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
|
Cheng Y, Lin S, Cao Z, Yu R, Fan Y, Chen J. The role of chronic low-grade inflammation in the development of sarcopenia: Advances in molecular mechanisms. Int Immunopharmacol 2025; 147:114056. [PMID: 39799736 DOI: 10.1016/j.intimp.2025.114056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 12/16/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
With the exacerbation of global population aging, sarcopenia has become an increasingly recognized public health issue. Sarcopenia, characterized by a progressive decline in skeletal muscle mass, strength, and function, significantly impacts the quality of life in the elderly. Herein, we explore the role of chroniclow-gradeinflammation in the development of sarcopenia and its underlying molecular mechanisms, including chronic inflammation-associated signaling pathways, immunosenescence, obesity and lipid infiltration, gut microbiota dysbiosis and intestinal barrier disruption, and the decline of satellite cells. The interplay and interaction of these molecular mechanisms provide new perspectives on the complexity of the pathogenesis of sarcopenia and offer a theoretical foundation for the development of future therapeutic strategies.
Collapse
Affiliation(s)
- Ying Cheng
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040 China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040 China
| | - Shangjin Lin
- Department of Orthopedics, Huadong Hospital Affiliated to Fudan University, Shanghai 200040 China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040 China
| | - Ziyi Cao
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040 China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040 China
| | - Runzhi Yu
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040 China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040 China
| | - Yongqian Fan
- Department of Orthopedics, Huadong Hospital Affiliated to Fudan University, Shanghai 200040 China.
| | - Jie Chen
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040 China.
| |
Collapse
|
3
|
Lokwani R, Fertil D, Hartigan DR, Josyula A, Ngo TB, Sadtler K. Eosinophils Respond to Extracellular Matrix Treated Muscle Injuries but are Not Required for Macrophage Polarization. Adv Healthc Mater 2025; 14:e2400134. [PMID: 39072935 PMCID: PMC11834370 DOI: 10.1002/adhm.202400134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 06/10/2024] [Indexed: 07/30/2024]
Abstract
The immune response to decellularized extracellular matrix (ECM) muscle injury is characterized by Th2 T cells, Tregs, M2-like macrophages, and an abundance of eosinophils. Eosinophils have previously been described as mediators of muscle regeneration but inhibit skin wound healing. In addition to response to wounding, a large number of eosinophils respond to biomaterial-treated muscle injury, specifically in response to decellularized ECM. ECM treatment of muscle wounds has been associated with positive outcomes in tissue regeneration, but the detailed mechanisms of action are still being evaluated. Here, this work investigates the role of these eosinophils in terms of their immunologic phenotype and subsequent effect on the local tissue microenvironment. These cells have a mixed phenotype showing both type-2 and regulatory gene upregulation and but are not required for macrophage polarization. Beyond the local tissue, ECM treatment is seen to induce a transient flux of eosinophils to the lungs but prevented a trauma-associated neutrophilia in the lungs of injured mice. This work believes this local and systemic immunomodulation contributes to the regenerative effects of the material and such distal tissue effects should be considered in therapeutic design and implementation.
Collapse
Affiliation(s)
- Ravi Lokwani
- Section on ImmunoengineeringCenter for Biomedical Engineering and Technology AccelerationNational Institute of Biomedical Imaging and BioengineeringNational Institutes of HealthBethesdaMD20892USA
| | - Daphna Fertil
- Section on ImmunoengineeringCenter for Biomedical Engineering and Technology AccelerationNational Institute of Biomedical Imaging and BioengineeringNational Institutes of HealthBethesdaMD20892USA
| | - Devon R. Hartigan
- Section on ImmunoengineeringCenter for Biomedical Engineering and Technology AccelerationNational Institute of Biomedical Imaging and BioengineeringNational Institutes of HealthBethesdaMD20892USA
| | - Aditya Josyula
- Section on ImmunoengineeringCenter for Biomedical Engineering and Technology AccelerationNational Institute of Biomedical Imaging and BioengineeringNational Institutes of HealthBethesdaMD20892USA
| | - Tran B. Ngo
- Section on ImmunoengineeringCenter for Biomedical Engineering and Technology AccelerationNational Institute of Biomedical Imaging and BioengineeringNational Institutes of HealthBethesdaMD20892USA
| | - Kaitlyn Sadtler
- Section on ImmunoengineeringCenter for Biomedical Engineering and Technology AccelerationNational Institute of Biomedical Imaging and BioengineeringNational Institutes of HealthBethesdaMD20892USA
| |
Collapse
|
4
|
Ahn J, Kim B, Bello AB, Moon JJ, Arai Y, Lee SH. Regenerative Functions of Regulatory T Cells and Current Strategies Utilizing Mesenchymal Stem Cells in Immunomodulatory Tissue Regeneration. Tissue Eng Regen Med 2025; 22:167-180. [PMID: 39804546 PMCID: PMC11794763 DOI: 10.1007/s13770-024-00690-w] [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: 11/21/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Regulatory T cells (Tregs) are essential for maintaining immune homeostasis and facilitating tissue regeneration by fostering an environment conducive to tissue repair. However, in damaged tissues, excessive inflammatory responses can overwhelm the immunomodulatory capacity of Tregs, compromising their functionality and potentially hindering effective regeneration. Mesenchymal stem cells (MSCs) play a key role in enhancing Treg function. MSCs enhance Treg activity through indirect interactions, such as cytokine secretion, and direct interactions via membrane proteins. METHODS This review examines the regenerative functions of Tregs across various tissues, including bone, cartilage, muscle, and skin, and explores strategies to enhance Treg functionality using MSCs. Advanced techniques, such as the overexpression of relevant genes in MSCs, are highlighted for their potential to further enhance Treg function. Additionally, emerging technologies utilizing extracellular vesicles (EVs) and cell membrane-derived vesicles derived from MSCs offer promising alternatives to circumvent the potential side effects associated with live cell therapies. This review proposes approaches to enhance Treg function and promote tissue regeneration and also outlines future research directions. RESULTS AND CONCLUSION This review elucidates recent technological advancements aimed at enhancing Treg function using MSCs and examines their potential to improve tissue regeneration efficiency.
Collapse
Grants
- 2022R1A2C3004850 Ministry of Science and ICT, South Korea
- RS-2024-00405381 Ministry of Science and ICT, South Korea
- RS-2023-00257290 Ministry of Science and ICT, South Korea
- RS-2023-00246418 Ministry of Education
- RS-2023-00275407 Ministry of Education
- 21C0703L1 Ministry of Science and ICT, Ministry of Health & Welfare
- HX23C1734 Ministry of Science and ICT, Ministry of Trade, Industry and Energy, Ministry of Health & Welfare, The Ministry of Food and Drug Safety
- Ministry of Science and ICT, Ministry of Health & Welfare
- Ministry of Science and ICT, Ministry of Trade, Industry and Energy, Ministry of Health & Welfare, The Ministry of Food and Drug Safety
Collapse
Affiliation(s)
- Jinsung Ahn
- Department of Biomedical Engineering, Dongguk University, Seoul, South Korea
| | - Bowon Kim
- Department of Biomedical Engineering, Dongguk University, Seoul, South Korea
| | - Alvin Bacero Bello
- Department of Biomedical Engineering, Dongguk University, Seoul, South Korea
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Yoshie Arai
- Department of Biomedical Engineering, Dongguk University, Seoul, South Korea.
| | - Soo-Hong Lee
- Department of Biomedical Engineering, Dongguk University, Seoul, South Korea.
| |
Collapse
|
5
|
Chang TH, Ho PC. Interferon-driven Metabolic Reprogramming and Tumor Microenvironment Remodeling. Immune Netw 2025; 25:e8. [PMID: 40078784 PMCID: PMC11896656 DOI: 10.4110/in.2025.25.e8] [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: 12/24/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 03/14/2025] Open
Abstract
IFNs play a critical role in cancer biology, including impacting tumor cell behavior and instructing the tumor microenvironment (TME). IFNs recently have been shown to reprogram tumor metabolism through distinct mechanisms. Furthermore, IFNs shape the TME by modulating immune cell infiltration and function, contributing to the intricate interaction between the tumor and stromal cells. This review summarizes the effects of IFNs on metabolic reprogramming and their impacts on the function of immune cells within the TME, with a particular focus on the dual roles of IFNs in mediating both anti-tumor and pro-tumor immune responses. Understanding the significance of IFNs-mediated processes aids to advise future therapeutic strategies in cancer treatment.
Collapse
Affiliation(s)
- Tzu-Hsuan Chang
- Department of Fundamental Oncology, University of Lausanne, 1015 Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, 1015 Lausanne, Switzerland
| | - Ping-Chih Ho
- Department of Fundamental Oncology, University of Lausanne, 1015 Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, 1015 Lausanne, Switzerland
| |
Collapse
|
6
|
Wu W, Hou C, Wu W, Shen H, Zeng Y, Chen L, Liao Y, Zhu H, Tian Y, Peng B, Chen WX, Li X. Cerebrospinal fluid neurofilament light chain levels in children with acquired demyelinating syndrome. Front Pediatr 2024; 12:1467020. [PMID: 39564383 PMCID: PMC11573574 DOI: 10.3389/fped.2024.1467020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024] Open
Abstract
Objective To study the cerebrospinal fluid (CSF) neurofilament light chain (NfL) in pediatric acquired demyelinating syndrome (ADS) and its association with factors of laboratory and imaging results. Methods We analyzed clinical data from children with ADS collected from May 2020 to January 2021 at the Department of Neurology of Guangzhou Women and Children's Medical Center. Enzyme-linked immunosorbent assays were used to detect the CSF NfL of patients. Results Thirty pediatric ADS patients (17 male, 13 female) were included in the study. The most frequent diagnosis was uncategorized ADS (36.7%, 11/30), followed by acute disseminating encephalomyelitis (ADEM) (23.3%, 7/30), myelin oligodendrocyte glycoprotein antibody-associated disorder (MOGAD) (20.0%, 6/30), NMO (6.7%, 2/30), multiple sclerosis (MS) (6.7%, 2/30), and neuromyelitis optic spectrum disorders (NMOSD) (6.7%, 2/30). The median CSF NfL for the first time was 7,425.28 pg/ml (interquartile range, 1,273.51, >10,000 pg/ml). CSF NfL increase over normal value (<290.00 pg/ml for people younger than 30 years old) was seen in 98.7% of patients. Patients were divided into uncategorized ADS, ADEM, MOGAD, and MS/NMO/NMOSD groups, with no significant difference in CSF NfL between each group. The CSF NfL positively correlated with the immunoglobulin (Ig) G (ρ = 0.473) and IgE (ρ = 0.366). However, the CSF NfL did not correlate with CSF white blood count and CSF protein. Furthermore, there was no significant difference between patients with oligoclonal bands positive and without. The CSF NfL negatively correlated with interferon γ (ρ = -0.501), CD45 + CD3+ T (ρ = -0.466), CD45 + CD3 + CD4+ T (ρ = -0.466), and CD45 + CD3 + CD8+ T cells (ρ = -0.521). However, it did not correlate with CD45 + CD19+ B cells. CSF NfL in patients with cerebral white matter lesions in MRI was higher than in patients without. Moreover, the CSF NfL positively correlated with the number of brain MRI locations (ρ = 0.362). Nine patients underwent multiple detections of CSF NfL, and their CSF NfL for the last detection was not significantly different from the first. Conclusions The CSF NfL increases significantly in pediatric ADS, and it can be a biomarker of neuro-axonal injury and a good indication of the extent of lesions.
Collapse
Affiliation(s)
- Wenlin Wu
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chi Hou
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wenxiao Wu
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Huiling Shen
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yiru Zeng
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lianfeng Chen
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yinting Liao
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Haixia Zhu
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yang Tian
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Bingwei Peng
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wen-Xiong Chen
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaojing Li
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
7
|
Lee JH, Shin SJ, Lee JH, Knowles JC, Lee HH, Kim HW. Adaptive immunity of materials: Implications for tissue healing and regeneration. Bioact Mater 2024; 41:499-522. [PMID: 39206299 PMCID: PMC11350271 DOI: 10.1016/j.bioactmat.2024.07.027] [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: 05/07/2024] [Revised: 07/16/2024] [Accepted: 07/21/2024] [Indexed: 09/04/2024] Open
Abstract
Recent cumulative findings signify the adaptive immunity of materials as a key agenda in tissue healing that can improve regenerative events and outcomes. Modulating immune responses, mainly the recruitment and functions of T and B cells and their further interplay with innate immune cells (e.g., dendritic cells, macrophages) can be orchestrated by materials. For instance, decellularized matrices have been shown to promote muscle healing by inducing T helper 2 (Th2) cell immunity, while synthetic biopolymers exhibit differential effects on B cell responses and fibrosis compared decellularized matrices. We discuss the recent findings on how implantable materials instruct the adaptive immune events and the subsequent tissue healing process. In particular, we dissect the materials' physicochemical properties (shape, size, topology, degradation, rigidity, and matrix dynamic mechanics) to demonstrate the relations of these parameters with the adaptive immune responses in vitro and the underlying biological mechanisms. Furthermore, we present evidence of recent in vivo phenomena, including tissue healing, cancer progression, and fibrosis, wherein biomaterials potentially shape adaptive immune cell functions and in vivo outcomes. Our discussion will help understand the materials-regulated immunology events more deeply, and offer the design rationale of materials with tunable matrix properties for accelerated tissue repair and regeneration.
Collapse
Affiliation(s)
- Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
| | - Seong-Jin Shin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
| | - Jun Hee Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
| | - Jonathan C. Knowles
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman Dental Institute, University College London, London NW3 2PX, United Kingdom
| | - Hae-Hyoung Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
8
|
Nayer B, Tan JL, Alshoubaki YK, Lu YZ, Legrand JMD, Lau S, Hu N, Park AJ, Wang XN, Amann-Zalcenstein D, Hickey PF, Wilson T, Kuhn GA, Müller R, Vasanthakumar A, Akira S, Martino MM. Local administration of regulatory T cells promotes tissue healing. Nat Commun 2024; 15:7863. [PMID: 39251592 PMCID: PMC11383969 DOI: 10.1038/s41467-024-51353-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 08/05/2024] [Indexed: 09/11/2024] Open
Abstract
Regulatory T cells (Tregs) are crucial immune cells for tissue repair and regeneration. However, their potential as a cell-based regenerative therapy is not yet fully understood. Here, we show that local delivery of exogenous Tregs into injured mouse bone, muscle, and skin greatly enhances tissue healing. Mechanistically, exogenous Tregs rapidly adopt an injury-specific phenotype in response to the damaged tissue microenvironment, upregulating genes involved in immunomodulation and tissue healing. We demonstrate that exogenous Tregs exert their regenerative effect by directly and indirectly modulating monocytes/macrophages (Mo/MΦ) in injured tissues, promoting their switch to an anti-inflammatory and pro-healing state via factors such as interleukin (IL)-10. Validating the key role of IL-10 in exogenous Treg-mediated repair and regeneration, the pro-healing capacity of these cells is lost when Il10 is knocked out. Additionally, exogenous Tregs reduce neutrophil and cytotoxic T cell accumulation and IFN-γ production in damaged tissues, further dampening the pro-inflammatory Mo/MΦ phenotype. Highlighting the potential of this approach, we demonstrate that allogeneic and human Tregs also promote tissue healing. Together, this study establishes exogenous Tregs as a possible universal cell-based therapy for regenerative medicine and provides key mechanistic insights that could be harnessed to develop immune cell-based therapies to enhance tissue healing.
Collapse
Affiliation(s)
- Bhavana Nayer
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Jean L Tan
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Yasmin K Alshoubaki
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Yen-Zhen Lu
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Julien M D Legrand
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Sinnee Lau
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Nan Hu
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Anthony J Park
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Xiao-Nong Wang
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Daniela Amann-Zalcenstein
- Advanced Genomics Facility, Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Peter F Hickey
- Advanced Genomics Facility, Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Trevor Wilson
- MHTP Medical Genomics Facility, Monash Health Translation Precinct, Clayton, VIC, Australia
| | - Gisela A Kuhn
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Ajithkumar Vasanthakumar
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- La Trobe University, Bundoora, VIC, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, Australia
| | - Shizuo Akira
- Laboratory of Host Defense, World Premier Institute Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Mikaël M Martino
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia.
- Laboratory of Host Defense, World Premier Institute Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Victorian Heart Institute, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
9
|
Zhang C, Li G, Zhang F, Zhang Y, Hong S, Gao S, Liu Y, Du J, Li Y. IL-33 Facilitates Fibro-Adipogenic Progenitors to Establish the Pro-Regenerative Niche after Muscle Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405299. [PMID: 39037903 PMCID: PMC11425282 DOI: 10.1002/advs.202405299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/03/2024] [Indexed: 07/24/2024]
Abstract
During the process of muscle regeneration post-injury in adults, muscle stem cells (MuSCs) function is facilitated by neighboring cells within the pro-regenerative niche. However, the precise mechanism triggering the initiation of signaling in the pro-regenerative niche remains unknown. Using single-cell RNA sequencing, 14 different muscle cells are comprehensively mapped during the initial stage following injury. Among these, macrophages and fibro-adipogenic progenitor cells (FAPs) exhibit the most pronounced intercellular communication with other cells. In the FAP subclusters, the study identifies an activated FAP phenotype that secretes chemokines, such as CXCL1, CXCL5, CCL2, and CCL7, to recruit macrophages after injury. Il1rl1, encoding the protein of the interleukin-33 (IL-33) receptor, is identified as a highly expressed signature surface marker of the FAP phenotype. Following muscle injury, autocrine IL-33, an alarmin, has been observed to activate quiescent FAPs toward this inflammatory phenotype through the IL1RL1-MAPK/NF-κB signaling pathway. Il1rl1 deficiency results in decreased chemokine expression and recruitment of macrophages, accompanied by impaired muscle regeneration. These findings elucidate a novel mechanism involving the IL-33/IL1RL1 signaling pathway in promoting the activation of FAPs and facilitating muscle regeneration, which can aid the development of therapeutic strategies for muscle-related disorders and injuries.
Collapse
Affiliation(s)
- Congcong Zhang
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
- Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, 100029, China
| | - Guoqi Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
- Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, 100029, China
| | - Fan Zhang
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
- Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, 100029, China
| | - Yanhong Zhang
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
- Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, 100029, China
| | - Shiyao Hong
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
- Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, 100029, China
| | - Shijuan Gao
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
- Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, 100029, China
| | - Yan Liu
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
- Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, 100029, China
| | - Jie Du
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
- Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, 100029, China
| | - Yulin Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
- Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, 100029, China
| |
Collapse
|
10
|
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
|
11
|
Zhu Y, Hu Y, Pan Y, Li M, Niu Y, Zhang T, Sun H, Zhou S, Liu M, Zhang Y, Wu C, Ma Y, Guo Y, Wang L. Fatty infiltration in the musculoskeletal system: pathological mechanisms and clinical implications. Front Endocrinol (Lausanne) 2024; 15:1406046. [PMID: 39006365 PMCID: PMC11241459 DOI: 10.3389/fendo.2024.1406046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/10/2024] [Indexed: 07/16/2024] Open
Abstract
Fatty infiltration denotes the anomalous accrual of adipocytes in non-adipose tissue, thereby generating toxic substances with the capacity to impede the ordinary physiological functions of various organs. With aging, the musculoskeletal system undergoes pronounced degenerative alterations, prompting heightened scrutiny regarding the contributory role of fatty infiltration in its pathophysiology. Several studies have demonstrated that fatty infiltration affects the normal metabolism of the musculoskeletal system, leading to substantial tissue damage. Nevertheless, a definitive and universally accepted generalization concerning the comprehensive effects of fatty infiltration on the musculoskeletal system remains elusive. As a result, this review summarizes the characteristics of different types of adipose tissue, the pathological mechanisms associated with fatty infiltration in bone, muscle, and the entirety of the musculoskeletal system, examines relevant clinical diseases, and explores potential therapeutic modalities. This review is intended to give researchers a better understanding of fatty infiltration and to contribute new ideas to the prevention and treatment of clinical musculoskeletal diseases.
Collapse
Affiliation(s)
- Yihua Zhu
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yue Hu
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yalan Pan
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Traditional Chinese Medicine (TCM) Nursing Intervention Laboratory of Chronic Disease Key Laboratory, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Muzhe Li
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yuanyuan Niu
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Tianchi Zhang
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Haitao Sun
- Department of Orthopedic Surgery, Affiliated Huishan Hospital of Xinglin College of Nantong University, Wuxi, Jiangsu, China
| | - Shijie Zhou
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Mengmin Liu
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yili Zhang
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Chengjie Wu
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yong Ma
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng TCM Hospital, Yancheng, Jiangsu, China
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, China
| | - Yang Guo
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, China
| | - Lining Wang
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Chinese Medicine Centre (International Collaboration between Western Sydney University and Beijing University of Chinese Medicine), Western Sydney University, Sydney, Australia
| |
Collapse
|
12
|
Liu J, Liu F, Liang T, Zhou Y, Su X, Li X, Zeng J, Qu P, Wang Y, Chen F, Lei Q, Li G, Cheng P. The roles of Th cells in myocardial infarction. Cell Death Discov 2024; 10:287. [PMID: 38879568 PMCID: PMC11180143 DOI: 10.1038/s41420-024-02064-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/19/2024] Open
Abstract
Myocardial infarction, commonly known as a heart attack, is a serious condition caused by the abrupt stoppage of blood flow to a part of the heart, leading to tissue damage. A significant aspect of this condition is reperfusion injury, which occurs when blood flow is restored but exacerbates the damage. This review first addresses the role of the innate immune system, including neutrophils and macrophages, in the cascade of events leading to myocardial infarction and reperfusion injury. It then shifts focus to the critical involvement of CD4+ T helper cells in these processes. These cells, pivotal in regulating the immune response and tissue recovery, include various subpopulations such as Th1, Th2, Th9, Th17, and Th22, each playing a unique role in the pathophysiology of myocardial infarction and reperfusion injury. These subpopulations contribute to the injury process through diverse mechanisms, with cytokines such as IFN-γ and IL-4 influencing the balance between tissue repair and injury exacerbation. Understanding the interplay between the innate immune system and CD4+ T helper cells, along with their cytokines, is crucial for developing targeted therapies to mitigate myocardial infarction and reperfusion injury, ultimately improving outcomes for cardiac patients.
Collapse
Affiliation(s)
- Jun Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Feila Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Tingting Liang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Yue Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Xiaohan Su
- Department of Breast and Thyroid Surgery, Biological Targeting Laboratory of Breast Cancer, Academician (expert) workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xue Li
- Department of Laboratory Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jiao Zeng
- Department of Breast and Thyroid Surgery, Biological Targeting Laboratory of Breast Cancer, Academician (expert) workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Peng Qu
- Department of Laboratory Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yali Wang
- Department of Breast and Thyroid Surgery, Biological Targeting Laboratory of Breast Cancer, Academician (expert) workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Fuli Chen
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qian Lei
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Gang Li
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Panke Cheng
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Chengdu, China.
| |
Collapse
|
13
|
Langston PK, Mathis D. Immunological regulation of skeletal muscle adaptation to exercise. Cell Metab 2024; 36:1175-1183. [PMID: 38670108 DOI: 10.1016/j.cmet.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024]
Abstract
Exercise has long been acknowledged for its powerful disease-preventing, health-promoting effects. However, the cellular and molecular mechanisms responsible for the beneficial effects of exercise are not fully understood. Inflammation is a component of the stress response to exercise. Recent work has revealed that such inflammation is not merely a symptom of exertion; rather, it is a key regulator of exercise adaptations, particularly in skeletal muscle. The purpose of this piece is to provide a conceptual framework that we hope will integrate exercise immunology with exercise physiology, muscle biology, and cellular immunology. We start with an overview of early studies in the field of exercise immunology, followed by an exploration of the importance of stromal cells and immunocytes in the maintenance of muscle homeostasis based on studies of experimental muscle injury. Subsequently, we discuss recent advances in our understanding of the functions and physiological relevance of the immune system in exercised muscle. Finally, we highlight a potential immunological basis for the benefits of exercise in musculoskeletal diseases and aging.
Collapse
Affiliation(s)
- P Kent Langston
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
14
|
Loffredo LF, Savage TM, Ringham OR, Arpaia N. Treg-tissue cell interactions in repair and regeneration. J Exp Med 2024; 221:e20231244. [PMID: 38668758 PMCID: PMC11046849 DOI: 10.1084/jem.20231244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/22/2024] [Accepted: 04/11/2024] [Indexed: 04/29/2024] Open
Abstract
Regulatory T (Treg) cells are classically known for their critical immunosuppressive functions that support peripheral tolerance. More recent work has demonstrated that Treg cells produce pro-repair mediators independent of their immunosuppressive function, a process that is critical to repair and regeneration in response to numerous tissue insults. These factors act on resident parenchymal and structural cells to initiate repair in a tissue-specific context. This review examines interactions between Treg cells and tissue-resident non-immune cells-in the context of tissue repair, fibrosis, and cancer-and discusses areas for future exploration.
Collapse
Affiliation(s)
- Lucas F. Loffredo
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | - Thomas M. Savage
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | - Olivia R. Ringham
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | - Nicholas Arpaia
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| |
Collapse
|
15
|
Caballero-Sánchez N, Alonso-Alonso S, Nagy L. Regenerative inflammation: When immune cells help to re-build tissues. FEBS J 2024; 291:1597-1614. [PMID: 36440547 PMCID: PMC10225019 DOI: 10.1111/febs.16693] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/29/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2022]
Abstract
Inflammation is an essential immune response critical for responding to infection, injury and maintenance of tissue homeostasis. Upon injury, regenerative inflammation promotes tissue repair by a timed and coordinated infiltration of diverse cell types and the secretion of growth factors, cytokines and lipids mediators. Remarkably, throughout evolution as well as mammalian development, this type of physiological inflammation is highly associated with immunosuppression. For instance, regenerative inflammation is the consequence of an in situ macrophage polarization resulting in a transition from pro-inflammatory to anti-inflammatory/pro-regenerative response. Immune cells are the first responders upon injury, infiltrating the damaged tissue and initiating a pro-inflammatory response depleting cell debris and necrotic cells. After phagocytosis, macrophages undergo multiple coordinated metabolic and transcriptional changes allowing the transition and dictating the initiation of the regenerative phase. Differences between a highly efficient, complete ad integrum tissue repair, such as, acute skeletal muscle injury, and insufficient regenerative inflammation, as the one developing in Duchenne Muscular Dystrophy (DMD), highlight the importance of a coordinated response orchestrated by immune cells. During regenerative inflammation, these cells interact with others and alter the niche, affecting the character of inflammation itself and, therefore, the progression of tissue repair. Comparing acute muscle injury and chronic inflammation in DMD, we review how the same cells and molecules in different numbers, concentration and timing contribute to very different outcomes. Thus, it is important to understand and identify the distinct functions and secreted molecules of macrophages, and potentially other immune cells, during tissue repair, and the contributors to the macrophage switch leveraging this knowledge in treating diseases.
Collapse
Affiliation(s)
- Noemí Caballero-Sánchez
- Doctoral School of Molecular Cell and Immunobiology, Faculty of Medicine, University of Debrecen, Hungary
- Department of Biochemistry and Molecular Biology, Nuclear Receptor Research Laboratory, Faculty of Medicine, University of Debrecen, Hungary
| | - Sergio Alonso-Alonso
- Instituto Oftalmológico Fernández-Vega, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Laszlo Nagy
- Department of Biochemistry and Molecular Biology, Nuclear Receptor Research Laboratory, Faculty of Medicine, University of Debrecen, Hungary
- Departments Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, and Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St Petersburg, Florida, USA
| |
Collapse
|
16
|
Diaz-Castro J, Reyes-Olavarría D, Toledano JM, Puche-Juarez M, Garcia-Vega JE, Ochoa JJ, Moreno-Fernandez J. Assessment of muscle endocrine function and inflammatory signalling in male school children following a physical activity programme. Clin Nutr 2024; 43:936-942. [PMID: 38422951 DOI: 10.1016/j.clnu.2024.02.024] [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: 09/29/2023] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND & AIMS Regular and planned physical activity can diminish the risk of numerous illnesses. However, school children and teenagers often exercise intermittently and for brief periods, restricting potential benefits. Furthermore, previous studies mainly focused on body composition, without providing molecular mechanisms elucidating the role of physical activity in muscle tissue and inflammatory signalling. The objective of this study was to determine the effect of a vigorous physical activity intervention on endocrine muscle function and cytokine output in children. METHODS 103 boys were divided into two groups: control (n = 51, did not perform additional physical activity) and exercise (n = 52, performed vigorous physical activity). Body composition measurements, endocrine muscle function and inflammatory signalling biomarkers were assessed at enrolment and after 6 months of intervention. RESULTS No statistical significance was found for fractalkine, oncostatin, EGF, TNF-α and eotaxin. However, LIF, FBAP3, IL-6, FGF21 and IL-15 increased in the exercise group at the end of the protocol, though myostatin got decreased. In contrast, IFN-γ was increased in the exercise group at the beginning and end of the exercise protocol, IL-10 was also increased in this group, IL-1α decreased in the exercise group before and after the exercise protocol, and IP-10 and MCP-1 also decreased in the exercise group. CONCLUSION It can be affirmed that a physical activity programme for boys was shown to produce changes in body composition (decreased fat mass, increased lean mass) and in markers of endocrine muscle function and cytokine release. It is possible that these changes, if sustained, could reduce the risk of chronic disease.
Collapse
Affiliation(s)
- Javier Diaz-Castro
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria (IBS), 18016 Granada, Spain
| | - Daniela Reyes-Olavarría
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, 18071 Granada, Spain; Nutrition and Food Sciences Ph.D. Program, University of Granada, 18071 Granada, Spain; Department of Physical Education, Sport, and Recreation, Universidad de La Frontera, Temuco 4780000, Chile
| | - Juan M Toledano
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, 18071 Granada, Spain; Nutrition and Food Sciences Ph.D. Program, University of Granada, 18071 Granada, Spain.
| | - María Puche-Juarez
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, 18071 Granada, Spain; Nutrition and Food Sciences Ph.D. Program, University of Granada, 18071 Granada, Spain.
| | - Jose Eulogio Garcia-Vega
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, 18071 Granada, Spain; Nutrition and Food Sciences Ph.D. Program, University of Granada, 18071 Granada, Spain
| | - Julio J Ochoa
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria (IBS), 18016 Granada, Spain
| | - Jorge Moreno-Fernandez
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria (IBS), 18016 Granada, Spain.
| |
Collapse
|
17
|
Marzetti E, Lozanoska-Ochser B, Calvani R, Landi F, Coelho-Júnior HJ, Picca A. Restoring Mitochondrial Function and Muscle Satellite Cell Signaling: Remedies against Age-Related Sarcopenia. Biomolecules 2024; 14:415. [PMID: 38672432 PMCID: PMC11048011 DOI: 10.3390/biom14040415] [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: 02/19/2024] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Sarcopenia has a complex pathophysiology that encompasses metabolic dysregulation and muscle ultrastructural changes. Among the drivers of intracellular and ultrastructural changes of muscle fibers in sarcopenia, mitochondria and their quality control pathways play relevant roles. Mononucleated muscle stem cells/satellite cells (MSCs) have been attributed a critical role in muscle repair after an injury. The involvement of mitochondria in supporting MSC-directed muscle repair is unclear. There is evidence that a reduction in mitochondrial biogenesis blunts muscle repair, thus indicating that the delivery of functional mitochondria to injured muscles can be harnessed to limit muscle fibrosis and enhance restoration of muscle function. Injection of autologous respiration-competent mitochondria from uninjured sites to damaged tissue has been shown to reduce infarct size and enhance cell survival in preclinical models of ischemia-reperfusion. Furthermore, the incorporation of donor mitochondria into MSCs enhances lung and cardiac tissue repair. This strategy has also been tested for regeneration purposes in traumatic muscle injuries. Indeed, the systemic delivery of mitochondria promotes muscle regeneration and restores muscle mass and function while reducing fibrosis during recovery after an injury. In this review, we discuss the contribution of altered MSC function to sarcopenia and illustrate the prospect of harnessing mitochondrial delivery and restoration of MSCs as a therapeutic strategy against age-related sarcopenia.
Collapse
Affiliation(s)
- Emanuele Marzetti
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (R.C.); (F.L.)
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy;
| | - Biliana Lozanoska-Ochser
- Department of Medicine and Surgery, LUM University, 70010 Casamassima, Italy;
- DAHFMO Unit of Histology and Medical Embryology, Sapienza Università di Roma, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Riccardo Calvani
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (R.C.); (F.L.)
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy;
| | - Francesco Landi
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (R.C.); (F.L.)
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy;
| | - Hélio José Coelho-Júnior
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy;
| | - Anna Picca
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (R.C.); (F.L.)
- Department of Medicine and Surgery, LUM University, 70010 Casamassima, Italy;
| |
Collapse
|
18
|
Becker M, Dirschl SM, Scherm MG, Serr I, Daniel C. Niche-specific control of tissue function by regulatory T cells-Current challenges and perspectives for targeting metabolic disease. Cell Metab 2024; 36:229-239. [PMID: 38218187 DOI: 10.1016/j.cmet.2023.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/30/2023] [Accepted: 12/15/2023] [Indexed: 01/15/2024]
Abstract
Tissue regulatory T cells (Tregs) exert pivotal functions in both immune and metabolic regulation, maintaining local tissue homeostasis, integrity, and function. Accordingly, Tregs play a crucial role in controlling obesity-induced inflammation and supporting efficient muscle function and repair. Depending on the tissue context, Tregs are characterized by unique transcriptomes, growth, and survival factors and T cell receptor (TCR) repertoires. This functional specialization offers the potential to selectively target context-specific Treg populations, tailoring therapeutic strategies to specific niches, thereby minimizing potential side effects. Here, we discuss challenges and perspectives for niche-specific Treg targeting, which holds promise for highly efficient and precise medical interventions to combat metabolic disease.
Collapse
Affiliation(s)
- Maike Becker
- Research Division Type 1 Diabetes Immunology, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany; Deutsches Zentrum für Diabetesforschung (DZD), 85764 Munich, Germany
| | - Sandra M Dirschl
- Research Division Type 1 Diabetes Immunology, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany; Deutsches Zentrum für Diabetesforschung (DZD), 85764 Munich, Germany
| | - Martin G Scherm
- Research Division Type 1 Diabetes Immunology, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany; Deutsches Zentrum für Diabetesforschung (DZD), 85764 Munich, Germany
| | - Isabelle Serr
- Research Division Type 1 Diabetes Immunology, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany; Deutsches Zentrum für Diabetesforschung (DZD), 85764 Munich, Germany
| | - Carolin Daniel
- Research Division Type 1 Diabetes Immunology, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany; Deutsches Zentrum für Diabetesforschung (DZD), 85764 Munich, Germany; Division of Clinical Pharmacology, Department of Medicine IV, Ludwig-Maximilians-Universität München, 80336 Munich, Germany.
| |
Collapse
|
19
|
Cui H, Wang N, Li H, Bian Y, Wen W, Kong X, Wang F. The dynamic shifts of IL-10-producing Th17 and IL-17-producing Treg in health and disease: a crosstalk between ancient "Yin-Yang" theory and modern immunology. Cell Commun Signal 2024; 22:99. [PMID: 38317142 PMCID: PMC10845554 DOI: 10.1186/s12964-024-01505-0] [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: 12/07/2023] [Accepted: 01/28/2024] [Indexed: 02/07/2024] Open
Abstract
The changes in T regulatory cell (Treg) and T helper cell (Th) 17 ratios holds paramount importance in ensuring internal homeostasis and disease progression. Recently, novel subsets of Treg and Th17, namely IL-17-producing Treg and IL-10-producing Th17 have been identified. IL-17-producing Treg and IL-10-producing Th17 are widely considered as the intermediates during Treg/Th17 transformation. These "bi-functional" cells exhibit plasticity and have been demonstrated with important roles in multiple physiological functions and disease processes. Yin and Yang represent opposing aspects of phenomena according to the ancient Chinese philosophy "Yin-Yang" theory. Furthermore, Yin can transform into Yang, and vice versa, under specific conditions. This theory has been widely used to describe the contrasting functions of immune cells and molecules. Therefore, immune-activating populations (Th17, M1 macrophage, etc.) and immune overreaction (inflammation, autoimmunity) can be considered Yang, while immunosuppressive populations (Treg, M2 macrophage, etc.) and immunosuppression (tumor, immunodeficiency) can be considered Yin. However, another important connotation of "Yin-Yang" theory, the conversion between Yin and Yang, has been rarely documented in immune studies. The discovery of IL-17-producing Treg and IL-10-producing Th17 enriches the meaning of "Yin-Yang" theory and further promotes the relationship between ancient "Yin-Yang" theory and modern immunology. Besides, illustrating the functions of IL-17-producing Treg and IL-10-producing Th17 and mechanisms governing their differentiation provides valuable insights into the mechanisms underlying the dynamically changing statement of immune statement in health and diseases.
Collapse
Affiliation(s)
- Huantian Cui
- First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Ning Wang
- First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Hanzhou Li
- College of Integrative Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yuhong Bian
- College of Integrative Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Weibo Wen
- First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China.
| | - Xiangying Kong
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Fudi Wang
- The First Affiliated Hospital, Institute of Translational Medicine, The Second Affiliated Hospital, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
20
|
Bartolacci JG, Behun MN, Warunek JP, Li T, Sahu A, Dwyer GK, Lucas A, Rong J, Ambrosio F, Turnquist HR, Badylak SF. Matrix-bound nanovesicle-associated IL-33 supports functional recovery after skeletal muscle injury by initiating a pro-regenerative macrophage phenotypic transition. NPJ Regen Med 2024; 9:7. [PMID: 38280914 PMCID: PMC10821913 DOI: 10.1038/s41536-024-00346-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/04/2024] [Indexed: 01/29/2024] Open
Abstract
Injuries to skeletal muscle are among the most common injuries in civilian and military populations, accounting for nearly 60% of extremity injuries. The standard of care for severe extremity injury has been focused upon limb salvage procedures and the utilization of tissue grafts or orthotics in conjunction with rehabilitation to avoid amputation. Nonetheless, many patients have persistent strength and functional deficits that permanently impact their quality of life. Preclinical and clinical studies have shown that partial restoration of functional skeletal muscle tissue following injury can be achieved by the implantation of a biologic scaffold composed of extracellular matrix (ECM). These favorable outcomes are mediated, at least in part, through local immunomodulation. The mechanisms underlying this immunomodulatory effect, however, are poorly understood. The present study investigates a potential mechanistic driver of the immunomodulatory effects; specifically, the effect of selected ECM components upon inflammation resolution and repair. Results show that the host response to skeletal muscle injury is profoundly altered and functional recovery decreased in il33-/- mice compared to age- and sex-matched wildtype counterparts by 14 days post-injury. Results also show that IL-33, contained within matrix-bound nanovesicles (MBV), supports skeletal muscle regeneration by regulating local macrophage activation toward a pro-remodeling phenotype via canonical and non-canonical pathways to improve functional recovery from injury compared to untreated il33-/- counterparts. Taken together, these data suggest that MBV and their associated IL-33 cargo represent a novel homeostatic signaling mechanism that contributes to skeletal muscle repair.
Collapse
Affiliation(s)
- J G Bartolacci
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - M N Behun
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - J P Warunek
- Departments of Surgery and Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - T Li
- Departments of Surgery and Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - A Sahu
- Department of Physical Medicine and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - G K Dwyer
- Departments of Surgery and Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - A Lucas
- Departments of Surgery and Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - J Rong
- McGowan Institute for Regenerative Medicine, Pittsburgh, USA
| | - F Ambrosio
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Physical Medicine and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, Pittsburgh, USA
| | - H R Turnquist
- Departments of Surgery and Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
- McGowan Institute for Regenerative Medicine, Pittsburgh, USA.
| | - S F Badylak
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Departments of Surgery and Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
- McGowan Institute for Regenerative Medicine, Pittsburgh, USA.
| |
Collapse
|
21
|
Lokwani R, Josyula A, Ngo TB, DeStefano S, Fertil D, Faust M, Adusei KM, Bhuiyan M, Lin A, Karkanitsa M, Maclean E, Fathi P, Su Y, Liu J, Vishwasrao HD, Sadtler K. Pro-regenerative biomaterials recruit immunoregulatory dendritic cells after traumatic injury. NATURE MATERIALS 2024; 23:147-157. [PMID: 37872423 DOI: 10.1038/s41563-023-01689-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 09/12/2023] [Indexed: 10/25/2023]
Abstract
During wound healing and surgical implantation, the body establishes a delicate balance between immune activation to fight off infection and clear debris and immune tolerance to control reactivity against self-tissue. Nonetheless, how such a balance is achieved is not well understood. Here we describe that pro-regenerative biomaterials for muscle injury treatment promote the proliferation of a BATF3-dependent CD103+XCR1+CD206+CD301b+ dendritic cell population associated with cross-presentation and self-tolerance. Upregulation of E-cadherin, the ligand for CD103, and XCL-1 in injured tissue suggests a mechanism for cell recruitment to trauma. Muscle injury recruited natural killer cells that produced Xcl1 when stimulated with fragmented extracellular matrix. Without cross-presenting cells, T-cell activation increases, pro-regenerative macrophage polarization decreases and there are alterations in myogenesis, adipogenesis, fibrosis and increased muscle calcification. These results, previously observed in cancer progression, suggest a fundamental mechanism of immune regulation in trauma and material implantation with implications for both short- and long-term injury recovery.
Collapse
Affiliation(s)
- Ravi Lokwani
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Aditya Josyula
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Tran B Ngo
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Sabrina DeStefano
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Daphna Fertil
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Mondreakest Faust
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Kenneth M Adusei
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Minhaj Bhuiyan
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Aaron Lin
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
- Unit for Nanoengineering and Microphysiological Systems, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Maria Karkanitsa
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Efua Maclean
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Parinaz Fathi
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
- Unit for Nanoengineering and Microphysiological Systems, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Yijun Su
- Advanced Imaging and Microscopy Resource, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Jiamin Liu
- Advanced Imaging and Microscopy Resource, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Harshad D Vishwasrao
- Advanced Imaging and Microscopy Resource, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Kaitlyn Sadtler
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
22
|
Jayewickreme T, Benoist C, Mathis D. Lymph node stromal cell responses to perinatal T cell waves, a temporal atlas. Proc Natl Acad Sci U S A 2023; 120:e2316957120. [PMID: 38079541 PMCID: PMC10740392 DOI: 10.1073/pnas.2316957120] [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: 09/29/2023] [Accepted: 10/30/2023] [Indexed: 12/18/2023] Open
Abstract
The perinatal period is a critical time window in establishing T cell tolerance. Regulatory T cells (Tregs) made during the first 2 wk of life are key drivers of perinatal tolerance induction, but how these cells are generated and operate has not been established. To elucidate the unique environment murine perinatal Tregs encounter within the lymph nodes (LNs) as they first emerge from the thymus, and how it evolves over the succeeding days, we employed single-cell RNA sequencing to generate an atlas of the early LN niche. A highly dynamic picture emerged, the stromal cell compartment showing the most striking changes and putative interactions with other LN cell compartments. In particular, LN stromal cells showed increasing potential for lymphocyte interactions with age. Analogous studies on mice lacking α:β T cells or enriched for autoreactive α:β T cells revealed an acute stromal cell response to α:β T cell dysfunction, largely reflecting dysregulation of Tregs. Punctual ablation of perinatal Tregs induced stromal cell activation that was dependent on both interferon-gamma signaling and activation of conventional CD4+ T cells. These findings elucidate some of the earliest cellular and molecular events in perinatal induction of T cell tolerance, providing a framework for future explorations.
Collapse
Affiliation(s)
| | | | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA02115
| |
Collapse
|
23
|
Yang W, Pan Z, Zhang J, Wang L, lai J, Zhou S, Zhang Z, Fan K, Deng D, Gao Z, Yu S. Extracellular vesicles from adipose stem cells ameliorate allergic rhinitis in mice by immunomodulatory. Front Immunol 2023; 14:1302336. [PMID: 38143758 PMCID: PMC10739383 DOI: 10.3389/fimmu.2023.1302336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/24/2023] [Indexed: 12/26/2023] Open
Abstract
Background Human adipose tissue-derived stem cells (hADSCs) exert potent immunosuppressive effects in the allogeneic transplantation treatment. In mouse model of allergic rhinitis (AR), ADSCs partially ameliorated AR. However, no study has evaluated the potential therapeutic effects of hADSC-derived extracellular vesicles (hADSC-EVs) on AR. Methods Female BALB/c mice were sensitized and challenged with ovalbumin (OVA) to induce AR. One day after the last nasal drop, each group received phosphate buffered saline (PBS) or hADSC-EVs treatment. Associated symptoms and biological changes were then assessed. Results hADSC-EV treatment significantly alleviated nasal symptoms, and reduced inflammatory infiltration. Serum levels of OVA-specific IgE, interleukin (IL)-4 and interferon (IFN)-γ were all significantly reduced. The mRNA levels of IL-4 and IFN-γ in the spleen also changed accordingly. The T helper (Th)1/Th2 cell ratio increased. The treatment efficacy index of hADSC-EV was higher than that of all human-derived MSCs in published reports on MSC treatment of AR. ADSC-EVs exhibited a greater therapeutic index in most measures when compared to our previous treatment involving ADSCs. Conclusion These results demonstrated that hADSC-EVs could ameliorate the symptoms of AR by modulating cytokine secretion and Th1/Th2 cell balance. hADSC-EVs could potentially be a viable therapeutic strategy for AR. Further animal studies are needed to elucidate the underlying mechanisms and to optimize potential clinical protocols.
Collapse
Affiliation(s)
- Wenhan Yang
- Department of Otorhinolaryngology Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Zhiyu Pan
- Department of Otorhinolaryngology Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiacheng Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lian Wang
- School of Medicine, Tongji University, Shanghai, China
| | - Ju lai
- Department of Otorhinolaryngology Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Shican Zhou
- Department of Otorhinolaryngology Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Medicine, Tongji University, Shanghai, China
| | - Zhili Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Kai Fan
- Department of Otorhinolaryngology Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dan Deng
- Department of Dermatology, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Dermatology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhengliang Gao
- Fundamental Research Center, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
| | - Shaoqing Yu
- Department of Otorhinolaryngology Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Allergy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
24
|
Yaghi OK, Hanna BS, Langston PK, Michelson DA, Jayewickreme T, Marin-Rodero M, Benoist C, Mathis D. A discrete 'early-responder' stromal-cell subtype orchestrates immunocyte recruitment to injured tissue. Nat Immunol 2023; 24:2053-2067. [PMID: 37932455 PMCID: PMC10792729 DOI: 10.1038/s41590-023-01669-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/05/2023] [Indexed: 11/08/2023]
Abstract
Following acute injury, stromal cells promote tissue regeneration by a diversity of mechanisms. Time-resolved single-cell RNA sequencing of muscle mesenchymal stromal cells (MmSCs) responding to acute injury identified an 'early-responder' subtype that spiked on day 1 and expressed a notable array of transcripts encoding immunomodulators. IL-1β, TNF-α and oncostatin M each strongly and rapidly induced MmSCs transcribing this immunomodulatory program. Macrophages amplified the program but were not strictly required for its induction. Transfer of the inflammatory MmSC subtype, tagged with a unique surface marker, into healthy hindlimb muscle induced inflammation primarily driven by neutrophils and macrophages. Among the abundant inflammatory transcripts produced by this subtype, Cxcl5 was stroma-specific and highly upregulated with injury. Depletion of this chemokine early after injury revealed a substantial impact on recruitment of neutrophils, a prolongation of inflammation to later times and an effect on tissue regeneration. Mesenchymal stromal cell subtypes expressing a comparable inflammatory program were found in a mouse model of muscular dystrophy and in several other tissues and pathologies in both mice and humans. These 'early-responder' mesenchymal stromal cells, already in place, permit rapid and coordinated mobilization and amplification of critical cell collaborators in response to injury.
Collapse
Affiliation(s)
- Omar K Yaghi
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Bola S Hanna
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - P Kent Langston
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Daniel A Michelson
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Teshika Jayewickreme
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Miguel Marin-Rodero
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Christophe Benoist
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA, USA.
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
25
|
Langston PK, Sun Y, Ryback BA, Mueller AL, Spiegelman BM, Benoist C, Mathis D. Regulatory T cells shield muscle mitochondria from interferon-γ-mediated damage to promote the beneficial effects of exercise. Sci Immunol 2023; 8:eadi5377. [PMID: 37922340 PMCID: PMC10860652 DOI: 10.1126/sciimmunol.adi5377] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 10/02/2023] [Indexed: 11/05/2023]
Abstract
Exercise enhances physical performance and reduces the risk of many disorders such as cardiovascular disease, type 2 diabetes, dementia, and cancer. Exercise characteristically incites an inflammatory response, notably in skeletal muscles. Although some effector mechanisms have been identified, regulatory elements activated in response to exercise remain obscure. Here, we have addressed the roles of Foxp3+CD4+ regulatory T cells (Tregs) in the healthful activities of exercise via immunologic, transcriptomic, histologic, metabolic, and biochemical analyses of acute and chronic exercise models in mice. Exercise rapidly induced expansion of the muscle Treg compartment, thereby guarding against overexuberant production of interferon-γ and consequent metabolic disruptions, particularly mitochondrial aberrancies. The performance-enhancing effects of exercise training were dampened in the absence of Tregs. Thus, exercise is a natural Treg booster with therapeutic potential in disease and aging contexts.
Collapse
Affiliation(s)
- P. Kent Langston
- Department of Immunology, Harvard Medical School; Boston, 02115, USA
| | - Yizhi Sun
- Department of Cancer Biology, Dana-Farber Cancer Institute; Boston, 02115, USA
- Department of Cell Biology, Harvard Medical School; Boston, 02115, USA
| | - Birgitta A. Ryback
- Department of Cancer Biology, Dana-Farber Cancer Institute; Boston, 02115, USA
| | - Amber L. Mueller
- Department of Genetics, Harvard Medical School; Boston, 02115, USA
| | - Bruce M. Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute; Boston, 02115, USA
- Department of Cell Biology, Harvard Medical School; Boston, 02115, USA
| | | | - Diane Mathis
- Department of Immunology, Harvard Medical School; Boston, 02115, USA
| |
Collapse
|
26
|
Tarban N, Papp AB, Deák D, Szentesi P, Halász H, Patsalos A, Csernoch L, Sarang Z, Szondy Z. Loss of adenosine A3 receptors accelerates skeletal muscle regeneration in mice following cardiotoxin-induced injury. Cell Death Dis 2023; 14:706. [PMID: 37898628 PMCID: PMC10613231 DOI: 10.1038/s41419-023-06228-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/13/2023] [Accepted: 10/18/2023] [Indexed: 10/30/2023]
Abstract
Skeletal muscle regeneration is a complex process orchestrated by multiple interacting steps. An increasing number of reports indicate that inflammatory responses play a central role in linking initial muscle injury responses to timely muscle regeneration following injury. The nucleoside adenosine has been known for a long time as an endogenously produced anti-inflammatory molecule that is generated in high amounts during tissue injury. It mediates its physiological effects via four types of adenosine receptors. From these, adenosine A3 receptors (A3Rs) are not expressed by the skeletal muscle but are present on the surface of various inflammatory cells. In the present paper, the effect of the loss of A3Rs was investigated on the regeneration of the tibialis anterior (TA) muscle in mice following cardiotoxin-induced injury. Here we report that regeneration of the skeletal muscle from A3R-/- mice is characterized by a stronger initial inflammatory response resulting in a larger number of transmigrating inflammatory cells to the injury site, faster clearance of cell debris, enhanced proliferation and faster differentiation of the satellite cells (the muscle stem cells), and increased fusion of the generated myoblasts. This leads to accelerated skeletal muscle tissue repair and the formation of larger myofibers. Though the infiltrating immune cells expressed A3Rs and showed an increased inflammatory profile in the injured A3R-/- muscles, bone marrow transplantation experiments revealed that the increased response of the tissue-resident cells to tissue injury is responsible for the observed phenomenon. Altogether our data indicate that A3Rs are negative regulators of injury-related regenerative inflammation and consequently also that of the muscle fiber growth in the TA muscle. Thus, inhibiting A3Rs might have a therapeutic value during skeletal muscle regeneration following injury.
Collapse
Affiliation(s)
- Nastaran Tarban
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Debrecen, Hungary
| | - Albert Bálint Papp
- Doctoral School of Dental Sciences, University of Debrecen, Debrecen, Hungary
| | - Dávid Deák
- Laboratory Animal Facility, Life Science Building, University of Debrecen, Debrecen, Hungary
| | - Péter Szentesi
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Hajnalka Halász
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Debrecen, Hungary
| | - Andreas Patsalos
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St, Petersburg, FL, USA
| | - László Csernoch
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Sarang
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsuzsa Szondy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
- Division of Dental Biochemistry, Department of Basic Medical Sciences, Faculty of Dentistry, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
27
|
Becker M, Joseph SS, Garcia-Carrizo F, Tom RZ, Opaleva D, Serr I, Tschöp MH, Schulz TJ, Hofmann SM, Daniel C. Regulatory T cells require IL6 receptor alpha signaling to control skeletal muscle function and regeneration. Cell Metab 2023; 35:1736-1751.e7. [PMID: 37734370 PMCID: PMC10563138 DOI: 10.1016/j.cmet.2023.08.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 02/27/2023] [Accepted: 08/31/2023] [Indexed: 09/23/2023]
Abstract
Muscle-residing regulatory T cells (Tregs) control local tissue integrity and function. However, the molecular interface connecting Treg-based regulation with muscle function and regeneration remains largely unexplored. Here, we show that exercise fosters a stable induction of highly functional muscle-residing Tregs with increased expression of amphiregulin (Areg), EGFR, and ST2. Mechanistically, we find that mice lacking IL6Rα on T cells (TKO) harbor significant reductions in muscle Treg functionality and satellite and fibro-adipogenic progenitor cells, which are required for muscle regeneration. Using exercise and sarcopenia models, IL6Rα TKO mice demonstrate deficits in Tregs, their functional maturation, and a more pronounced decline in muscle mass. Muscle injury models indicate that IL6Rα TKO mice have significant disabilities in muscle regeneration. Treg gain of function restores impaired muscle repair in IL6Rα TKO mice. Of note, pharmacological IL6R blockade in WT mice phenocopies deficits in muscle function identified in IL6Rα TKO mice, thereby highlighting the clinical implications of the findings.
Collapse
Affiliation(s)
- Maike Becker
- Research Unit Type 1 Diabetes Immunology, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Munich-Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany
| | - Sini S Joseph
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Munich-Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 80333 Munich, Germany
| | - Francisco Garcia-Carrizo
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany
| | - Robby Z Tom
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Institute for Diabetes and Regeneration, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Munich-Neuherberg, Germany; Department of Medicine IV, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Daria Opaleva
- Research Unit Type 1 Diabetes Immunology, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Munich-Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany
| | - Isabelle Serr
- Research Unit Type 1 Diabetes Immunology, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Munich-Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany
| | - Matthias H Tschöp
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Munich-Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 80333 Munich, Germany
| | - Tim J Schulz
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany; University of Potsdam, Institute of Nutritional Science, Potsdam-Rehbrücke, 14558 Nuthetal, Germany
| | - Susanna M Hofmann
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Institute for Diabetes and Regeneration, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Munich-Neuherberg, Germany; Department of Medicine IV, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Carolin Daniel
- Research Unit Type 1 Diabetes Immunology, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Munich-Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Division of Clinical Pharmacology, Department of Medicine IV, Ludwig-Maximilians-Universität München, 80337 Munich, Germany.
| |
Collapse
|
28
|
Wu H, Guo C, Liu Z, Cai J, Wang C, Yi H, Sanyal A, Puri P, Zhou H, Wang XY. Neutrophils exacerbate acetaminophen-induced liver injury by producing cytotoxic interferon-γ. Int Immunopharmacol 2023; 123:110734. [PMID: 37541108 PMCID: PMC10603570 DOI: 10.1016/j.intimp.2023.110734] [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: 03/20/2023] [Revised: 07/27/2023] [Accepted: 07/27/2023] [Indexed: 08/06/2023]
Abstract
BACKGROUND Drug (e.g., acetaminophen, APAP)-associated hepatotoxicity is the major cause of acute liver failure. Emerging evidence shows that initial tissue damage caused by APAP triggers molecular and cellular immune responses, which can modulate the severity of hepatoxicity. The pro-inflammatory and cytotoxic cytokine interferon (IFN)-γ has been reported as a key molecule contributing to APAP-induced liver injury (AILI). However, its cellular source remains undetermined. RESULTS In the current study, we show that elevation of serum IFN-γ in patients with drug hepatotoxicity correlates with disease severity. Neutralization of IFN-γ in a mouse model of AILI effectively reduces hepatotoxicity. Strikingly, we reveal that IFN-γ is expressed primarily by hepatic neutrophils, not by conventional immune cells with known IFN-γ-producing capability, e.g., CD8+ T cells, CD4+ T cells, natural killer cells, or natural killer T cells. Upon encountering APAP-injured hepatocytes, neutrophils secrete cytotoxic IFN-γ further causing cell stress and damage, which can be abrogated in the presence of blocking antibodies for IFN-γ or IFN-γreceptor. Furthermore, removal of neutrophils in vivo substantially decreases hepatic IFN-γ levels concomitantly with reduced APAP hepatotoxicity, whereas adoptive transfer of IFN-γ-producing neutrophils confers IFN-γ-/- mice susceptibility to APAP administration. CONCLUSIONS Our findings uncover a novel mechanism of neutrophil action in promoting AILI and provide new insights into immune modulation of the disease pathogenesis.
Collapse
Affiliation(s)
- Hao Wu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Chunqing Guo
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA; Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA; Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA.
| | - Zheng Liu
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Jinyang Cai
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Chong Wang
- Nephrology Department, First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Huanfa Yi
- Central Laboratory, The First Hospital of Jilin University, Changchun, Jilin 130021, China; Key Laboratory of Organ Regeneration and Transplantation, Ministry of Education, Changchun, Jilin 130021, China
| | - Arun Sanyal
- Division of Gastroenterology, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Puneet Puri
- Richmond VA Medical Center, Richmond 23249, VA, USA
| | - Huiping Zhou
- Richmond VA Medical Center, Richmond 23249, VA, USA; Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Xiang-Yang Wang
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA; Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA; Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA; Richmond VA Medical Center, Richmond 23249, VA, USA.
| |
Collapse
|
29
|
Barrachina F, Ottino K, Elizagaray ML, Gervasi MG, Tu LJ, Markoulaki S, Spallanzani RG, Capen D, Brown D, Battistone MA. Regulatory T cells play a crucial role in maintaining sperm tolerance and male fertility. Proc Natl Acad Sci U S A 2023; 120:e2306797120. [PMID: 37676910 PMCID: PMC10500189 DOI: 10.1073/pnas.2306797120] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/31/2023] [Indexed: 09/09/2023] Open
Abstract
Regulatory T cells (Tregs) modulate tissue homeostatic processes and immune responses. Understanding tissue-Treg biology will contribute to developing precision-targeting treatment strategies. Here, we show that Tregs maintain the tolerogenic state of the testis and epididymis, where sperm are produced and mature. We found that Treg depletion induces severe autoimmune orchitis and epididymitis, manifested by an exacerbated immune cell infiltration [CD4 T cells, monocytes, and mononuclear phagocytes (MPs)] and the development of antisperm antibodies (ASA). In Treg-depleted mice, MPs increased projections toward the epididymal lumen as well as invading the lumen. ASA-bound sperm enhance sperm agglutination and might facilitate sperm phagocytosis. Tolerance breakdown impaired epididymal epithelial function and altered extracellular vesicle cargo, both of which play crucial roles in the acquisition of sperm fertilizing ability and subsequent embryo development. The affected mice had reduced sperm number and motility and severe fertility defects. Deciphering these immunoregulatory mechanisms may help to design new strategies to treat male infertility, as well as to identify potential targets for immunocontraception.
Collapse
Affiliation(s)
- Ferran Barrachina
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA02129
| | - Kiera Ottino
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA02129
| | - Maia Lina Elizagaray
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA02129
| | - Maria Gracia Gervasi
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA01003
- Genetically Engineered Models Center, Whitehead Institute of Biomedical Research, Cambridge, MA02142
| | - Leona J. Tu
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA02129
| | - Styliani Markoulaki
- Genetically Engineered Models Center, Whitehead Institute of Biomedical Research, Cambridge, MA02142
| | - Raul G. Spallanzani
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA02115
| | - Diane Capen
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA02129
| | - Dennis Brown
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA02129
| | - Maria Agustina Battistone
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA02129
| |
Collapse
|
30
|
Sun R, Zhao H, Gao DS, Ni A, Li H, Chen L, Lu X, Chen K, Lu B. Amphiregulin couples IL1RL1 + regulatory T cells and cancer-associated fibroblasts to impede antitumor immunity. SCIENCE ADVANCES 2023; 9:eadd7399. [PMID: 37611111 PMCID: PMC10446484 DOI: 10.1126/sciadv.add7399] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 06/27/2023] [Indexed: 08/25/2023]
Abstract
Regulatory T (Treg) cells and cancer-associated fibroblasts (CAFs) jointly promote tumor immune tolerance and tumorigenesis. The molecular apparatus that drives Treg cell and CAF coordination in the tumor microenvironment (TME) remains elusive. Interleukin 33 (IL-33) has been shown to enhance fibrosis and IL1RL1+ Treg cell accumulation during tumorigenesis and tissue repair. We demonstrated that IL1RL1 signaling in Treg cells greatly dampened the antitumor activity of both IL-33 and PD-1 blockade. Whole tumor single-cell RNA sequencing (scRNA-seq) analysis and blockade experiments revealed that the amphiregulin (AREG)-epidermal growth factor receptor (EGFR) axis mediated cross-talk between IL1RL1+ Treg cells and CAFs. We further demonstrated that the AREG/EGFR axis enables Treg cells to promote a profibrotic and immunosuppressive functional state of CAFs. Moreover, AREG mAbs and IL-33 concertedly inhibited tumor growth. Our study reveals a previously unidentified AREG/EGFR-mediated Treg/CAF coupling that controls the bifurcation of fibroblast functional states and is a critical barrier for cancer immunotherapy.
Collapse
Affiliation(s)
- Runzi Sun
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hongyu Zhao
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - David Shihong Gao
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Andrew Ni
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Haochen Li
- Department of Biomedical informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lujia Chen
- Department of Biomedical informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xinghua Lu
- Department of Biomedical informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kong Chen
- Department of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Binfeng Lu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| |
Collapse
|
31
|
Hymel LA, Anderson SE, Turner TC, York WY, Zhang H, Liversage AR, Lim HS, Qiu P, Mortensen LJ, Jang YC, Willett NJ, Botchwey EA. Identifying dysregulated immune cell subsets following volumetric muscle loss with pseudo-time trajectories. Commun Biol 2023; 6:749. [PMID: 37468760 PMCID: PMC10356763 DOI: 10.1038/s42003-023-04790-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/31/2023] [Indexed: 07/21/2023] Open
Abstract
Volumetric muscle loss (VML) results in permanent functional deficits and remains a substantial regenerative medicine challenge. A coordinated immune response is crucial for timely myofiber regeneration, however the immune response following VML has yet to be fully characterized. Here, we leveraged dimensionality reduction and pseudo-time analysis techniques to elucidate the cellular players underlying a functional or pathological outcome as a result of subcritical injury or critical VML in the murine quadriceps, respectively. We found that critical VML resulted in a sustained presence of M2-like and CD206hiLy6Chi 'hybrid' macrophages whereas subcritical defects resolved these populations. Notably, the retained M2-like macrophages from critical VML injuries presented with aberrant cytokine production which may contribute to fibrogenesis, as indicated by their co-localization with fibroadipogenic progenitors (FAPs) in areas of collagen deposition within the defect. Furthermore, several T cell subpopulations were significantly elevated in critical VML compared to subcritical injuries. These results demonstrate a dysregulated immune response in critical VML that is unable to fully resolve the chronic inflammatory state and transition to a pro-regenerative microenvironment within the first week after injury. These data provide important insights into potential therapeutic strategies which could reduce the immune cell burden and pro-fibrotic signaling characteristic of VML.
Collapse
Affiliation(s)
- Lauren A Hymel
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Shannon E Anderson
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Thomas C Turner
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - William Y York
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Hongmanlin Zhang
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Adrian R Liversage
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA, USA
| | - Hong Seo Lim
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Peng Qiu
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Luke J Mortensen
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA, USA
| | - Young C Jang
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
- Department of Orthopaedics, Emory University, Atlanta, GA, USA.
| | - Nick J Willett
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
- Department of Orthopaedics, Emory University, Atlanta, GA, USA.
- Atlanta Veterans Affairs Medical Center, Decatur, GA, USA.
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, USA.
- The Veterans Affairs Portland Health Care System, Portland, OR, USA.
| | - Edward A Botchwey
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
32
|
McDaniels JM, Shetty AC, Kuscu C, Kuscu C, Bardhi E, Rousselle T, Drachenberg C, Talwar M, Eason JD, Muthukumar T, Maluf DG, Mas VR. Single nuclei transcriptomics delineates complex immune and kidney cell interactions contributing to kidney allograft fibrosis. Kidney Int 2023; 103:1077-1092. [PMID: 36863444 PMCID: PMC10200746 DOI: 10.1016/j.kint.2023.02.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 01/16/2023] [Accepted: 02/07/2023] [Indexed: 03/04/2023]
Abstract
Chronic allograft dysfunction (CAD), characterized histologically by interstitial fibrosis and tubular atrophy, is the major cause of kidney allograft loss. Here, using single nuclei RNA sequencing and transcriptome analysis, we identified the origin, functional heterogeneity, and regulation of fibrosis-forming cells in kidney allografts with CAD. A robust technique was used to isolate individual nuclei from kidney allograft biopsies and successfully profiled 23,980 nuclei from five kidney transplant recipients with CAD and 17,913 nuclei from three patients with normal allograft function. Our analysis revealed two distinct states of fibrosis in CAD; low and high extracellular matrix (ECM) with distinct kidney cell subclusters, immune cell types, and transcriptional profiles. Imaging mass cytometry analysis confirmed increased ECM deposition at the protein level. Proximal tubular cells transitioned to an injured mixed tubular (MT1) phenotype comprised of activated fibroblasts and myofibroblast markers, generated provisional ECM which recruited inflammatory cells, and served as the main driver of fibrosis. MT1 cells in the high ECM state achieved replicative repair evidenced by dedifferentiation and nephrogenic transcriptional signatures. MT1 in the low ECM state showed decreased apoptosis, decreased cycling tubular cells, and severe metabolic dysfunction, limiting the potential for repair. Activated B, T and plasma cells were increased in the high ECM state, while macrophage subtypes were increased in the low ECM state. Intercellular communication between kidney parenchymal cells and donor-derived macrophages, detected several years post-transplantation, played a key role in injury propagation. Thus, our study identified novel molecular targets for interventions aimed to ameliorate or prevent allograft fibrogenesis in kidney transplant recipients.
Collapse
Affiliation(s)
- Jennifer M McDaniels
- Division of Surgical Sciences, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Amol C Shetty
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Cem Kuscu
- Transplant Research Institute, James D. Eason Transplant Institute, University of Tennessee Health Science Center, Memphis, Tennessee, USA; Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Canan Kuscu
- Transplant Research Institute, James D. Eason Transplant Institute, University of Tennessee Health Science Center, Memphis, Tennessee, USA; Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Elissa Bardhi
- Division of Surgical Sciences, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Thomas Rousselle
- Division of Surgical Sciences, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Cinthia Drachenberg
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Manish Talwar
- Transplant Research Institute, James D. Eason Transplant Institute, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - James D Eason
- Transplant Research Institute, James D. Eason Transplant Institute, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Thangamani Muthukumar
- Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Daniel G Maluf
- Division of Surgical Sciences, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA; Program in Transplantation, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Valeria R Mas
- Division of Surgical Sciences, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
33
|
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
|
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
|
Astarita JL, Dominguez CX, Tan C, Guillen J, Pauli ML, Labastida R, Valle J, Kleinschek M, Lyons J, Zarrin AA. Treg specialization and functions beyond immune suppression. Clin Exp Immunol 2023; 211:176-183. [PMID: 36571811 PMCID: PMC10019124 DOI: 10.1093/cei/uxac123] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/25/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
The actions of the immune system are finely tuned, involving complex communication and coordination between diverse immune and non-immune cells across the tissues of the body. A healthy immune system requires a precise balance between immunity and tolerance. Regulatory T cells (Tregs) have long been appreciated as one of the master regulators of this balance; their importance is underscored by the autoimmunity that develops in mice and humans when Tregs are missing or dysfunctional. In addition to the immunoregulatory roles of Tregs in suppressing autoimmunity and inflammation via control of adaptive and innate immune responses, several non-immune modulatory functions of Tregs have been identified in recent years. In this review, we have highlighted the growing literature on the action of Tregs in metabolism, stem cell maintenance, tissue repair, and angiogenesis. Alongside Tregs' immune suppressive role, these non-suppressive activities comprise a key function of Tregs in regulating health and disease. As Tregs receive increasing attention as therapeutic targets, understanding their non-canonical functions may become an important feature of Treg-directed interventions.
Collapse
Affiliation(s)
| | | | - Corey Tan
- TRex Biosciences, South San Francisco, CA, USA
| | | | | | | | - Jose Valle
- TRex Biosciences, South San Francisco, CA, USA
| | | | - Jesse Lyons
- TRex Biosciences, South San Francisco, CA, USA
| | - Ali A Zarrin
- Correspondence: TRexBio, fourth floor, 681 Gateway Blvd., South San Francisco, CA 94080, USA.
| |
Collapse
|
36
|
Habanjar O, Bingula R, Decombat C, Diab-Assaf M, Caldefie-Chezet F, Delort L. Crosstalk of Inflammatory Cytokines within the Breast Tumor Microenvironment. Int J Mol Sci 2023; 24:4002. [PMID: 36835413 PMCID: PMC9964711 DOI: 10.3390/ijms24044002] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Several immune and immunocompetent cells, including dendritic cells, macrophages, adipocytes, natural killer cells, T cells, and B cells, are significantly correlated with the complex discipline of oncology. Cytotoxic innate and adaptive immune cells can block tumor proliferation, and others can prevent the immune system from rejecting malignant cells and provide a favorable environment for tumor progression. These cells communicate with the microenvironment through cytokines, a chemical messenger, in an endocrine, paracrine, or autocrine manner. These cytokines play an important role in health and disease, particularly in host immune responses to infection and inflammation. They include chemokines, interleukins (ILs), adipokines, interferons, colony-stimulating factors (CSFs), and tumor necrosis factor (TNF), which are produced by a wide range of cells, including immune cells, such as macrophages, B-cells, T-cells, and mast cells, as well as endothelial cells, fibroblasts, a variety of stromal cells, and some cancer cells. Cytokines play a crucial role in cancer and cancer-related inflammation, with direct and indirect effects on tumor antagonistic or tumor promoting functions. They have been extensively researched as immunostimulatory mediators to promote the generation, migration and recruitment of immune cells that contribute to an effective antitumor immune response or pro-tumor microenvironment. Thus, in many cancers such as breast cancer, cytokines including leptin, IL-1B, IL-6, IL-8, IL-23, IL-17, and IL-10 stimulate while others including IL-2, IL-12, and IFN-γ, inhibit cancer proliferation and/or invasion and enhance the body's anti-tumor defense. Indeed, the multifactorial functions of cytokines in tumorigenesis will advance our understanding of cytokine crosstalk pathways in the tumor microenvironment, such as JAK/STAT, PI3K, AKT, Rac, MAPK, NF-κB, JunB, cFos, and mTOR, which are involved in angiogenesis, cancer proliferation and metastasis. Accordingly, targeting and blocking tumor-promoting cytokines or activating and amplifying tumor-inhibiting cytokines are considered cancer-directed therapies. Here, we focus on the role of the inflammatory cytokine system in pro- and anti-tumor immune responses, discuss cytokine pathways involved in immune responses to cancer and some anti-cancer therapeutic applications.
Collapse
Affiliation(s)
- Ola Habanjar
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Rea Bingula
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Caroline Decombat
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Mona Diab-Assaf
- Equipe Tumorigénèse Pharmacologie Moléculaire et Anticancéreuse, Faculté des Sciences II, Université Libanaise Fanar, Beyrouth 1500, Lebanon
| | - Florence Caldefie-Chezet
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Laetitia Delort
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| |
Collapse
|
37
|
Cardoso Dal Pont G, Lee A, Bortoluzzi C, Farnell YZ, Gougoulias C, Kogut MH. Novel model for chronic intestinal inflammation in chickens: (2) Immunologic mechanism behind the inflammatory response. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 138:104524. [PMID: 36067905 DOI: 10.1016/j.dci.2022.104524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/25/2022] [Accepted: 08/25/2022] [Indexed: 06/15/2023]
Abstract
Intestinal inflammation in poultry is a complex response that involves immune and intestinal cells which is still not fully understood. Thus, to better understand the mechanisms that drive the chronic intestinal inflammation in fowl we conducted an experiment applying a previously established nutritional model of low-grade chronic intestinal inflammation to evaluate cytokine and chemokine profiles in the chicken intestine. For this, we placed 90 one-day chickens into two treatments: (1) a control group (CNT) fed a corn-soybean diet, and (2) a group fed a diet high in non-starch polysaccharides (NSP). At days 14, 22, 28 and 36 of age, 6 birds from each treatment were euthanized, jejunal and ileal samples were collected for histological examination and cytokine measurements. The cytokines interferon-alpha (IFN-α), IFN-γ, interleukin-16 (IL-16), IL-10, IL-21, IL-6, macrophage-colony stimulating factor (M-CSF), chemokine C-C motif ligand 20 (CCL20), CCL4, CCL5 and vascular endothelial growth factor (VEGF) were quantified in the intestinal tissue. Histologically, both jejunum and ileum of broilers fed NSP diet showed marked infiltration of mononuclear immune cells into the villi. Further, these birds exhibited a significant (P < 0.05) increase in CCL20 concentration in the jejunum at 14d, but a dramatic reduction of M-CSF at 14 and 21d. Later at 28d and 36d, birds fed the NSP diet exhibited increased IL-16 concentration in the jejunum. Since M-CSF is a monocyte stimulatory cytokine and CCL20 a chemokine of T-cells, the reduced M-CSF and increased production of CCL20 may indicate the involvement of the adaptive immune response, specifically driven by T-cells, occurring around the third week of age in the NSP model. Lastly, as a result of the mononuclear cell infiltration and activation of T-cells, IL-16, a pro-inflammatory T-cell cytokine, increased. Therefore, the current work indicates the importance of adaptive immune cells, especially T-cells, in the chronic intestinal inflammation in broiler chicken.
Collapse
Affiliation(s)
- Gabriela Cardoso Dal Pont
- Department of Poultry Science, Texas A&M AgriLife Research, Texas A&M University, College Station, TX, USA.
| | - A Lee
- Department of Poultry Science, Texas A&M AgriLife Research, Texas A&M University, College Station, TX, USA
| | - C Bortoluzzi
- Department of Poultry Science, Texas A&M AgriLife Research, Texas A&M University, College Station, TX, USA
| | - Y Z Farnell
- Department of Poultry Science, Texas A&M AgriLife Research, Texas A&M University, College Station, TX, USA
| | - C Gougoulias
- Innovad NV/SA, Postbaan 69, 2910, Essen, Belgium
| | - M H Kogut
- USDA-ARS, Southern Plains Agricultural Research Center, College Station, TX, USA.
| |
Collapse
|
38
|
Yang S, Zhao M, Jia S. Macrophage: Key player in the pathogenesis of autoimmune diseases. Front Immunol 2023; 14:1080310. [PMID: 36865559 PMCID: PMC9974150 DOI: 10.3389/fimmu.2023.1080310] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/09/2023] [Indexed: 02/16/2023] Open
Abstract
The macrophage is an essential part of the innate immune system and also serves as the bridge between innate immunity and adaptive immune response. As the initiator and executor of the adaptive immune response, macrophage plays an important role in various physiological processes such as immune tolerance, fibrosis, inflammatory response, angiogenesis and phagocytosis of apoptotic cells. Consequently, macrophage dysfunction is a vital cause of the occurrence and development of autoimmune diseases. In this review, we mainly discuss the functions of macrophages in autoimmune diseases, especially in systemic lupus erythematosus (SLE), rheumatic arthritis (RA), systemic sclerosis (SSc) and type 1 diabetes (T1D), providing references for the treatment and prevention of autoimmune diseases.
Collapse
Affiliation(s)
- Shuang Yang
- Dapartment of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ming Zhao
- Dapartment of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.,Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| | - Sujie Jia
- Department of Pharmacy, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| |
Collapse
|
39
|
Depletion of regulatory T cells exacerbates inflammatory responses after chronic cerebral hypoperfusion in mice. Mol Cell Neurosci 2022; 123:103788. [PMID: 36302461 DOI: 10.1016/j.mcn.2022.103788] [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: 05/11/2022] [Revised: 10/04/2022] [Accepted: 10/17/2022] [Indexed: 11/27/2022] Open
Abstract
Vascular cognitive impairment is the second most common cause of dementia which can be induced by chronic cerebral hypoperfusion. Regulatory T cells (Tregs) have been proven to provide beneficial effects in several central nervous system (CNS) diseases, but the roles of Tregs in chronic cerebral hypoperfusion-induced white matter damage have not been explored. In this study, Foxp3-diphtheria toxin receptor (DTR) mice treated with diphtheria toxin (DT) and wild type C57BL/6 mice treated with anti-CD25 antibody were subjected to bilateral carotid artery stenosis (BCAS). Flow cytometry analysis showed Tregs were widely distributed in spleen whereas barely distributed in brain under normal conditions. The distribution of lymphocytes and Tregs did not change significantly in spleen and brain after BCAS. Depletion of Tregs decreased the numbers of mature oligodendrocytes and anti-inflammatory microglia at 14 days and 28 days following BCAS. And pro-inflammatory cytokines including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6) and interferon-γ (IFN-γ) showed higher expression after Tregs depletion. In contrast, Tregs depletion did not change the overall severity of white matter injury as shown by the expression of myelin-associated glycoprotein (MAG), myelin basic protein (MBP), luxol fast blue (LFB) staining and electron microscopy assay. Moreover, Tregs depletion had marginal effect on cognition defects after BCAS revealed by Morris water maze and novel object recognition examination at 28 days after BCAS. In summary, our results suggest an anti-inflammatory role of Tregs with marginal effects on white matter damage in mice after BCAS-induced chronic cerebral hypoperfusion.
Collapse
|
40
|
Chen Z, Lan H, Liao Z, Huang J, Jian X, Hu J, Liao H. Regulatory T cells-centered regulatory networks of skeletal muscle inflammation and regeneration. Cell Biosci 2022; 12:112. [PMID: 35869487 PMCID: PMC9308315 DOI: 10.1186/s13578-022-00847-x] [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: 09/27/2021] [Accepted: 07/08/2022] [Indexed: 11/10/2022] Open
Abstract
As the understanding of skeletal muscle inflammation is increasingly clarified, the role of Treg cells in the treatment of skeletal muscle diseases has attracted more attention in recent years. A consensus has been reached that the regulation of Treg cells is the key to completing the switch of inflammation and repair of skeletal muscle, whose presence directly determine the repairing quality of the injured skeletal muscle. However, the functioning process of Treg cells remains unreported, thereby making it necessary to summarize the current role of Treg cells in skeletal muscle. In this review, the characteristics, origins, and cellular kinetics of these Treg cells are firstly described; Then, the relationship between Treg cells and muscle satellite cells (MuSCs), conventional T cells (Tconv) is discussed (the former is involved in the entire repair and regeneration process, while the latter matters considerably in causing most skeletal muscle autoimmune diseases); Next, focus is placed on the control of Treg cells on the phenotypic switch of macrophages, which is the key to the switch of inflammation; Finally, factors regulating the functional process of Treg cells are analyzed, and a regulatory network centered on Treg cells is summarized. The present study summarizes the cell-mediated interactions in skeletal muscle repair over the past decade, and elucidates the central role of regulatory T cells in this process, so that other researchers can more quickly and comprehensively understand the development and direction of this very field. It is believed that the hereby proposed viewpoints and problems can provide fresh visions for the latecomers.
Collapse
|
41
|
Hao Y, Zhang W, Qin J, Tan L, Luo Y, Chen H. Biological Cardiac Patch Based on Extracellular Vesicles and Extracellular Matrix for Regulating Injury-Related Microenvironment and Promoting Cardiac Tissue Recovery. ACS APPLIED BIO MATERIALS 2022; 5:5218-5230. [PMID: 36265007 DOI: 10.1021/acsabm.2c00659] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cardiac patches are widely investigated to repair or regenerate diseased and aging cardiac tissues. While numerous studies looked into engineering the biochemical/biomechanical/cellular microenvironment and components in the heart tissue, the changes induced by cardiac patches and how they should be controlled to promote cardiac tissue repair/regeneration remains an important yet untapped direction, especially immunological responses. In this study, we designed and fabricated a bilaminated cardiac patch based on extracellular matrix (ECM) materials loaded with the extracellular vesicles (EVs) derived from mesenchymal stromal cells. The function of the biological material to modulate the injury-related microenvironment in a cardiac infarction model in mice was investigated. The study showed that the treatment of EV-ECM patches to the infarcted area increased the level of immunomodulatory major histocompatibility complex class IIlo macrophages in the early stage of myocardial injury to mitigate excessive inflammatory responses due to injury. The intensity of the acquired proinflammatory immune response in systemic immune organs was reduced. Further analyses indicated that the EV-ECM patches exhibited proangiogenic functions and decreased the infarct size with improved cardiac recovery in mice. The study provided insights into shaping the injury-related microenvironment through the incorporation of extracellular vesicles into cardiac patches, and the EV-ECM material is a promising design paradigm to improve the function of cardiac patches to treat myocardial injuries and diseases.
Collapse
Affiliation(s)
- Yaoyao Hao
- Department of Biomedical Engineering, College of Engineering, Peking University, 5 Yiheyuan Road, Haidian District, Beijing 100871, China.,College of Future Technology, Peking University, 5 Yiheyuan Road, Haidian District, Beijing 100871, China
| | - Wei Zhang
- Guangzhou Keyue Biotech Co., Ltd., 6 Xin-Rui Road, Luogang District, Guangzhou 510535, China
| | - Jiahang Qin
- Department of Biomedical Engineering, College of Engineering, Peking University, 5 Yiheyuan Road, Haidian District, Beijing 100871, China.,College of Future Technology, Peking University, 5 Yiheyuan Road, Haidian District, Beijing 100871, China
| | - Lindan Tan
- Department of Biomedical Engineering, College of Engineering, Peking University, 5 Yiheyuan Road, Haidian District, Beijing 100871, China.,College of Future Technology, Peking University, 5 Yiheyuan Road, Haidian District, Beijing 100871, China
| | - Ying Luo
- Department of Biomedical Engineering, College of Engineering, Peking University, 5 Yiheyuan Road, Haidian District, Beijing 100871, China
| | - Haifeng Chen
- Department of Biomedical Engineering, College of Engineering, Peking University, 5 Yiheyuan Road, Haidian District, Beijing 100871, China.,College of Future Technology, Peking University, 5 Yiheyuan Road, Haidian District, Beijing 100871, China
| |
Collapse
|
42
|
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
|
43
|
Kharraz Y, Lukesova V, Serrano AL, Davison A, Muñoz-Cánoves P. Full spectrum cytometry improves the resolution of highly autofluorescent biological samples: Identification of myeloid cells in regenerating skeletal muscles. Cytometry A 2022; 101:862-876. [PMID: 35608022 DOI: 10.1002/cyto.a.24568] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 03/30/2022] [Accepted: 04/22/2022] [Indexed: 01/27/2023]
Abstract
Autofluorescence (AF) is an intrinsic characteristic of cells caused by the presence of fluorescent biological compounds within the cell; these can include structural proteins (e.g., collagen and elastin), cellular organelles, and metabolites (e.g., aromatic amino acids). In flow cytometric studies, the presence of AF can lead to reduced antigen and population resolution, as well as the presence of artifacts due to false positive events. Here, we describe a methodology that uses the inherent ability of full spectrum cytometry to treat AF as a fluorochrome and to thereby separate it from the other fluorochromes of the assay. This method can be applied to complex inflamed tissues; for instance, in regenerating skeletal muscle we have developed a 16-color panel targeting highly autofluorescent myeloid cells. This represents a first step toward overcoming technological limitations in flow cytometry due to AF.
Collapse
Affiliation(s)
- Yacine Kharraz
- Application Department, Cytek Biosciences, Inc., Fremont, California, USA
| | - Vera Lukesova
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Antonio L Serrano
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Adam Davison
- Application Department, Cytek Biosciences, Inc., Fremont, California, USA
| | - Pura Muñoz-Cánoves
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.,Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,ICREA, Barcelona, Spain
| |
Collapse
|
44
|
Liang Z, Zhang T, Liu H, Li Z, Peng L, Wang C, Wang T. Inflammaging: The ground for sarcopenia? Exp Gerontol 2022; 168:111931. [PMID: 35985553 DOI: 10.1016/j.exger.2022.111931] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/02/2022] [Accepted: 08/14/2022] [Indexed: 12/15/2022]
Abstract
Sarcopenia is a progressive skeletal muscle disease that occurs most commonly in the elderly population, contributing to increased costs and hospitalization. Exercise and nutritional therapy have been proven to be effective for sarcopenia, and some drugs can also alleviate declines in muscle mass and function due to sarcopenia. However, there is no specific pharmacological treatment for sarcopenia at present. This review will mainly discuss the relationship between inflammaging and sarcopenia. The increased secretion of proinflammatory cytokines with aging may be because of cellular senescence, immunosenescence, alterations in adipose tissue, damage-associated molecular patterns (DAMPs), and gut microbes due to aging. These sources of inflammaging can impact the sarcopenia process through direct or indirect pathways. Conversely, sarcopenia can also aggravate the process of inflammaging, creating a vicious cycle. Targeting sources of inflammaging can influence muscle function, which could be considered a therapeutic target for sarcopenia. Moreover, not only proinflammatory cytokines but also anti-inflammatory cytokines can influence muscle and inflammation and participate in the progression of sarcopenia. This review focuses on the effects of TNF-α, IL-6, and IL-10, which can be detected in plasma. Therefore, clearing chronic inflammation by targeting proinflammatory cytokines (TNF-α, IL-1, IL-6) and the inflammatory pathway (JAK/STAT, autophagy, NF-κB) may be effective in treating sarcopenia.
Collapse
Affiliation(s)
- Zejun Liang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Tianxiao Zhang
- School of Healthcare Sciences, Cardiff University, Health Park, CF14 4XN Wales, UK
| | - Honghong Liu
- West China School of Nursing/West China Hospital, Sichuan University, NO.37 Alley, Chengdu 610041, Sichuan, PR China
| | - Zhenlin Li
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Lihong Peng
- Department of Rehabilitation Medicine, West China Second University Hospital, Sichuan University, PR China
| | - Changyi Wang
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Tiantian Wang
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
45
|
Regulatory T cells in skeletal muscle repair and regeneration: recent insights. Cell Death Dis 2022; 13:680. [PMID: 35931697 PMCID: PMC9356005 DOI: 10.1038/s41419-022-05142-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 01/21/2023]
Abstract
Skeletal muscle repair and regeneration after injury is a multi-stage process, involving a dynamic inflammatory microenvironment consisting of a complex network formed by the interaction of immune cells and their secreted cytokines. The homeostasis of the inflammatory microenvironment determines whether skeletal muscle repair tissues will ultimately form scar tissue or regenerative tissue. Regulatory T cells (Tregs) regulate homeostasis within the immune system and self-immune tolerance, and play a crucial role in skeletal muscle repair and regeneration. Dysregulated Tregs function leads to abnormal repair. In this review, we discuss the role and mechanisms of Tregs in skeletal muscle repair and regeneration after injury and provide new strategies for Treg immunotherapy in skeletal muscle diseases.
Collapse
|
46
|
Role of Regulatory T Cells in Skeletal Muscle Regeneration: A Systematic Review. Biomolecules 2022; 12:biom12060817. [PMID: 35740942 PMCID: PMC9220893 DOI: 10.3390/biom12060817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/08/2022] [Accepted: 03/11/2022] [Indexed: 02/06/2023] Open
Abstract
Muscle injuries are frequent in individuals with genetic myopathies and in athletes. Skeletal muscle regeneration depends on the activation and differentiation of satellite cells present in the basal lamina of muscle fibers. The skeletal muscle environment is critical for repair, metabolic and homeostatic function. Regulatory T cells (Treg) residing within skeletal muscle comprise a distinct and special cell population that modifies the inflammatory environment by secreting cytokines and amphiregulin, an epidermal growth factor receptor (EGFR) ligand that acts directly upon satellite cells, promoting tissue regeneration. This systematic review summarizes the current knowledge regarding the role of Treg in muscle repair and discusses their therapeutic potential in skeletal muscle injuries. A bibliographic search was carried out using the terms Treg and muscle regeneration and repair, covering all articles up to April 2021 indexed in the PubMed and EMBASE databases. The search included only published original research in human and experimental animal models, with further data analysis based on the PICO methodology, following PRISMA definitions and Cochrane guidelines.
Collapse
|
47
|
Terry J, Bedaiwy MA. Placental interferon signaling is involved in chronic intervillositis of unknown etiology. Placenta 2022; 124:5-11. [DOI: 10.1016/j.placenta.2022.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/02/2022] [Accepted: 05/10/2022] [Indexed: 11/29/2022]
|
48
|
Shang L, Shao J, Ge S. Immunomodulatory Properties: The Accelerant of Hydroxyapatite-Based Materials for Bone Regeneration. Tissue Eng Part C Methods 2022; 28:377-392. [PMID: 35196904 DOI: 10.1089/ten.tec.2022.00111112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The immunoinflammatory response is the prerequisite step for wound healing and tissue regeneration, and the immunomodulatory effects of biomaterials have attracted increasing attention. Hydroxyapatite [Ca10(PO4)6(OH)2] (HAp), a common calcium phosphate ceramic, due to its structural and functional similarity to the inorganic constituent of natural bones, has been developed for different application purposes such as bone substitutes, tissue engineering scaffolds, and implant coatings. Recently, the interaction between HAp-based materials and the immune system (various immune cells), and the immunomodulatory effects of HAp-based materials on bone tissue regeneration have been explored extensively. Macrophages-mediated regenerative effect by HAp stimulation occupies the mainstream status of immunomodulatory strategies. The immunomodulation of HAp can be manipulated by tuning the physical, chemical, and biological cues such as surface functionalization (physical or chemical modifications), structural and textural characteristics (size, shape, and surface topography), and the incorporation of bioactive substances (cytokines, rare-earth elements, and bioactive ions). Therefore, HAp ceramic materials can contribute to bone regeneration by creating a favorable osteoimmune microenvironment, which would provide a more comprehensive theoretical basis for their further clinical applications. Considering the rapidly developed HAp-based materials as well as their excellent biological performances in the field of regenerative medicine, this review discusses the recent advances concerning the immunomodulatory methods for HAp-based biomaterials and their roles in bone tissue regeneration.
Collapse
Affiliation(s)
- Lingling Shang
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Jinlong Shao
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Shaohua Ge
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| |
Collapse
|
49
|
Larouche JA, Fraczek PM, Kurpiers SJ, Yang BA, Davis C, Castor-Macias JA, Sabin K, Anderson S, Harrer J, Hall M, Brooks SV, Jang YC, Willett N, Shea LD, Aguilar CA. Neutrophil and natural killer cell imbalances prevent muscle stem cell-mediated regeneration following murine volumetric muscle loss. Proc Natl Acad Sci U S A 2022; 119:e2111445119. [PMID: 35377804 PMCID: PMC9169656 DOI: 10.1073/pnas.2111445119] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 02/09/2022] [Indexed: 02/06/2023] Open
Abstract
Volumetric muscle loss (VML) overwhelms the innate regenerative capacity of mammalian skeletal muscle (SkM), leading to numerous disabilities and reduced quality of life. Immune cells are critical responders to muscle injury and guide tissue resident stem cell– and progenitor-mediated myogenic repair. However, how immune cell infiltration and intercellular communication networks with muscle stem cells are altered following VML and drive pathological outcomes remains underexplored. Herein, we contrast the cellular and molecular mechanisms of VML injuries that result in the fibrotic degeneration or regeneration of SkM. Following degenerative VML injuries, we observed the heightened infiltration of natural killer (NK) cells as well as the persistence of neutrophils beyond 2 wk postinjury. Functional validation of NK cells revealed an antagonistic role in neutrophil accumulation in part via inducing apoptosis and CCR1-mediated chemotaxis. The persistent infiltration of neutrophils in degenerative VML injuries was found to contribute to impairments in muscle stem cell regenerative function, which was also attenuated by transforming growth factor beta 1 (TGFβ1). Blocking TGFβ signaling reduced neutrophil accumulation and fibrosis and improved muscle-specific force. Collectively, these results enhance our understanding of immune cell–stem cell cross talk that drives regenerative dysfunction and provide further insight into possible avenues for fibrotic therapy exploration.
Collapse
Affiliation(s)
- Jacqueline A. Larouche
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109
| | - Paula M. Fraczek
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109
| | - Sarah J. Kurpiers
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
| | - Benjamin A. Yang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109
| | - Carol Davis
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109
| | - Jesus A. Castor-Macias
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109
| | - Kaitlyn Sabin
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109
| | - Shannon Anderson
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332
| | - Julia Harrer
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332
| | - Matthew Hall
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
| | - Susan V. Brooks
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109
| | - Young C. Jang
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332
| | - Nick Willett
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR 97403
| | - Lonnie D. Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
| | - Carlos A. Aguilar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
50
|
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
|