1
|
Humer B, Dik WA, Versnel MA. Advocating the role of trained immunity in the pathogenesis of ME/CFS: a mini review. Front Immunol 2025; 16:1483764. [PMID: 40201181 PMCID: PMC11975576 DOI: 10.3389/fimmu.2025.1483764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 03/11/2025] [Indexed: 04/10/2025] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a complex chronic disease of which the underlying (molecular) mechanisms are mostly unknown. An estimated 0.89% of the global population is affected by ME/CFS. Most patients experience a multitude of symptoms that severely affect their lives. These symptoms include post-exertional malaise, chronic fatigue, sleep disorder, impaired cognitive functions, flu-like symptoms, and chronic immune activation. Therapy focusses on symptom management, as there are no drugs available. Approximately 60% of patients develop ME/CFS following an acute infection. Such a preceding infection may induce a state of trained immunity; defined as acquired, nonspecific, immunological memory of innate immune cells. Trained immune cells undergo long term epigenetic reprogramming, which leads to changes in chromatin accessibility, metabolism, and results in a hyperresponsive phenotype. Initially, trained immunity has only been demonstrated in peripheral blood monocytes and macrophages. However, more recent findings indicate that hematopoietic stem cells in the bone marrow are required for long-term persistence of trained immunity. While trained immunity is beneficial to combat infections, a disproportionate response may cause disease. We hypothesize that pronounced hyperresponsiveness of innate immune cells to stimuli could account for the aberrant activation of various immune pathways, thereby contributing to the pathophysiology of ME/CFS. In this mini review, we elaborate on the concept of trained immunity as a factor involved in the pathogenesis of ME/CFS by presenting evidence from other post-infectious diseases with symptoms that closely resemble those of ME/CFS.
Collapse
Affiliation(s)
- Bart Humer
- Department of Immunology, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Willem A. Dik
- Laboratory Medical Immunology, Department of Immunology, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Marjan A. Versnel
- Department of Immunology, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
2
|
Pacilio S, Lombardi S, Costa R, Paris F, Petrocelli G, Marrazzo P, Cenacchi G, Alviano F. Role of Perinatal Stem Cell Secretome as Potential Therapy for Muscular Dystrophies. Biomedicines 2025; 13:458. [PMID: 40002871 PMCID: PMC11852414 DOI: 10.3390/biomedicines13020458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/02/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Inflammation mechanisms play a critical role in muscle homeostasis, and in Muscular Dystrophies (MDs), the myofiber damage triggers chronic inflammation which significantly controls the disease progression. Immunomodulatory strategies able to target inflammatory pathways and mitigate the immune-mediated damage in MDs may provide new therapeutic options. Owing to its capacity of influencing the immune response and enhancing tissue repair, stem cells' secretome has been proposed as an adjunct or standalone treatment for MDs. In this review study, we discuss the challenging points related to the inflammation condition characterizing MD pathology and provide a concise summary of the literature supporting the potential of perinatal stem cells in targeting and modulating the MD inflammation.
Collapse
Affiliation(s)
- Serafina Pacilio
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Sara Lombardi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Roberta Costa
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Francesca Paris
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Giovannamaria Petrocelli
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| | - Pasquale Marrazzo
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Giovanna Cenacchi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Francesco Alviano
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| |
Collapse
|
3
|
Hernández Rodríguez MY, Biswas DD, Slyne AD, Lee J, Scarrow E, Abdelbarr SM, Daniels H, O’Halloran KD, Ferreira LF, Gersbach CA, ElMallah MK. Respiratory pathology in the mdx/utrn -/- mouse: A murine model for Duchenne Muscular Dystrophy (DMD). PLoS One 2025; 20:e0316295. [PMID: 39919154 PMCID: PMC11805407 DOI: 10.1371/journal.pone.0316295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/09/2024] [Indexed: 02/09/2025] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked devastating disease caused by a lack of dystrophin which results in progressive muscle weakness. As muscle weakness progresses, respiratory insufficiency and hypoventilation result in significant morbidity and mortality. The most studied DMD mouse model- the mdx mouse- has a milder respiratory phenotype compared to humans, likely due to compensatory overexpression of utrophin. mdx/utrn-/- mice lack both dystrophin and utrophin proteins. These mice have an early onset of muscular dystrophy, severe muscle weakness, and premature death, but the respiratory pathophysiology is unclear. The objective of this study is to characterize the respiratory pathophysiology and histopathology using whole body plethysmography to measure breathing and metabolism, diaphragm muscle functional analysis, histology, and immunohistochemistry. The mdx/utrn-/- mice have significant respiratory and metabolic deficits with respiratory insufficiency and hypoventilation when exposed to hypoxia and hypercarbia as early as 6 weeks of age. They also have significant diaphragmatic weakness and disrupted diaphragmatic structural pathology. The mdx/utrn-/- mice display respiratory dysfunction that mimics the DMD phenotype and therefore can provide a useful model to study the impact of novel therapies on respiratory function for DMD.
Collapse
Affiliation(s)
- Marán Y. Hernández Rodríguez
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Duke University, Durham, North Carolina, United States of America
| | - Debolina D. Biswas
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Duke University, Durham, North Carolina, United States of America
| | - Aoife D. Slyne
- Department of Physiology, University College Cork, Cork, Ireland
| | - Jane Lee
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Duke University, Durham, North Carolina, United States of America
| | - Evelyn Scarrow
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Duke University, Durham, North Carolina, United States of America
| | - Sarra M. Abdelbarr
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Duke University, Durham, North Carolina, United States of America
| | - Heather Daniels
- Center for Advanced Genomic Technologies, Duke University, Durham, North Carolina, United States of America
| | | | - Leonardo F. Ferreira
- Department of Orthopedic Surgery, Duke University, Durham, North Carolina, United States of America
| | - Charles A. Gersbach
- Center for Advanced Genomic Technologies, Duke University, Durham, North Carolina, United States of America
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
- Department of Surgery, Duke University Hospital, Durham, North Carolina, United States of America
| | - Mai K. ElMallah
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Duke University, Durham, North Carolina, United States of America
- Department of Neurobiology, Duke University, Durham, North Carolina, United States of America
- Department of Cell Biology, Duke University, Durham, North Carolina, United States of America
| |
Collapse
|
4
|
Fennel ZJ, O'Connell RM, Drummond MJ. Macrophage immunometabolism: emerging targets for regrowth in aging muscle. Am J Physiol Endocrinol Metab 2025; 328:E186-E197. [PMID: 39763086 DOI: 10.1152/ajpendo.00403.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/31/2025]
Abstract
The recovery from muscle atrophy is impaired with aging as characterized by improper muscle remodeling and sustained functional deficits. Age-related deficits in muscle regrowth are tightly linked with the loss of early pro-inflammatory macrophage responses and subsequent cellular dysregulation within the skeletal muscle niche. Macrophage inflammatory phenotype is regulated at the metabolic level, highlighting immunometabolism as an emerging strategy to enhance macrophage responses and restore functional muscle regrowth. Accordingly, metabolic targets with an emphasis on glycolytic, hypoxia, and redox-related pathways stand out for their role in promoting macrophage inflammation and enhancing muscle regrowth in aging. Here we highlight promising immuno-metabolic targets that could be leveraged to restore optimal pro-inflammatory macrophage function in aging and enhance muscle regrowth following muscular atrophy.
Collapse
Affiliation(s)
- Zachary J Fennel
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, United States
| | - Ryan M O'Connell
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, United States
| | - Micah J Drummond
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, United States
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
5
|
Hajishengallis G, Netea MG, Chavakis T. Trained immunity in chronic inflammatory diseases and cancer. Nat Rev Immunol 2025:10.1038/s41577-025-01132-x. [PMID: 39891000 DOI: 10.1038/s41577-025-01132-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2025] [Indexed: 02/03/2025]
Abstract
A decade after the term 'trained immunity' (TRIM) was coined to reflect the long-lasting hyper-responsiveness of innate immune cells with an epigenetically imprinted 'memory' of earlier stimuli, our understanding has broadened to include the potential implications of TRIM in health and disease. Here, after summarizing the well-documented beneficial effects of TRIM against infections, we discuss emerging evidence that TRIM is also a major underlying mechanism in chronic inflammation-related disorders such as periodontitis, rheumatoid arthritis and cardiovascular disease. Furthermore, mounting evidence indicates that the induction of TRIM by certain agonists confers protective antitumour responses. Although the mechanisms underlying TRIM require further study, the current knowledge enables the experimental development of innovative therapeutic approaches to stimulate or inhibit TRIM in a context-appropriate manner, such as the stimulation of TRIM in cancer or its inhibition in inflammatory disorders.
Collapse
Affiliation(s)
- George Hajishengallis
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands.
- Department of Immunology and Metabolism, LIMES, University of Bonn, Bonn, Germany.
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
6
|
Cheng X, Sui H, Chen F, Li C, Du M, Zhang S, Chen J, Dou J, Huang Y, Xie X, Cheng C, Yang R, Yang C, Shi B, Shao D, Leong KW, Huang H. Nanomaterial-Mediated Reprogramming of Macrophages to Inhibit Refractory Muscle Fibrosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2410368. [PMID: 39548911 PMCID: PMC11849413 DOI: 10.1002/adma.202410368] [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: 07/17/2024] [Revised: 11/05/2024] [Indexed: 11/18/2024]
Abstract
Orofacial muscles are particularly prone to refractory fibrosis after injury, leading to a negative effect on the patient's quality of life and limited therapeutic options. Gaining insights into innate inflammatory response-fibrogenesis homeostasis can aid in the development of new therapeutic strategies for muscle fibrosis. In this study, the crucial role of macrophages is identified in the regulation of orofacial muscle fibrogenesis after injury. Hypothesizing that orchestrating macrophage polarization and functions will be beneficial for fibrosis treatment, nanomaterials are engineered with polyethylenimine functionalization to regulate the macrophage phenotype by capturing negatively charged cell-free nucleic acids (cfNAs). This cationic nanomaterial reduces macrophage-related inflammation in vitr and demonstrates excellent efficacy in preventing orofacial muscle fibrosis in vivo. Single-cell RNA sequencing reveals that the cationic nanomaterial reduces the proportion of profibrotic Gal3+ macrophages through the cfNA-mediated TLR7/9-NF-κB signaling pathway, resulting in a shift in profibrotic fibro-adipogenic progenitors (FAPs) from the matrix-producing Fabp4+ subcluster to the matrix-degrading Igf1+ subcluster. The study highlights a strategy to target innate inflammatory response-fibrogenesis homeostasis and suggests that cationic nanomaterials can be exploited for treating refractory fibrosis.
Collapse
Affiliation(s)
- Xu Cheng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hao Sui
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Fangman Chen
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Chenghao Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Meijun Du
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Shiming Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jiali Chen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jinfeng Dou
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yixuan Huang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiaochun Xie
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Chuanxu Cheng
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Renjie Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Eastern Clinic, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Chao Yang
- Department of Orthopedics, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, 510630, China
| | - Bing Shi
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Dan Shao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
- Department of Systems Biology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Hanyao Huang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| |
Collapse
|
7
|
Ren S, Fu X, Guo W, Bai R, Li S, Zhang T, Liu J, Wang Z, Zhao H, Suo S, Zhang W, Jia M, Ji W, Hu P, Chen Y. Profound cellular defects attribute to muscular pathogenesis in the rhesus monkey model of Duchenne muscular dystrophy. Cell 2024; 187:6669-6686.e16. [PMID: 39305903 DOI: 10.1016/j.cell.2024.08.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 05/29/2024] [Accepted: 08/20/2024] [Indexed: 11/17/2024]
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscle-wasting disease caused by mutations in the DMD gene. Muscle fibers rely on the coordination of multiple cell types for repair and regenerative capacity. To elucidate the cellular and molecular changes in these cell types under pathologic conditions, we generated a rhesus monkey model for DMD that displays progressive muscle deterioration and impaired motor function, mirroring human conditions. By leveraging these DMD monkeys, we analyzed freshly isolated muscle tissues using single-cell RNA sequencing (scRNA-seq). Our analysis revealed changes in immune cell landscape, a reversion of lineage progressing directions in fibrotic fibro-adipogenic progenitors (FAPs), and TGF-β resistance in FAPs and muscle stem cells (MuSCs). Furthermore, MuSCs displayed cell-intrinsic defects, leading to differentiation deficiencies. Our study provides important insights into the pathogenesis of DMD, offering a valuable model and dataset for further exploration of the underlying mechanisms, and serves as a suitable platform for developing and evaluating therapeutic interventions.
Collapse
Affiliation(s)
- Shuaiwei Ren
- State Key Laboratory of Primate Biomedical Research Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500 Kunming, China; Faculty of Life Science and Technology, Kunming University of Science and Technology, 650500 Kunming, China; Yunnan Key Laboratory of Primate Biomedical Research, 650500 Kunming, China
| | - Xin Fu
- Spine Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China
| | - Wenting Guo
- State Key Laboratory of Primate Biomedical Research Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500 Kunming, China; Yunnan Key Laboratory of Primate Biomedical Research, 650500 Kunming, China
| | - Raoxian Bai
- State Key Laboratory of Primate Biomedical Research Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500 Kunming, China; Faculty of Life Science and Technology, Kunming University of Science and Technology, 650500 Kunming, China; Yunnan Key Laboratory of Primate Biomedical Research, 650500 Kunming, China
| | - Sheng Li
- Spine Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China
| | - Ting Zhang
- State Key Laboratory of Primate Biomedical Research Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500 Kunming, China; Yunnan Key Laboratory of Primate Biomedical Research, 650500 Kunming, China; Southwest United Graduate School, 650092 Kunming, China
| | - Jie Liu
- State Key Laboratory of Primate Biomedical Research Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500 Kunming, China; Yunnan Key Laboratory of Primate Biomedical Research, 650500 Kunming, China
| | - Zhengbo Wang
- State Key Laboratory of Primate Biomedical Research Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500 Kunming, China; Yunnan Key Laboratory of Primate Biomedical Research, 650500 Kunming, China
| | - Hui Zhao
- Guangzhou Laboratory, 510005 Guangzhou, China; Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, 510005 Guangzhou, China
| | | | - Weikang Zhang
- Guangzhou Laboratory, 510005 Guangzhou, China; College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China
| | - Minzhi Jia
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 200031 Shanghai, China
| | - Weizhi Ji
- State Key Laboratory of Primate Biomedical Research Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500 Kunming, China; Yunnan Key Laboratory of Primate Biomedical Research, 650500 Kunming, China.
| | - Ping Hu
- Spine Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China; Guangzhou Laboratory, 510005 Guangzhou, China; Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, 510005 Guangzhou, China; The Tenth People's Hospital Affiliated to Tongji University, 200072 Shanghai, China.
| | - Yongchang Chen
- State Key Laboratory of Primate Biomedical Research Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500 Kunming, China; Faculty of Life Science and Technology, Kunming University of Science and Technology, 650500 Kunming, China; Yunnan Key Laboratory of Primate Biomedical Research, 650500 Kunming, China; Southwest United Graduate School, 650092 Kunming, China.
| |
Collapse
|
8
|
Bhargavi G, Subbian S. The causes and consequences of trained immunity in myeloid cells. Front Immunol 2024; 15:1365127. [PMID: 38665915 PMCID: PMC11043514 DOI: 10.3389/fimmu.2024.1365127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Conventionally, immunity in humans has been classified as innate and adaptive, with the concept that only the latter type has an immunological memory/recall response against specific antigens or pathogens. Recently, a new concept of trained immunity (a.k.a. innate memory response) has emerged. According to this concept, innate immune cells can exhibit enhanced responsiveness to subsequent challenges, after initial stimulation with antigen/pathogen. Thus, trained immunity enables the innate immune cells to respond robustly and non-specifically through exposure or re-exposure to antigens/infections or vaccines, providing enhanced resistance to unrelated pathogens or reduced infection severity. For example, individuals vaccinated with BCG to protect against tuberculosis were also protected from malaria and SARS-CoV-2 infections. Epigenetic modifications such as histone acetylation and metabolic reprogramming (e.g. shift towards glycolysis) and their inter-linked regulations are the key factors underpinning the immune activation of trained cells. The integrated metabolic and epigenetic rewiring generates sufficient metabolic intermediates, which is crucial to meet the energy demand required to produce proinflammatory and antimicrobial responses by the trained cells. These factors also determine the efficacy and durability of trained immunity. Importantly, the signaling pathways and regulatory molecules of trained immunity can be harnessed as potential targets for developing novel intervention strategies, such as better vaccines and immunotherapies against infectious (e.g., sepsis) and non-infectious (e.g., cancer) diseases. However, aberrant inflammation caused by inappropriate onset of trained immunity can lead to severe autoimmune pathological consequences, (e.g., systemic sclerosis and granulomatosis). In this review, we provide an overview of conventional innate and adaptive immunity and summarize various mechanistic factors associated with the onset and regulation of trained immunity, focusing on immunologic, metabolic, and epigenetic changes in myeloid cells. This review underscores the transformative potential of trained immunity in immunology, paving the way for developing novel therapeutic strategies for various infectious and non-infectious diseases that leverage innate immune memory.
Collapse
Affiliation(s)
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| |
Collapse
|
9
|
Dowling P, Trollet C, Negroni E, Swandulla D, Ohlendieck K. How Can Proteomics Help to Elucidate the Pathophysiological Crosstalk in Muscular Dystrophy and Associated Multi-System Dysfunction? Proteomes 2024; 12:4. [PMID: 38250815 PMCID: PMC10801633 DOI: 10.3390/proteomes12010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
This perspective article is concerned with the question of how proteomics, which is a core technique of systems biology that is deeply embedded in the multi-omics field of modern bioresearch, can help us better understand the molecular pathogenesis of complex diseases. As an illustrative example of a monogenetic disorder that primarily affects the neuromuscular system but is characterized by a plethora of multi-system pathophysiological alterations, the muscle-wasting disease Duchenne muscular dystrophy was examined. Recent achievements in the field of dystrophinopathy research are described with special reference to the proteome-wide complexity of neuromuscular changes and body-wide alterations/adaptations. Based on a description of the current applications of top-down versus bottom-up proteomic approaches and their technical challenges, future systems biological approaches are outlined. The envisaged holistic and integromic bioanalysis would encompass the integration of diverse omics-type studies including inter- and intra-proteomics as the core disciplines for systematic protein evaluations, with sophisticated biomolecular analyses, including physiology, molecular biology, biochemistry and histochemistry. Integrated proteomic findings promise to be instrumental in improving our detailed knowledge of pathogenic mechanisms and multi-system dysfunction, widening the available biomarker signature of dystrophinopathy for improved diagnostic/prognostic procedures, and advancing the identification of novel therapeutic targets to treat Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland;
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
| | - Capucine Trollet
- Center for Research in Myology U974, Sorbonne Université, INSERM, Myology Institute, 75013 Paris, France; (C.T.); (E.N.)
| | - Elisa Negroni
- Center for Research in Myology U974, Sorbonne Université, INSERM, Myology Institute, 75013 Paris, France; (C.T.); (E.N.)
| | - Dieter Swandulla
- Institute of Physiology, Faculty of Medicine, University of Bonn, D53115 Bonn, Germany;
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland;
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
| |
Collapse
|