1
|
Li Z, Si P, Meng T, Zhao X, Zhu C, Zhang D, Meng S, Li N, Liu R, Ni T, Yan J, Li H, Zhao N, Zhong C, Qin Y, Chen W, Chen ZJ, Jiao X. CCR8 + decidual regulatory T cells maintain maternal-fetal immune tolerance during early pregnancy. Sci Immunol 2025; 10:eado2463. [PMID: 40249828 DOI: 10.1126/sciimmunol.ado2463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 12/13/2024] [Accepted: 02/18/2025] [Indexed: 04/20/2025]
Abstract
Regulatory T (Treg) cells play a vital role in maintaining maternal immune tolerance to the semiallogeneic fetus during pregnancy. Treg cell population heterogeneity and tissue-specific functions in the human decidua remain largely unknown. Here, using single-cell transcriptomic and T cell receptor sequencing of human CD4+ T cells from first-trimester deciduae and matched peripheral blood of pregnant women, we identified a highly activated, immunosuppressive CCR8+ Treg cell subset specifically enriched in the decidua (dTreg cells). CCR8+ dTreg cells were decreased in patients with recurrent pregnancy loss (RPL) and an abortion-prone mouse model. Depletion of CCR8+ dTreg cells increased susceptibility to fetal loss, with altered decidual immune profiles. Adoptive transfer of CCR8+ Treg cells rescued fetal loss in abortion-prone mice. The CCR8 ligand CCL1 was mainly produced by decidual CD49a+ natural killer cells and was significantly decreased in patients with RPL. Our data demonstrate that CCR8+ dTreg cells are required to maintain maternal-fetal tolerance and highlight potential avenues for RPL therapies.
Collapse
Affiliation(s)
- Zhuqing Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
- Department of Reproductive Medicine, Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Pinxin Si
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| | - Tingting Meng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| | - Xiaoran Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| | - Chendi Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| | - Dunfang Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shutong Meng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| | - Nianyu Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| | - Ran Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| | - Tianxiang Ni
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| | - Junhao Yan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| | - Hongchang Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| | - Ning Zhao
- Analytical Biosciences Limited, Beijing 100191, China
| | - Chao Zhong
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing 100191, China
| | - Yingying Qin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
- Department of Reproductive Medicine, Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - WanJun Chen
- Mucosal Immunology Section, National Institutes of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zi-Jiang Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
- Department of Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xue Jiao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| |
Collapse
|
2
|
Ma S, Lu Y, Sui S, Yang JS, Fu BB, Tan PX, Chai Y, Lv J, Kong L, Wu X, Gao YB, Yan T. Unraveling the triad of immunotherapy, tumor microenvironment, and skeletal muscle biomechanics in oncology. Front Immunol 2025; 16:1572821. [PMID: 40242775 PMCID: PMC12000078 DOI: 10.3389/fimmu.2025.1572821] [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: 02/07/2025] [Accepted: 03/03/2025] [Indexed: 04/18/2025] Open
Abstract
The intricate interaction between skeletal muscle biomechanics, the tumor microenvironment, and immunotherapy constitutes a pivotal research focus oncology. This work provides a comprehensive review of methodologies for evaluating skeletal muscle biomechanics, including handheld dynamometry, advanced imaging techniques, electrical impedance myography, elastography, and single-fiber experiments to assess muscle quality and performance. Furthermore, it elucidates the mechanisms, applications, and limitations of various immunotherapy modalities, including immune checkpoint inhibitors, adoptive cell therapy, cancer vaccines, and combined chemoimmunotherapy, while examining their effects on skeletal muscle function and systemic immune responses. Key findings indicate that although immunotherapy is effective in augmenting antitumor immunity, it frequently induces muscle-related adverse effects such as weakness, fatigue, or damage, primarily mediated by cytokine release and immune activation. This work underscores the significance of immune niches within the tumor microenvironment in influencing treatment outcomes and proposes strategies to optimize therapy through personalized regimens and combinatorial approaches. This review highlights the need for further research on the formation of immune niches and interactions muscle-tumor. Our work is crucial for advancing the efficacy of immunotherapy, reducing adverse effects, and ultimately improving survival rates and quality of life of patients with cancer.
Collapse
Affiliation(s)
- Shuang Ma
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Ying Lu
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Shang Sui
- St. John’s Kilmarnock School, Breslau, ON, Canada
| | - Jia-shuo Yang
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bing-bing Fu
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pei-xin Tan
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yicheng Chai
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Jiaqi Lv
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Lingyu Kong
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Xiaolin Wu
- School of Mathematics and Statistics, Liaoning University, Shenyang, China
| | - Yi-bo Gao
- Department of Oral and Maxillofacial Surgery, Taikang Bybo Dental, Beijing, China
| | - Tao Yan
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
3
|
Sadeghi M, Moghaddam A, Amiri AM, Charoghdoozi K, Mohammadi M, Dehnavi S, Orazizadeh M. Improving the Wound Healing Process: Pivotal role of Mesenchymal stromal/stem Cells and Immune Cells. Stem Cell Rev Rep 2025; 21:680-697. [PMID: 39921839 DOI: 10.1007/s12015-025-10849-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2025] [Indexed: 02/10/2025]
Abstract
Wound healing, a physiological process, involves several different types of cells, from immune cells to non-immune cells, including mesenchymal stromal/stem cells (MSC), and their interactions. Immune cells including macrophages, neutrophils, dendritic cells (DC), innate lymphoid cells (ILC), natural killer (NK) cells, and B and T lymphocytes participate in wound healing by secreting various mediators and interacting with other cells. MSCs, as self-renewing, fast proliferating, and multipotent stromal/stem cells, are found in a wide variety of tissues and critically involved in different phases of wound healing by secreting various molecules that help to improve tissue healing and regeneration. In this review, first, we described the four main phases of wound healing, second, we reviewed the function of MSCs, MSC secretome and immune cells in improving the progress of wound repair (mainly focusing on skin wound healing), third, we explained the immune cells/MSCs interactions in the process of wound healing and regeneration, and finally, we introduce clinical applications of MSCs to improve the process of wound healing.
Collapse
Affiliation(s)
- Mahvash Sadeghi
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Asma Moghaddam
- Cellular and Molecular Research Center, Medical Basic Sciences Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Anatomical Sciences, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Amir Mohammad Amiri
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Kianush Charoghdoozi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojgan Mohammadi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sajad Dehnavi
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mahmoud Orazizadeh
- Cellular and Molecular Research Center, Medical Basic Sciences Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Department of Anatomical Sciences, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
4
|
Chen YJ, Yen CL, Lee CH, Liao KC, Fang JT, Tsai TS, Chen YC, Lin CY. Prognostic potential of neutrophil-to-lymphocyte ratio for appendicular skeletal muscle mass reduction in males aged 70 and older. Aging (Albany NY) 2025; 17:863-871. [PMID: 40053874 PMCID: PMC11984423 DOI: 10.18632/aging.206217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 01/10/2025] [Indexed: 03/09/2025]
Abstract
Inflammation plays a pivotal role in the age-related decline of skeletal muscle mass, leading to sarcopenia in the elderly. The prevalence of sarcopenia notably increases among males aged ≥ 70. However, it remains unclear whether inflammatory indexes are associated with the reduction in skeletal muscle mass in the elderly population. Thirty-one males aged ≥ 70, without severe diseases or dementia, were enrolled in the study. They underwent muscle mass measurements, physical measurements, and hematological tests at the onset of the study and after a one-year follow-up. Twenty-eight participants were successfully followed for one year. Appendicular skeletal muscle mass index (ASMI) decreased by 3.30 ± 2.41% in 14 participants and increased by 2.66 ± 1.61% in the other 14 participants compared to baseline levels. The baseline neutrophil-to-lymphocyte ratio (NLR) was 2.14 ± 0.56 in the ASMI-decreased group and 1.66 ± 0.62 in the ASMI-increased group. A statistically significant negative correlation was found between baseline NLR and the change in ASMI in linear regression analyses. The area under the curve (AUC) of the baseline NLR for predicting ASMI decline was 0.724, with an optimal sensitivity of 64.3% and specificity of 78.6% at a cut-off value of 1.94. NLR emerged as a potential prognostic marker for ASMI reduction in elderly males. However, further studies are necessary to assess its clinical utility.
Collapse
Affiliation(s)
- Ying-Jen Chen
- Geriatric Medical Center, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Geriatrics and General Internal Medicine, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chieh-Li Yen
- Geriatric Medical Center, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Geriatrics and General Internal Medicine, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chern-Horng Lee
- Geriatric Medical Center, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Geriatrics and General Internal Medicine, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kuo-Chen Liao
- Geriatric Medical Center, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Geriatrics and General Internal Medicine, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ji-Tseng Fang
- Department of Nephrology, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tz-Shiu Tsai
- Geriatric Medical Center, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Geriatrics and General Internal Medicine, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yi-Ching Chen
- Department of Geriatrics and General Internal Medicine, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chun-Yen Lin
- Division of Hepatology, Department of Gastroenterology and Hepatology, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
5
|
Zhang P, Wang J, Miao J, Zhu P. The dual role of tissue regulatory T cells in tissue repair: return to homeostasis or fibrosis. Front Immunol 2025; 16:1560578. [PMID: 40114929 PMCID: PMC11922884 DOI: 10.3389/fimmu.2025.1560578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 02/18/2025] [Indexed: 03/22/2025] Open
Abstract
Tissue resident regulatory T cells (tissue Tregs) are vital for maintaining immune homeostasis and controlling inflammation. They aid in repairing damaged tissues and influencing the progression of fibrosis. However, despite extensive research on how tissue Tregs interact with immune and non-immune cells during tissue repair, their pro- and anti-fibrotic effects in chronic tissue injury remain unclear. Understanding how tissue Tregs interact with various cell types, as well as their roles in chronic injury and fibrosis, is crucial for uncovering the mechanisms behind these conditions. In this review, we describe the roles of tissue Tregs in repair and fibrosis across different tissues and explore potential strategies for regulating tissue homeostasis. These insights hold promise for providing new perspectives and approaches for the treatment of irreversible fibrotic diseases.
Collapse
Affiliation(s)
- Peiyan Zhang
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jiawei Wang
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jinlin Miao
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ping Zhu
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
6
|
Sun J, Chen X, Ruan Y, Xu J, Xu H. MEF2A promoter methylation negatively regulates mRNA transcription and affects myoblast physiological function in cattle. Genomics 2025; 117:111016. [PMID: 40024578 DOI: 10.1016/j.ygeno.2025.111016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/27/2025] [Accepted: 02/17/2025] [Indexed: 03/04/2025]
Abstract
This study investigates the regulatory effects of methylation in the promoter region of the bovine MEF2A gene on its transcription levels and the impact on bovine myoblasts. Transcription levels and promoter methylation status of MEF2A in the same tissues of calves and adult cattle were assessed using qRT-PCR and BSP methods. The results indicated that MEF2A expression levels in calves were significantly lower than those in adult cattle (P < 0.05), while the methylation rate of MEF2A was significantly higher in calves (P < 0.05), suggesting a correlation between high methylation levels and reduced gene expression. Subsequently, MEF2A overexpression and interference vectors were transfected into bovine myoblasts to examine the effects of altered MEF2A expression on its promoter methylation status. The findings revealed that MEF2A overexpression significantly reduced the methylation rate (P < 0.01), whereas MEF2A interference increased the methylation rate (P < 0.01), aligning with the expression trends of DNMT1. Furthermore, bovine myoblasts were treated with varying concentrations of the methylation inhibitor 5-Aza-dC to evaluate changes in MEF2A promoter methylation and mRNA levels. The effects on cell cycle progression, apoptosis, and other growth parameters were assessed using flow cytometry, ELISA, and qRT-PCR. Results showed that a concentration of 1 μM 5-Aza-dC effectively reduced MEF2A promoter methylation and significantly upregulated MEF2A expression, leading to accelerated cell cycle progression and increased secretion levels of GH and INS, all differences being statistically significant (P < 0.01). Additionally, 1 μM of 5-Aza-dC promoted apoptosis, with qRT-PCR results for relevant genes supporting this finding. In conclusion, methylation of the MEF2A promoter negatively regulates its mRNA transcription levels, thereby impacting the growth and development of Guanling cattle myoblasts. These results provide valuable insights for the genetic improvement of cattle through marker-assisted selection.
Collapse
Affiliation(s)
- Jinkui Sun
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, PR China; College of Animal Science, Guizhou University, Guiyang 550025, PR China
| | - Xiang Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, PR China; College of Animal Science, Guizhou University, Guiyang 550025, PR China
| | - Yong Ruan
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, PR China; College of Animal Science, Guizhou University, Guiyang 550025, PR China
| | - Jiali Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, PR China; College of Animal Science, Guizhou University, Guiyang 550025, PR China
| | - Houqiang Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, PR China; College of Animal Science, Guizhou University, Guiyang 550025, PR China.
| |
Collapse
|
7
|
Yao Y, Luo Y, Liang X, Zhong L, Wang Y, Hong Z, Song C, Xu Z, Wang J, Zhang M. The role of oxidative stress-mediated fibro-adipogenic progenitor senescence in skeletal muscle regeneration and repair. Stem Cell Res Ther 2025; 16:104. [PMID: 40025535 PMCID: PMC11872320 DOI: 10.1186/s13287-025-04242-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 02/18/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Stem cells play a pivotal role in tissue regeneration and repair. Skeletal muscle comprises two main stem cells: muscle stem cells (MuSCs) and fibro-adipogenic progenitors (FAPs). FAPs are essential for maintaining the regenerative milieu of muscle tissue and modulating the activation of muscle satellite cells. However, during acute skeletal muscle injury, the alterations and mechanisms of action of FAPs remain unclear. METHODS we employed the GEO database for bioinformatics analysis of skeletal muscle injury. A skeletal muscle injury model was established through cardiotoxin (CTX, 10µM, 50µL) injection into the tibialis anterior (TA) of C57BL/6 mice. Three days post-injury, we extracted the TA, isolated FAPs (CD31-CD45-PDGFRα+Sca-1+), and assessed the senescence phenotype through SA-β-Gal staining and Western blot. Additionally, we established a co-culture system to evaluate the capacity of FAPs to facilitate MuSCs differentiation. Finally, we alleviated the senescent of FAPs through in vitro (100 µM melatonin, 5 days) and in vivo (20 mg/kg/day melatonin, 15 days) administration experiments, confirming melatonin's pivotal role in the regeneration and repair processes of skeletal muscle. RESULTS In single-cell RNA sequencing analysis, we discovered the upregulation of senescence-related pathways in FAPs following injury. Immunofluorescence staining revealed the co-localization of FAPs and senescent markers in injured muscles. We established the CTX injury model and observed a reduction in the number of FAPs post-injury, accompanied by the manifestation of a senescent phenotype. Melatonin treatment was found to attenuate the injury-induced senescence of FAPs. Further co-culture experiments revealed that melatonin facilitated the restoration of FAPs' capacity to promote myoblast differentiation. Through GO and KEGG analysis, we found that the administration of melatonin led to the upregulation of AMPK pathway in FAPs, a pathway associated with antioxidant stress response. Finally, drug administration experiments corroborated that melatonin enhances skeletal muscle regeneration and repair by alleviating FAP senescence in vivo. CONCLUSION In this study, we first found FAPs underwent senescence and redox homeostasis imbalance after injury. Next, we utilized melatonin to enhance FAPs regenerative and repair capabilities by activating AMPK signaling pathway. Taken together, this work provides a novel theoretical foundation for treating skeletal muscle injury.
Collapse
Affiliation(s)
- Yuqing Yao
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Yusheng Luo
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xiaomei Liang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Li Zhong
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yannan Wang
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-Sen University, Shenzhen, China
| | - Zhengchao Hong
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Chao Song
- School of Electronics and Communication Engineering, Shenzhen Campus of Sun Yat-sen University, Sun Yat-Sen University, Shenzhen, China
| | - Zeyu Xu
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-Sen University, Shenzhen, China
| | - Jiancheng Wang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| | - Miao Zhang
- Department of Physical Education, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
8
|
Soussi S, Maione AS, Lefèvre L, Pizzinat N, Iacovoni J, Gonzalez-Fuentes I, Cussac D, Iengo L, Santin Y, Tundo F, Tondo C, Pompilio G, Parini A, Douin-Echinard V, Sommariva E. Analysis of effector/memory regulatory T cells from arrhythmogenic cardiomyopathy patients identified IL-32 as a novel player in ACM pathogenesis. Cell Death Dis 2025; 16:87. [PMID: 39934117 DOI: 10.1038/s41419-025-07364-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/23/2024] [Accepted: 01/16/2025] [Indexed: 02/13/2025]
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited cardiac disorder that causes sudden cardiac death and progressive heart failure. Besides fibro-fatty replacement and myocyte degenerative changes, inflammatory patchy infiltrates are found in myocardial histological analysis of ACM patients. Inflammatory cells could actively participate in ACM pathogenesis, contributing to the alteration of cardiac microenvironment homeostasis, thus triggering disease evolution. In order to characterize the immune-derived mediators involved in ACM pathogenesis, peripheral blood mononuclear cells from ACM patients were characterized and compared to healthy controls' ones. Flow cytometry analysis revealed a lower frequency of CD4+ T helper type 1 cells, NK cells, and terminally differentiated CD8+ EMRA+ T cells in ACM patients compared to age-matched controls. In contrast, a higher proportion of effector/memory FOXP3+ CCR4+ CD45RO+ regulatory CD4+ T cells (Treg) were found in ACM patients. Single-cell RNA-seq performed on isolated memory Treg cells (mTreg) from ACM patients and healthy controls identified 6 clusters characterized by specific gene signatures related to tissue repair and immunosuppressive pathways. Notably, interleukin 32 (IL-32) was the most differentially expressed gene in ACM patients mTreg with respect to healthy controls. Treatment of human cardiac mesenchymal stromal cells with recombinant IL-32 in vitro promoted lipid droplet accumulation and collagen deposition, thus identifying IL-32 as a new potential player in the immune-mediated trigger of cardiac fibro-fatty replacement in ACM. Overall, we here provide the first complete characterization of circulating ACM immune cells, revealing an abundance of Treg. The high expression of IL-32 in ACM Treg may contribute to accelerated cardiac remodeling in ACM patients' hearts.
Collapse
Affiliation(s)
| | - Angela Serena Maione
- Unit of Inherited Cardiomyopathies, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
| | - Lise Lefèvre
- I2MC, INSERM, UMR-1297, Toulouse, France
- RESTORE Research Center, UMR-1301, Paul Sabatier University, Toulouse, France
| | | | | | | | | | - Lara Iengo
- Unit of Inherited Cardiomyopathies, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy
| | - Yohan Santin
- Unit of Inherited Cardiomyopathies, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
| | - Fabrizio Tundo
- Department of Clinical Electrophysiology and Cardiac Pacing, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
| | - Claudio Tondo
- Department of Clinical Electrophysiology and Cardiac Pacing, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, Milan, Italy
| | - Giulio Pompilio
- Unit of Inherited Cardiomyopathies, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, Milan, Italy
| | | | - Victorine Douin-Echinard
- I2MC, INSERM, UMR-1297, Toulouse, France
- RESTORE Research Center, UMR-1301, Paul Sabatier University, Toulouse, France
| | - Elena Sommariva
- Unit of Inherited Cardiomyopathies, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
| |
Collapse
|
9
|
Noureddine M, Chang LA, El Ayache F, Laghlali G, Burgess E, Gruneberg L, Warang P, Jiang K, Nijhuis H, Coughlan L, Diego JGB, Park S, Levican J, Schotsaert M. Muscle macrophage regenerative response after squalene-adjuvanted influenza vaccination drives Th2-skewed response and is reduced with age. RESEARCH SQUARE 2025:rs.3.rs-5760877. [PMID: 39975920 PMCID: PMC11838721 DOI: 10.21203/rs.3.rs-5760877/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Squalene-based adjuvants like MF59, and its research alternative AddaVax, induce transient muscle injury, but their working mechanisms downstream of muscle injury remain unclear. We show that an AddaVax-adjuvanted quadrivalent inactivated influenza virus vaccine (QIV) intramuscular injection triggers muscle regeneration-like immune processes and increases CX3CR1+Ly6C+ macrophages in the muscle and inguinal lymph nodes by day 4 post-injection. This leads to a Th2 skewed vaccine response with higher levels of vaccine specific IgG1 titers, and Th2-associated cytokines in the lungs 5 days after subsequent influenza viral challenge. In aged mice, the macrophage recruitment and polarization is diminished, which is consistent with age-associated muscle mass loss, reflecting the age-related decline in muscle regeneration. Unlike young mice, aged mice exhibit a reduction in magnitude and skewing of AddaVax-mediated Th2 responses to QIV. We found that adoptive transfer of bone marrow-derived macrophages derived from young mice into aged mice at the moment of vaccination leads to their infiltration into the injected muscle, where they collect vaccine antigens, drain to the lymph node, and enhance the Th2 response, recapitulating the young host response but in an older host. However, rescuing the Th2-skewing effects of AddaVax alone was not sufficient to enhance protection against mismatched subsequent influenza viral infection in aged mice, suggesting additional factors at play in the diminished vaccine response in aged hosts. This underscores the importance of the macrophage-driven muscle regenerative response in the mechanism of action for squalene-based adjuvants like AddaVax and emphasizes the need to study how muscle damage and regenerative pathways in intramuscular vaccine responses contribute to vaccine effectiveness.
Collapse
Affiliation(s)
- Moataz Noureddine
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lauren A. Chang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Farah El Ayache
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Gabriel Laghlali
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Eleanor Burgess
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Leonie Gruneberg
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kaijun Jiang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Haye Nijhuis
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Lynda Coughlan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Center for Vaccine Development and Global Health (CVD), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Juan Garcia-Bernalt Diego
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Seokchan Park
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jorge Levican
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| |
Collapse
|
10
|
Sabapathy V, Price A, Cheru NT, Venkatadri R, Dogan M, Costlow G, Mohammad S, Sharma R. ST2 + T-Regulatory Cells in Renal Inflammation and Fibrosis after Ischemic Kidney Injury. J Am Soc Nephrol 2025; 36:73-86. [PMID: 39186386 PMCID: PMC11706559 DOI: 10.1681/asn.0000000000000471] [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: 01/05/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024] Open
Abstract
Key Points IL-33/ST2 alarmin pathway regulates inflammation, fibrosis, and resolution of ischemia-reperfusion injury of kidneys. ST2 regulates the transcriptome of T-regulatory cells related to suppressive and reparative functions. The secretome of ST2+ T-regulatory cells regulates hypoxic injury in an amphiregulin-dependent manner. Background Inflammation is a major cause of kidney injury. IL-1 family cytokine IL-33 is released from damaged cells and modulates the immune response through its receptor ST2 expressed on many cell types, including regulatory T cells (Tregs). Although a proinflammatory role of IL-33 has been proposed, exogenous IL-33 expanded Tregs and suppressed renal inflammation. However, the contribution of endogenous IL-33/ST2 for the role of Tregs in the resolution of kidney injury has not been investigated. Methods We used murine renal ischemia-reperfusion injury and kidney organoids (KDOs) to delineate the role of the ST2 and amphiregulin (AREG) specifically in Tregs using targeted deletion. Bulk and single-cell RNA sequencing were performed on flow-sorted Tregs from spleen and CD4 T cells from postischemic kidneys, respectively. The protective role of ST2-sufficient Tregs was analyzed using a novel coculture system of syngeneic KDOs and Tregs under hypoxic conditions. Results Bulk RNA sequencing of splenic and single-cell RNA sequencing of kidney CD4 T cells showed that ST2+ Tregs are enriched for genes related to Treg proliferation and function. Genes for reparative factors, such as Areg , were also enriched in ST2+ Tregs. Treg-specific deletion of ST2 or AREG exacerbated kidney injury and fibrosis in the unilateral ischemia-reperfusion injury model. In coculture studies, wild-type but not ST2-deficient Tregs preserved hypoxia-induced loss of kidney organoid viability, which was restored by AREG supplementation. Conclusions Our study identified the role of the IL-33/ST2 pathway in Tregs for resolution of kidney injury. The transcriptome of ST2+ Tregs was enriched for reparative factors including Areg . Lack of ST2 or AREG in Tregs worsened kidney injury. Tregs protected KDOs from hypoxia in a ST2- and AREG-dependent manner.
Collapse
Affiliation(s)
- Vikram Sabapathy
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
| | - Airi Price
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
- Department of International Health, Georgetown University, Washington, DC
| | - Nardos Tesfaye Cheru
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
- Department of Immunobiology, School of Medicine, Yale University, New Haven, Connecticut
| | - Rajkumar Venkatadri
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
- Immunology Research Unit, GlaxoSmithKline (GSK), Collegeville, Pennsylvania
| | - Murat Dogan
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
- Department of Transplant Surgery, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Gabrielle Costlow
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
| | - Saleh Mohammad
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
| | - Rahul Sharma
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, Virginia
| |
Collapse
|
11
|
Alibhai FJ, Tobin SW. Characterization of Age-Dependent Changes in Skeletal Muscle Repair and Regeneration Using a Mouse Model of Acute Muscle Injury. Methods Mol Biol 2025; 2857:169-180. [PMID: 39348065 DOI: 10.1007/978-1-0716-4128-6_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Acute skeletal muscle injury initiates a process of necrosis, debris clearance, and ultimately tissue regeneration via myogenesis. While skeletal muscle stem cells (MuSCs) are responsible for populating the proliferative myogenic progenitor pool to fuel muscle repair, recruited and resident immune cells have a central role in the regulation of muscle regeneration via the execution of phagocytosis and release of soluble factors that act directly on MuSCs to regulate myogenic differentiation. Therefore, the timing of MuSC proliferation and differentiation is closely linked to the populations and behaviors of immune cells present within skeletal muscle. This has important implications for aging and muscle repair, as systemic changes in immune system function contribute to a decline in muscle regenerative capacity. Here, we present adapted protocols for the isolation of mononuclear cells from skeletal muscles for the quantification of immune cell populations using flow cytometry. We also describe a cardiotoxin skeletal muscle injury protocol and detail the expected outcomes including immune cell infiltration to the injured sites and formation of new myocytes. As immune cell function is substantially influenced by aging, we extend these approaches and outcomes to aged mice.
Collapse
Affiliation(s)
| | - Stephanie W Tobin
- Department of Biology, Trent University, Peterborough, ON, Canada.
- Environmental and Life Sciences Graduate Program, Trent University, Peterborough, ON, Canada.
| |
Collapse
|
12
|
Sisto M, Lisi S. Updates on Inflammatory Molecular Pathways Mediated by ADAM17 in Autoimmunity. Cells 2024; 13:2092. [PMID: 39768182 PMCID: PMC11674862 DOI: 10.3390/cells13242092] [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: 11/13/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
ADAM17 is a member of the disintegrin and metalloproteinase (ADAM) family of transmembrane proteases with immunoregulatory activity in multiple signaling pathways. The functional ADAM17 is involved in the shedding of the ectodomain characterizing many substrates belonging to growth factors, cytokines, receptors, and adhesion molecules. The ADAM17-dependent pathways are known to be crucial in tumor development and progression and in the modulation of many pathological and physiological processes. In the last decade, ADAM17 was considered the driver of several autoimmune pathologies, and numerous substrate-mediated signal transduction pathways were identified. However, the discoveries made to date have led researchers to try to clarify the multiple mechanisms in which ADAM17 is involved and to identify any molecular gaps between the different transductional cascades. In this review, we summarize the most recent updates on the multiple regulatory activities of ADAM17, focusing on reported data in the field of autoimmunity.
Collapse
Affiliation(s)
- Margherita Sisto
- Department of Translational Biomedicine and Neuroscience (DiBraiN), Section of Human Anatomy and Histology, University of Bari “Aldo Moro”, Piazza Giulio Cesare 1, I-70124 Bari, Italy;
| | | |
Collapse
|
13
|
Qi J, Wu Y, Liu Y, Ma J, Wang Z. hsa_circ_0007755 competitively adsorbs miR-27b-3p to mediate CXCL2 expression and recruit Th1 cells to promote hypertrophic scars development. Heliyon 2024; 10:e39169. [PMID: 39524791 PMCID: PMC11544067 DOI: 10.1016/j.heliyon.2024.e39169] [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: 12/28/2023] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
Background and objective The circular RNA hsa_circ_0007755 is markedly upregulated in hypertrophic scars (HS), yet its functional roles in this fibroproliferative disorder remain to be elucidated. This investigation aims to delineate the regulatory mechanisms of hsa_circ_0007755 in HS and to decode its downstream molecular signaling pathways. Methods We established a murine model of HS. Tissue histopathology was assessed using Hematoxylin and Eosin and Masson's trichrome staining. Peripheral blood from the animals was collected and the ratio of T-helper 1 (Th1) to T-helper 2 (Th2) cells was quantified via flow cytometry. The proliferation and apoptosis rates of human hypertrophic scar fibroblasts (hHSFs) were evaluated using the Cell Counting Kit-8 assay and flow cytometry, respectively. The invasive capacity of hHSFs was assessed via a Transwell assay. Co-culture experiments of hHSFs with T cells were conducted, and alterations in Th1/Th2 ratios were monitored using flow cytometry. Levels of cytokines, fibrosis-associated proteins, nuclear factor-kappaB (NF-κB) pathway-related protein, and C-X-C Motif Chemokine Ligand 2 (CXCL2) were quantified using Enzyme-Linked Immunosorbent Assay or Western blot analysis. The interactions between hsa_circ_0007755, miR-27b-3p, and CXCL2 were investigated using dual-luciferase reporter assays and RNA immunoprecipitation. Results Both hsa_circ_0007755 and CXCL2 were highly expressed in HS, whereas miR-27b-3p was downregulated. Knockdown of hsa_circ_0007755 inhibited the proliferation and invasion of hHSFs, promoted apoptosis, and reduced the expression of fibrotic proteins α-SMA and Collagen I, as well as the phosphorylation of the inflammatory pathway protein p65. Co-culture experiments confirmed that hHSFs lowly expressing hsa_circ_0007755 showed a decreased Th1 cell proportion and an increased Th2 cell proportion, alongside lower levels of TNF-α and INF-γ and higher levels of IL-4 and IL-10. The effects of either knocking down or overexpressing hsa_circ_0007755 were reversed by knocking down either miR-27b-3p or CXCL2, respectively. hsa_circ_0007755 acted as a "molecular sponge" for miR-27b-3p, sequestering and diminishing its availability, thereby alleviating its suppression of the target gene CXCL2. Conclusion hsa_circ_0007755 plays a pivotal role in modulating the immune response of HS by influencing the miR-27b-3p/CXCL2 axis, regulating the function and proportion of Th1 and Th2 cells, and thereby affecting the inflammatory and fibrotic processes in scar tissue.
Collapse
Affiliation(s)
- Jun Qi
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong City, Jiangsu Province, 226001, China
| | - YangYang Wu
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong City, Jiangsu Province, 226001, China
| | - YiFei Liu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong City, Jiangsu Province, 226001, China
| | - JiuCheng Ma
- Nantong University, Nantong City, Jiangsu Province, 226001, China
| | - ZhaoNan Wang
- Nantong University, Nantong City, Jiangsu Province, 226001, China
| |
Collapse
|
14
|
Singer M, Elsayed AM, Husseiny MI. Regulatory T-cells: The Face-off of the Immune Balance. FRONT BIOSCI-LANDMRK 2024; 29:377. [PMID: 39614434 DOI: 10.31083/j.fbl2911377] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/29/2024] [Accepted: 08/13/2024] [Indexed: 12/01/2024]
Abstract
Regulatory T-cells (Tregs) play a crucial role in maintaining immune homeostasis, ensuring a balanced immune response. Tregs primarily operate in an antigen-specific fashion, facilitated by their distinct distribution within discrete niches. Tregs have been studied extensively, from their point of origin in the thymus origin to their fate in the periphery or organs. Signals received from antigen-presenting cells (APCs) stimulate Tregs to dampen inflammation. Almost all tumors are characterized by a pathological abundance of immune suppression in their microenvironment. Conversely, the lack thereof proves detrimental to immunological disorders. Achieving a balanced expression of Tregs in relation to other immune compartments is important in establishing an effective and adaptable immune tolerance towards cancer cells and autoantigens. In the context of cancer, it is essential to decrease the frequency of Tregs to overcome tumor suppression. A lower survival rate is associated with the presence of excessive exhausted effector immune cells and an increased frequency of regulatory cells. However, when it comes to treating graft rejection and autoimmune diseases, the focus lies on immune tolerance and the transfer of Tregs. Here, we explore the complex mechanisms that Tregs use in human disease to balance effector immune cells.
Collapse
Affiliation(s)
- Mahmoud Singer
- School of Medicine, University of California Irvine, Irvine, CA 92617, USA
| | - Ahmed M Elsayed
- Division of Infectious Diseases, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Mohamed I Husseiny
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
15
|
Russo C, Surdo S, Valle MS, Malaguarnera L. The Gut Microbiota Involvement in the Panorama of Muscular Dystrophy Pathogenesis. Int J Mol Sci 2024; 25:11310. [PMID: 39457092 PMCID: PMC11508360 DOI: 10.3390/ijms252011310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/16/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024] Open
Abstract
Muscular dystrophies (MDs) are genetically heterogeneous diseases characterized by primary skeletal muscle atrophy. The collapse of muscle structure and irreversible degeneration of tissues promote the occurrence of comorbidities, including cardiomyopathy and respiratory failure. Mitochondrial dysfunction leads to inflammation, fibrosis, and adipogenic cellular infiltrates that exacerbate the symptomatology of MD patients. Gastrointestinal disorders and metabolic anomalies are common in MD patients and may be determined by the interaction between the intestine and its microbiota. Therefore, the gut-muscle axis is one of the actors involved in the spread of inflammatory signals to all muscles. In this review, we aim to examine in depth how intestinal dysbiosis can modulate the metabolic state, the immune response, and mitochondrial biogenesis in the course and progression of the most investigated MDs such as Duchenne Muscular Dystrophy (DMD) and Myotonic Dystrophy (MD1), to better identify gut microbiota metabolites working as therapeutic adjuvants to improve symptoms of MD.
Collapse
Affiliation(s)
- Cristina Russo
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Sofia Surdo
- Italian Center for the Study of Osteopathy (CSDOI), 95124 Catania, Italy;
| | - Maria Stella Valle
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
| | - Lucia Malaguarnera
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| |
Collapse
|
16
|
Ge J, Yin X, Chen L. Regulatory T cells: masterminds of immune equilibrium and future therapeutic innovations. Front Immunol 2024; 15:1457189. [PMID: 39290699 PMCID: PMC11405253 DOI: 10.3389/fimmu.2024.1457189] [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: 06/30/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Regulatory T cells (Tregs), a subset of CD4+T cells marked by the expression of the transcription factor forkhead box protein 3 (Foxp3), are pivotal in maintaining immune equilibrium and preventing autoimmunity. In our review, we addressed the functional distinctions between Foxp3+Tregs and other T cells, highlighting their roles in autoimmune diseases and cancer. We uncovered the dual nature of Tregs: they prevented autoimmune diseases by maintaining self-tolerance while contributing to tumor evasion by suppressing anti-tumor immunity. This study underscored the potential for targeted therapeutic strategies, such as enhancing Treg activity to restore balance in autoimmune diseases or depleting Foxp3+Tregs to augment anti-tumor immune responses in cancer. These insights laid the groundwork for future research and clinical applications, emphasizing the critical role of Foxp3+Tregs in immune regulation and the advancement of next-generation immunotherapies.
Collapse
Affiliation(s)
- Junwei Ge
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Xuan Yin
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| |
Collapse
|
17
|
Zhang N, Zhai L, Wong RMY, Cui C, Law SW, Chow SKH, Goodman SB, Cheung WH. Harnessing immunomodulation to combat sarcopenia: current insights and possible approaches. Immun Ageing 2024; 21:55. [PMID: 39103919 DOI: 10.1186/s12979-024-00458-9] [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: 05/21/2024] [Accepted: 07/24/2024] [Indexed: 08/07/2024]
Abstract
Sarcopenia is a complex age-associated syndrome of progressive loss of muscle mass and strength. Although this condition is influenced by many factors, age-related changes in immune function including immune cell dynamics, and chronic inflammation contribute to its progression. The complex interplay between the immune system, gut-muscle axis, and autophagy further underscores their important roles in sarcopenia pathogenesis. Immunomodulation has emerged as a promising strategy to counteract sarcopenia. Traditional management approaches to treat sarcopenia including physical exercise and nutritional supplementation, and the emerging technologies of biophysical stimulation demonstrated the importance of immunomodulation and regulation of macrophages and T cells and reduction of chronic inflammation. Treatments to alleviate low-grade inflammation in older adults by modulating gut microbial composition and diversity further combat sarcopenia. Furthermore, some pharmacological interventions, nano-medicine, and cell therapies targeting muscle, gut microbiota, or autophagy present additional avenues for immunomodulation in sarcopenia. This narrative review explores the immunological underpinnings of sarcopenia, elucidating the relationship between the immune system and muscle during ageing. Additionally, the review discusses new areas such as the gut-muscle axis and autophagy, which bridge immune system function and muscle health. Insights into current and potential approaches for sarcopenia management through modulation of the immune system are provided, along with suggestions for future research directions and therapeutic strategies. We aim to guide further investigation into clinical immunological biomarkers and identify indicators for sarcopenia diagnosis and potential treatment targets to combat this condition. We also aim to draw attention to the importance of considering immunomodulation in the clinical management of sarcopenia.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China.
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| | - Liting Zhai
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ronald Man Yeung Wong
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Can Cui
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Sheung-Wai Law
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | | | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Wing-Hoi Cheung
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China.
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
18
|
Yue Y, Ren Y, Lu C, Li P, Zhang G. Epigenetic regulation of human FOXP3+ Tregs: from homeostasis maintenance to pathogen defense. Front Immunol 2024; 15:1444533. [PMID: 39144146 PMCID: PMC11323565 DOI: 10.3389/fimmu.2024.1444533] [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: 06/05/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024] Open
Abstract
Regulatory T cells (Tregs), characterized by the expression of Forkhead Box P3 (FOXP3), constitute a distinct subset of T cells crucial for immune regulation. Tregs can exert direct and indirect control over immune homeostasis by releasing inhibitory factors or differentiating into Th-like Treg (Th-Treg), thereby actively contributing to the prevention and treatment of autoimmune diseases. The epigenetic regulation of FOXP3, encompassing DNA methylation, histone modifications, and post-translational modifications, governs the development and optimal suppressive function of Tregs. In addition, Tregs can also possess the ability to maintain homeostasis in diverse microenvironments through non-suppressive mechanisms. In this review, we primarily focus on elucidating the epigenetic regulation of Tregs as well as their multifaceted roles within diverse physiological contexts while looking forward to potential strategies involving augmentation or suppression of Tregs activity for disease management, particularly in light of the ongoing global COVID-19 pandemic.
Collapse
Affiliation(s)
| | | | | | | | - Guojun Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
19
|
Sousa AR, Cunha AF, Santos-Coquillat A, Estrada BH, Spiller KL, Barão M, Rodrigues AF, Simões S, Vilaça A, Ferreira L, Oliveira MB, Mano JF. Shape-Versatile Fixed Cellular Materials for Multiple Target Immunomodulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405367. [PMID: 38739450 PMCID: PMC11272431 DOI: 10.1002/adma.202405367] [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: 04/15/2024] [Indexed: 05/16/2024]
Abstract
Therapeutic cells are usually administered as living agents, despite the risks of undesired cell migration and acquisition of unpredictable phenotypes. Additionally, most cell-based therapies rely on the administration of single cells, often associated with rapid in vivo clearance. 3D cellular materials may be useful to prolong the effect of cellular therapies and offer the possibility of creating structural volumetric constructs. Here, the manufacturing of shape-versatile fixed cell-based materials with immunomodulatory properties is reported. Living cell aggregates with different shapes (spheres and centimeter-long fibers) are fixed using a method compatible with maintenance of structural integrity, robustness, and flexibility of 3D constructs. The biological properties of living cells can be modulated before fixation, rendering an in vitro anti-inflammatory effect toward human macrophages, in line with a decreased activation of the nuclear factor kappa B (NF-κB) pathway that preponderantly correlated with the surface area of the materials. These findings are further corroborated in vivo in mouse skin wounds. Contact with fixed materials also reduces the proliferation of activated primary T lymphocytes, while promoting regulatory populations. The fixation of cellular constructs is proposed as a versatile phenotypic stabilization method that can be easily implemented to prepare immunomodulatory materials with therapeutic potential.
Collapse
Affiliation(s)
- Ana Rita Sousa
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Ana F Cunha
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Ana Santos-Coquillat
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Beatriz Hernaez Estrada
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Chestnut Street, Philadelphia, PA, 19104, USA
| | - Kara L Spiller
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Chestnut Street, Philadelphia, PA, 19104, USA
| | - Marta Barão
- CNC-Center for Neurosciences and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, 3004-517, Portugal
| | - Artur Filipe Rodrigues
- CNC-Center for Neurosciences and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, 3004-517, Portugal
| | - Susana Simões
- CNC-Center for Neurosciences and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, 3004-517, Portugal
| | - Andreia Vilaça
- CNC-Center for Neurosciences and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, 3004-517, Portugal
| | - Lino Ferreira
- CNC-Center for Neurosciences and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, 3004-517, Portugal
- FMUC-Faculty of Medicine, University of Coimbra, Coimbra, 3004-517, Portugal
| | - Mariana B Oliveira
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, 3810-193, Portugal
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, 3810-193, Portugal
| |
Collapse
|
20
|
Han J, Rindone AN, Elisseeff JH. Immunoengineering Biomaterials for Musculoskeletal Tissue Repair across Lifespan. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311646. [PMID: 38416061 PMCID: PMC11239302 DOI: 10.1002/adma.202311646] [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: 11/03/2023] [Revised: 01/23/2024] [Indexed: 02/29/2024]
Abstract
Musculoskeletal diseases and injuries are among the leading causes of pain and morbidity worldwide. Broad efforts have focused on developing pro-regenerative biomaterials to treat musculoskeletal conditions; however, these approaches have yet to make a significant clinical impact. Recent studies have demonstrated that the immune system is central in orchestrating tissue repair and that targeting pro-regenerative immune responses can improve biomaterial therapeutic outcomes. However, aging is a critical factor negatively affecting musculoskeletal tissue repair and immune function. Hence, understanding how age affects the response to biomaterials is essential for improving musculoskeletal biomaterial therapies. This review focuses on the intersection of the immune system and aging in response to biomaterials for musculoskeletal tissue repair. The article introduces the general impacts of aging on tissue physiology, the immune system, and the response to biomaterials. Then, it explains how the adaptive immune system guides the response to injury and biomaterial implants in cartilage, muscle, and bone and discusses how aging impacts these processes in each tissue type. The review concludes by highlighting future directions for the development and translation of personalized immunomodulatory biomaterials for musculoskeletal tissue repair.
Collapse
Affiliation(s)
- Jin Han
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD 21231, USA
| | - Alexandra N. Rindone
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD 21231, USA
| | - Jennifer H. Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD 21231, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine; Baltimore, MD 21231, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University; Baltimore, MD 21231, USA
| |
Collapse
|
21
|
Miao Y, Li Z, Feng J, Lei X, Shan J, Qian C, Li J. The Role of CD4 +T Cells in Nonalcoholic Steatohepatitis and Hepatocellular Carcinoma. Int J Mol Sci 2024; 25:6895. [PMID: 39000005 PMCID: PMC11240980 DOI: 10.3390/ijms25136895] [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: 05/06/2024] [Revised: 06/14/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
Hepatocellular carcinoma (HCC) has become the fourth leading cause of cancer-related deaths worldwide; annually, approximately 830,000 deaths related to liver cancer are diagnosed globally. Since early-stage HCC is clinically asymptomatic, traditional treatment modalities, including surgical ablation, are usually not applicable or result in recurrence. Immunotherapy, particularly immune checkpoint blockade (ICB), provides new hope for cancer therapy; however, immune evasion mechanisms counteract its efficiency. In addition to viral exposure and alcohol addiction, nonalcoholic steatohepatitis (NASH) has become a major cause of HCC. Owing to NASH-related aberrant T cell activation causing tissue damage that leads to impaired immune surveillance, NASH-associated HCC patients respond much less efficiently to ICB treatment than do patients with other etiologies. In addition, abnormal inflammation contributes to NASH progression and NASH-HCC transition, as well as to HCC immune evasion. Therefore, uncovering the detailed mechanism governing how NASH-associated immune cells contribute to NASH progression would benefit HCC prevention and improve HCC immunotherapy efficiency. In the following review, we focused our attention on summarizing the current knowledge of the role of CD4+T cells in NASH and HCC progression, and discuss potential therapeutic strategies involving the targeting of CD4+T cells for the treatment of NASH and HCC.
Collapse
Affiliation(s)
- Yadi Miao
- School of Medicine, Chongqing University, Chongqing 400030, China
| | - Ziyong Li
- School of Medicine, Chongqing University, Chongqing 400030, China
| | - Juan Feng
- Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Xia Lei
- School of Medicine, Chongqing University, Chongqing 400030, China
| | - Juanjuan Shan
- School of Medicine, Chongqing University, Chongqing 400030, China
- Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Cheng Qian
- School of Medicine, Chongqing University, Chongqing 400030, China
- Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Jiatao Li
- School of Medicine, Chongqing University, Chongqing 400030, China
- Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| |
Collapse
|
22
|
Guglielmi V, Cheli M, Tonin P, Vattemi G. Sporadic Inclusion Body Myositis at the Crossroads between Muscle Degeneration, Inflammation, and Aging. Int J Mol Sci 2024; 25:2742. [PMID: 38473988 PMCID: PMC10932328 DOI: 10.3390/ijms25052742] [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: 12/27/2023] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Sporadic inclusion body myositis (sIBM) is the most common muscle disease of older people and is clinically characterized by slowly progressive asymmetrical muscle weakness, predominantly affecting the quadriceps, deep finger flexors, and foot extensors. At present, there are no enduring treatments for this relentless disease that eventually leads to severe disability and wheelchair dependency. Although sIBM is considered a rare muscle disorder, its prevalence is certainly higher as the disease is often undiagnosed or misdiagnosed. The histopathological phenotype of sIBM muscle biopsy includes muscle fiber degeneration and endomysial lymphocytic infiltrates that mainly consist of cytotoxic CD8+ T cells surrounding nonnecrotic muscle fibers expressing MHCI. Muscle fiber degeneration is characterized by vacuolization and the accumulation of congophilic misfolded multi-protein aggregates, mainly in their non-vacuolated cytoplasm. Many players have been identified in sIBM pathogenesis, including environmental factors, autoimmunity, abnormalities of protein transcription and processing, the accumulation of several toxic proteins, the impairment of autophagy and the ubiquitin-proteasome system, oxidative and nitrative stress, endoplasmic reticulum stress, myonuclear degeneration, and mitochondrial dysfunction. Aging has also been proposed as a contributor to the disease. However, the interplay between these processes and the primary event that leads to the coexistence of autoimmune and degenerative changes is still under debate. Here, we outline our current understanding of disease pathogenesis, focusing on degenerative mechanisms, and discuss the possible involvement of aging.
Collapse
Affiliation(s)
- Valeria Guglielmi
- Cellular and Molecular Biology of Cancer Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA;
- Immunity and Pathogenesis Program, Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Marta Cheli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (M.C.); (P.T.)
| | - Paola Tonin
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (M.C.); (P.T.)
| | - Gaetano Vattemi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (M.C.); (P.T.)
| |
Collapse
|
23
|
Jiang H, Liu B, Lin J, Xue T, Han Y, Lu C, Zhou S, Gu Y, Xu F, Shen Y, Xu L, Sun H. MuSCs and IPCs: roles in skeletal muscle homeostasis, aging and injury. Cell Mol Life Sci 2024; 81:67. [PMID: 38289345 PMCID: PMC10828015 DOI: 10.1007/s00018-023-05096-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: 10/04/2023] [Revised: 12/01/2023] [Accepted: 12/17/2023] [Indexed: 02/01/2024]
Abstract
Skeletal muscle is a highly specialized tissue composed of myofibres that performs crucial functions in movement and metabolism. In response to external stimuli and injuries, a range of stem/progenitor cells, with muscle stem cells or satellite cells (MuSCs) being the predominant cell type, are rapidly activated to repair and regenerate skeletal muscle within weeks. Under normal conditions, MuSCs remain in a quiescent state, but become proliferative and differentiate into new myofibres in response to injury. In addition to MuSCs, some interstitial progenitor cells (IPCs) such as fibro-adipogenic progenitors (FAPs), pericytes, interstitial stem cells expressing PW1 and negative for Pax7 (PICs), muscle side population cells (SPCs), CD133-positive cells and Twist2-positive cells have been identified as playing direct or indirect roles in regenerating muscle tissue. Here, we highlight the heterogeneity, molecular markers, and functional properties of these interstitial progenitor cells, and explore the role of muscle stem/progenitor cells in skeletal muscle homeostasis, aging, and muscle-related diseases. This review provides critical insights for future stem cell therapies aimed at treating muscle-related diseases.
Collapse
Affiliation(s)
- Haiyan Jiang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Boya Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Junfei Lin
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Tong Xue
- Department of Paediatrics, Medical School of Nantong University, Nantong University, Nantong, 226001, People's Republic of China
| | - Yimin Han
- Department of Paediatrics, Medical School of Nantong University, Nantong University, Nantong, 226001, People's Republic of China
| | - Chunfeng Lu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, Jiangsu, People's Republic of China
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yun Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Feng Xu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| | - Lingchi Xu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| |
Collapse
|
24
|
Kang JH, Zappasodi R. Modulating Treg stability to improve cancer immunotherapy. Trends Cancer 2023; 9:911-927. [PMID: 37598003 DOI: 10.1016/j.trecan.2023.07.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/23/2023] [Accepted: 07/25/2023] [Indexed: 08/21/2023]
Abstract
Immunosuppressive regulatory T cells (Tregs) provide a main mechanism of tumor immune evasion. Targeting Tregs, especially in the tumor microenvironment (TME), continues to be investigated to improve cancer immunotherapy. Recent studies have unveiled intratumoral Treg heterogeneity and plasticity, furthering the complexity of the role of Tregs in tumor immunity and immunotherapy response. The phenotypic and functional diversity of intratumoral Tregs can impact their response to therapy and may offer new targets to modulate specific Treg subsets. In this review we provide a unifying framework of critical factors contributing to Treg heterogeneity and plasticity in the TME, and we discuss how this information can guide the development of more specific Treg-targeting therapies for cancer immunotherapy.
Collapse
Affiliation(s)
- Jee Hye Kang
- Weill Cornell Medicine, Weill Cornell Medical College of Cornell University, New York, NY, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School, New York, NY, USA
| | - Roberta Zappasodi
- Weill Cornell Medicine, Weill Cornell Medical College of Cornell University, New York, NY, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School, New York, NY, USA.
| |
Collapse
|
25
|
Benamar M, Chen Q, Martinez-Blanco M, Chatila TA. Regulatory T cells in allergic inflammation. Semin Immunol 2023; 70:101847. [PMID: 37837939 PMCID: PMC10842049 DOI: 10.1016/j.smim.2023.101847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/16/2023]
Abstract
Regulatory T (Treg) cells maintain immune tolerance to allergens at the environmental interfaces in the airways, skin and gut, marshalling in the process distinct immune regulatory circuits operative in the respective tissues. Treg cells are coordinately mobilized with allergic effector mechanisms in the context of a tissue-protective allergic inflammatory response against parasites, toxins and potentially harmful allergens, serving to both limit the inflammation and promote local tissue repair. Allergic diseases are associated with subverted Treg cell responses whereby a chronic allergic inflammatory environment can skew Treg cells toward pathogenic phenotypes that both perpetuate and aggravate disease. Interruption of Treg cell subversion in chronic allergic inflammatory conditions may thus provide novel therapeutic strategies by re-establishing effective immune regulation.
Collapse
Affiliation(s)
- Mehdi Benamar
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Qian Chen
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Monica Martinez-Blanco
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Talal A Chatila
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA; Lead Contact, USA.
| |
Collapse
|
26
|
Lao P, Chen J, Tang L, Zhang J, Chen Y, Fang Y, Fan X. Regulatory T cells in lung disease and transplantation. Biosci Rep 2023; 43:BSR20231331. [PMID: 37795866 PMCID: PMC10611924 DOI: 10.1042/bsr20231331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/28/2023] [Accepted: 10/04/2023] [Indexed: 10/06/2023] Open
Abstract
Pulmonary disease can refer to the disease of the lung itself or the pulmonary manifestations of systemic diseases, which are often connected to the malfunction of the immune system. Regulatory T (Treg) cells have been shown to be important in maintaining immune homeostasis and preventing inflammatory damage, including lung diseases. Given the increasing amount of evidence linking Treg cells to various pulmonary conditions, Treg cells might serve as a therapeutic strategy for the treatment of lung diseases and potentially promote lung transplant tolerance. The most potent and well-defined Treg cells are Foxp3-expressing CD4+ Treg cells, which contribute to the prevention of autoimmune lung diseases and the promotion of lung transplant rejection. The protective mechanisms of Treg cells in lung disease and transplantation involve multiple immune suppression mechanisms. This review summarizes the development, phenotype and function of CD4+Foxp3+ Treg cells. Then, we focus on the therapeutic potential of Treg cells in preventing lung disease and limiting lung transplant rejection. Furthermore, we discussed the possibility of Treg cell utilization in clinical applications. This will provide an overview of current research advances in Treg cells and their relevant application in clinics.
Collapse
Affiliation(s)
- Peizhen Lao
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Jingyi Chen
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Longqian Tang
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Jiwen Zhang
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Yuxi Chen
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Yuyin Fang
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| | - Xingliang Fan
- Institute of Biological and Food Engineering, Guangdong University of Education, 351 Xingang Middle Road, Guangzhou 510303, PR China
| |
Collapse
|
27
|
Gandolfi S, Pileyre B, Drouot L, Dubus I, Auquit-Auckbur I, Martinet J. Stromal vascular fraction in the treatment of myositis. Cell Death Discov 2023; 9:346. [PMID: 37726262 PMCID: PMC10509179 DOI: 10.1038/s41420-023-01605-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 08/01/2023] [Accepted: 08/14/2023] [Indexed: 09/21/2023] Open
Abstract
Muscle regeneration is a physiological process that converts satellite cells into mature myotubes under the influence of an inflammatory environment progressively replaced by an anti-inflammatory environment, with precise crosstalk between immune and muscular cells. If the succession of these phases is disturbed, the immune system can sometimes become auto-reactive, leading to chronic muscular inflammatory diseases, such as myositis. The triggers of these autoimmune myopathies remain mostly unknown, but the main mechanisms of pathogenesis are partially understood. They involve chronic inflammation, which could be associated with an auto-reactive immune response, and gradually with a decrease in the regenerative capacities of the muscle, leading to its degeneration, fibrosis and vascular architecture deterioration. Immunosuppressive treatments can block the first part of the process, but sometimes muscle remains weakened, or even still deteriorates, due to the exhaustion of its capacities. For patients refractory to immunosuppressive therapies, mesenchymal stem cells have shown interesting effects but their use is limited by their availability. Stromal vascular fraction, which can easily be extracted from adipose tissue, has shown good tolerance and possible therapeutic benefits in several degenerative and autoimmune diseases. However, despite the increasing use of stromal vascular fraction, the therapeutically active components within this heterogeneous cellular product are ill-defined and the mechanisms by which this therapy might be active remain insufficiently understood. We review herein the current knowledge on the mechanisms of action of stromal vascular fraction and hypothesise on how it could potentially respond to some of the unmet treatment needs of refractory myositis.
Collapse
Affiliation(s)
- S Gandolfi
- Univ Rouen Normandie, INSERM U1234, FOCIS Center of Excellence PAn'THER, F-76000, Rouen, France
- Toulouse University Hospital, Department of Plastic and Reconstructive Surgery, F-31000, Toulouse, France
| | - B Pileyre
- Univ Rouen Normandie, INSERM U1234, FOCIS Center of Excellence PAn'THER, F-76000, Rouen, France.
- Centre Henri Becquerel, Department of Pharmacy, F-76000, Rouen, France.
| | - L Drouot
- Univ Rouen Normandie, INSERM U1234, FOCIS Center of Excellence PAn'THER, F-76000, Rouen, France
| | - I Dubus
- Univ Rouen Normandie, INSERM U1234, FOCIS Center of Excellence PAn'THER, F-76000, Rouen, France
| | - I Auquit-Auckbur
- Univ Rouen Normandie, INSERM U1234, FOCIS Center of Excellence PAn'THER, CHU Rouen, Department of Plastic, Reconstructive and Hand Surgery, F-76000, Rouen, France
| | - J Martinet
- Univ Rouen Normandie, INSERM U1234, FOCIS Center of Excellence PAn'THER, CHU Rouen, Department of Immunology and Biotherapy, F-76000, Rouen, France
| |
Collapse
|
28
|
Li Z, Peng L, Sun L, Si J. A link between mitochondrial damage and the immune microenvironment of delayed onset muscle soreness. BMC Med Genomics 2023; 16:196. [PMID: 37612729 PMCID: PMC10464284 DOI: 10.1186/s12920-023-01621-9] [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: 03/30/2023] [Accepted: 07/31/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND Delayed onset muscle soreness (DOMS) is a self-healing muscle pain disorder. Inflammatory pain is the main feature of DOMS. More and more researchers have realized that changes in mitochondrial morphology are related to pain. However, the role of mitochondria in the pathogenesis of DOMS and the abnormal immune microenvironment is still unknown. METHODS Mitochondria-related genes and gene expression data were obtained from MitoCarta3.0 and NCBI GEO databases. The network of mitochondrial function and the immune microenvironment of DOMS was constructed by computer algorithm. Subsequently, the skeletal muscle of DOMS rats was subjected to qPCR to verify the bioinformatics results. DOMS and non-DOMS histological samples were further studied by staining and transmission electron microscopy. RESULTS Bioinformatics results showed that expression of mitochondria-related genes was changed in DOMS. The results of qPCR showed that four hub genes (AMPK, PGC1-α, SLC25A25, and ARMCX1) were differentially expressed in DOMS. These hub genes are related to the degree of skeletal muscle immune cell infiltration, mitochondrial respiratory chain complex, DAMPs, the TCA cycle, and mitochondrial metabolism. Bayesian network inference showed that IL-6 and PGC1-α may be the main regulatory genes of mitochondrial damage in DOMS. Transmission electron microscopy revealed swelling of skeletal muscle mitochondria and disorganization of myofilaments. CONCLUSIONS Our study found that skeletal muscle mitochondrial damage is one of the causes of inflammatory factor accumulation in DOMS. According to the screened-out hub genes, this study provides a reference for follow-up clinical application.
Collapse
Affiliation(s)
- Zheng Li
- College of Sport Human Sciences, Harbin Sport University, No. 1, Dacheng Road, Nangang District, 150008, Harbin, China
| | - Lina Peng
- College of Sport Human Sciences, Harbin Sport University, No. 1, Dacheng Road, Nangang District, 150008, Harbin, China
| | - Lili Sun
- College of Sport Human Sciences, Harbin Sport University, No. 1, Dacheng Road, Nangang District, 150008, Harbin, China.
| | - Juncheng Si
- College of Sport Human Sciences, Harbin Sport University, No. 1, Dacheng Road, Nangang District, 150008, Harbin, China
| |
Collapse
|
29
|
Martins L, Amorim WW, Gregnani MF, de Carvalho Araújo R, Qadri F, Bader M, Pesquero JB. Kinin receptors regulate skeletal muscle regeneration: differential effects for B1 and B2 receptors. Inflamm Res 2023; 72:1583-1601. [PMID: 37464053 PMCID: PMC10499706 DOI: 10.1007/s00011-023-01766-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/20/2023] [Accepted: 07/02/2023] [Indexed: 07/20/2023] Open
Abstract
OBJECTIVE AND DESIGN After traumatic skeletal muscle injury, muscle healing is often incomplete and produces extensive fibrosis. Bradykinin (BK) reduces fibrosis in renal and cardiac damage models through the B2 receptor. The B1 receptor expression is induced by damage, and blocking of the kallikrein-kinin system seems to affect the progression of muscular dystrophy. We hypothesized that both kinin B1 and B2 receptors could play a differential role after traumatic muscle injury, and the lack of the B1 receptor could produce more cellular and molecular substrates for myogenesis and fewer substrates for fibrosis, leading to better muscle healing. MATERIAL AND METHODS To test this hypothesis, tibialis anterior muscles of kinin receptor knockout animals were subjected to traumatic injury. Myogenesis, angiogenesis, fibrosis, and muscle functioning were evaluated. RESULTS Injured B1KO mice showed a faster healing progression of the injured area with a larger amount of central nucleated fiber post-injury when compared to control mice. In addition, they exhibited higher neovasculogenic capacity, maintaining optimal tissue perfusion for the post-injury phase; had higher amounts of myogenic markers with less inflammatory infiltrate and tissue destruction. This was followed by higher amounts of SMAD7 and lower amounts of p-SMAD2/3, which resulted in less fibrosis. In contrast, B2KO and B1B2KO mice showed more severe tissue destruction and excessive fibrosis. B1KO animals had better results in post-injury functional tests compared to control animals. CONCLUSIONS We demonstrate that injured skeletal muscle tissues have a better repair capacity with less fibrosis in the presence of B2 receptor and absence of B1 receptor, including better performances in functional tests.
Collapse
Affiliation(s)
- Leonardo Martins
- Division of Medical Sciences, Laboratory of Transcriptional Regulation, Institute of Medical Biology of Polish Academy of Sciences (IMB-PAN), 3a Tylna St., 90-364, Łódź, Poland.
- Center for Research and Molecular Diagnosis of Genetic Diseases, Federal University of São Paulo, Rua Pedro de Toledo 669, 9th Floor, São Paulo, 04039032, Brazil.
- Department of Biochemistry and Molecular Biology, Federal University of São Paulo, Rua Três de Maio 100, 4th Floor, São Paulo, 04044-020, Brazil.
| | - Weslley Wallace Amorim
- Center for Research and Molecular Diagnosis of Genetic Diseases, Federal University of São Paulo, Rua Pedro de Toledo 669, 9th Floor, São Paulo, 04039032, Brazil
| | - Marcos Fernandes Gregnani
- Laboratory of Exercise Genetics and Metabolism, Federal University of São Paulo, Rua Pedro de Toledo 669, 9th Floor, São Paulo, 04039032, Brazil
| | - Ronaldo de Carvalho Araújo
- Laboratory of Exercise Genetics and Metabolism, Federal University of São Paulo, Rua Pedro de Toledo 669, 9th Floor, São Paulo, 04039032, Brazil
| | - Fatimunnisa Qadri
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Michael Bader
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany
- Institute for Biology, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
- Charité University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Potsdamer Str. 58, 10785, Berlin, Germany
| | - João Bosco Pesquero
- Center for Research and Molecular Diagnosis of Genetic Diseases, Federal University of São Paulo, Rua Pedro de Toledo 669, 9th Floor, São Paulo, 04039032, Brazil.
- Department of Biophysics, Federal University of São Paulo, Rua Botucatu 862, 6th Floor, São Paulo, 04023-062, Brazil.
| |
Collapse
|
30
|
Moon S, Hong J, Go S, Kim BS. Immunomodulation for Tissue Repair and Regeneration. Tissue Eng Regen Med 2023; 20:389-409. [PMID: 36920675 PMCID: PMC10219918 DOI: 10.1007/s13770-023-00525-0] [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: 10/20/2022] [Revised: 01/11/2023] [Accepted: 01/26/2023] [Indexed: 03/16/2023] Open
Abstract
Various immune cells participate in repair and regeneration following tissue injury or damage, orchestrating tissue inflammation and regeneration processes. A deeper understanding of the immune system's involvement in tissue repair and regeneration is critical for the development of successful reparatory and regenerative strategies. Here we review recent technologies that facilitate cell-based and biomaterial-based modulation of the immune systems for tissue repair and regeneration. First, we summarize the roles of various types of immune cells in tissue repair. Second, we review the principle, examples, and limitations of regulatory T (Treg) cell-based therapy, a representative cell-based immunotherapy. Finally, we discuss biomaterial-based immunotherapy strategies that aim to modulate immune cells using various biomaterials for tissue repair and regeneration.
Collapse
Affiliation(s)
- Sangjun Moon
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jihye Hong
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seokhyeong Go
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea.
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea.
- Institute of Chemical Processes, Institute of Engineering Research, BioMAX, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
31
|
Koike H, Manabe I, Oishi Y. Mechanisms of cooperative cell-cell interactions in skeletal muscle regeneration. Inflamm Regen 2022; 42:48. [DOI: 10.1186/s41232-022-00234-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/08/2022] [Indexed: 11/17/2022] Open
Abstract
AbstractSkeletal muscles have an extraordinary capacity to regenerate themselves when injured. Skeletal muscle stem cells, called satellite cells, play a central role in muscle regeneration via three major steps: activation, proliferation, and differentiation. These steps are affected by multiple types of cells, such as immune cells, fibro-adipogenic progenitor cells, and vascular endothelial cells. The widespread use of single-cell sequencing technologies has enabled the identification of novel cell subpopulations associated with muscle regeneration and their regulatory mechanisms. This review summarizes the dynamism of the cellular community that controls and promotes muscle regeneration, with a particular focus on skeletal muscle stem cells.
Collapse
|
32
|
Crescioli C. Vitamin D, exercise, and immune health in athletes: A narrative review. Front Immunol 2022; 13:954994. [PMID: 36211340 PMCID: PMC9539769 DOI: 10.3389/fimmu.2022.954994] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022] Open
Abstract
Vitamin D exerts important extra-skeletal effects, exhibiting an exquisite immune regulatory ability, affecting both innate and adaptive immune responses through the modulation of immunocyte function and signaling. Remarkably, the immune function of working skeletal muscle, which is fully recognized to behave as a secretory organ with immune capacity, is under the tight control of vitamin D as well. Vitamin D status, meaning hormone sufficiency or insufficiency, can push toward strengthening/stabilization or decline of immune surveillance, with important consequences for health. This aspect is particularly relevant when considering the athletic population: while exercising is, nowadays, the recommended approach to maintain health and counteract inflammatory processes, “too much” exercise, often experienced by athletes, can increase inflammation, decrease immune surveillance, and expose them to a higher risk of diseases. When overexercise intersects with hypovitaminosis D, the overall effects on the immune system might converge into immune depression and higher vulnerability to diseases. This paper aims to provide an overview of how vitamin D shapes human immune responses, acting on the immune system and skeletal muscle cells; some aspects of exercise-related immune modifications are addressed, focusing on athletes. The crossroad where vitamin D and exercise meet can profile whole-body immune response and health.
Collapse
|