versus Arthritis Research Centre for Integrated research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK.
| |
Collapse
|