1
|
Wu G, Su T, Zhou P, Tang R, Zhu X, Wang J, Chao M, Fan L, Yan H, Ye P, Yu D, Gao F, Chen H. Engineering M2 macrophage-derived exosomes modulate activated T cell cuproptosis to promote immune tolerance in rheumatoid arthritis. Biomaterials 2025; 315:122943. [PMID: 39509857 DOI: 10.1016/j.biomaterials.2024.122943] [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: 09/11/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/15/2024]
Abstract
Nanomedicines for immune modulation have made advancements in the treatment of rheumatoid arthritis (RA). However, due to aberrations in patients' immune systems, inducing antigen-specific immune tolerance while halting disease progression remains a significant challenge. Here, we develop a highly targeted multifunctional nanocomplex, termed M2Exo@CuS-CitP-Rapa (M2CPR), with the aim of selectively inhibiting inflammatory immune reactions while promoting immune tolerance towards specific antigens. M2CPR specifically targets inflammatory tissues in RA, delivering CuS NPs, CitP, Rapa, and endogenous anti-inflammatory factors, thereby ameliorating the inflammatory joint microenvironment. CuS NPs induce Cuproptosis of activated T cells, whose fragments are engulfed by resident or recruited macrophages, resulting in abundant production of TGF-β. TGF-β acts synergistically with Rapa to induce the iDCs into tDCs. tDCs present CitP to Naive T cells, promoting Tregs differentiation. Tregs, in turn, produce more TGF-β, inducing tDCs differentiation, thereby establishing a cycle of immune tolerance. Through in vitro and in vivo experiments, we validate that M2CPR can induce robust and durable antigen-specific immune tolerance, offering a new paradigm for RA therapy.
Collapse
Affiliation(s)
- Guoquan Wu
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Tianyu Su
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Peng Zhou
- Department of Orthopedics, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huai'an, 223002, China
| | - Rongze Tang
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Xu Zhu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Jin Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Minghao Chao
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Liying Fan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Hanrong Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Peng Ye
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Dehong Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Fenglei Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| | - Hongliang Chen
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China.
| |
Collapse
|
2
|
Lu L, Qi Z, Wang H, Chen Z, Song Z, Li Z, Wang X, Zhao B, Wei X, Shao Y, Wang Z, Tu J, Song X. The Hcp2b of APEC induces mitochondrial damage in chicken DF-1 cells. Avian Pathol 2024:1-9. [PMID: 39552458 DOI: 10.1080/03079457.2024.2431803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/15/2024] [Accepted: 10/19/2024] [Indexed: 11/19/2024]
Abstract
The haemolysin co-regulatory protein (Hcp) plays a significant role in the pathogenicity of avian pathogenic Escherichia coli (APEC) as an effector protein of the type VI secretion system (T6SS) to the host. Meanwhile, mitochondria in the host are the target of effector proteins of various secretion systems. Here, we explored the effects of APEC effector Hcp2b on the mitochondria of DF-1 cells and found that Hcp2b results in damage in mitochondria. Next, 68 target proteins in DF-1 cell lysates were identified that interacted with Hcp2b by streptavidin-biotin pull-down assay combined with LC-MS/MS, among which ADP/ATP transporter carrier (SLC25A4) is a mitochondria-associated protein; protein docking analysis showed that Hcp2b binds well to SLC25A4. Therefore, we hypothesize that the Hcp2b contributes to mitochondrial damage in DF-1 cells through interaction with the SLC25A4.
Collapse
Affiliation(s)
- Liting Lu
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
| | - Zhao Qi
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
| | - Haiyang Wang
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
| | - Zhe Chen
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
| | - Zichao Song
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
| | - Ziqi Li
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
| | - Xiaoru Wang
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
| | - Bingyu Zhao
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
| | - Xiyang Wei
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
| | - Ying Shao
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
| | - Zhenyu Wang
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
| | - Jian Tu
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
| | - Xiangjun Song
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei, People's Republic of China
| |
Collapse
|
3
|
Tan YN, Jiang GG, Meng XW, Lu ZY, Yan-Ma, Li J, Nan-Xiang, Sun XG, Wang Q, Wang X, Jia XY, Zhang M. CMPK2 Promotes CD4 + T Cell Activation and Apoptosis through Modulation of Mitochondrial Dysfunction in Systemic Lupus Erythematosus. Cell Biochem Biophys 2024; 82:3547-3557. [PMID: 39078538 DOI: 10.1007/s12013-024-01443-1] [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] [Accepted: 07/16/2024] [Indexed: 07/31/2024]
Abstract
Systemic lupus erythematosus (SLE) is a classic autoimmune disease characterized by abnormal autoantibodies, immune complex deposition, and tissue inflammation. Despite extensive research, the exact etiology and progression of SLE remain elusive. Cytidine/uridine monophosphate kinase 2 (CMPK2), a mitochondrial nucleoside monophosphate kinase, has garnered attention for its potential involvement in the development of various diseases, including SLE, where it has been observed to be dysregulated in affected individuals. However, the specific involvement of CMPK2 in the pathogenesis of SLE remains unclear. This study aims to clarify the expression level of CMPK2 in SLE CD4+ T cells and explore its impact on CD4+ T cells. The expression levels of the CMPK2 gene and the corresponding CMPK2 protein in CD4+ T cells of SLE patients were quantified using RT-qPCR and Western blot, respectively. Immunofluorescence and RT-qPCR were used to assess the mitochondrial function of SLE CD4+ T cells. Flow cytometry was used to assess CD4+ T cell activation and apoptosis levels. The impact of CMPK2 on CD4+ T cells was investigated by gene transfection experiment. We found that CMPK2 was significantly upregulated in SLE CD4+ T cells at both gene and protein levels. These cells demonstrated aberrant mitochondrial function, as evidenced by elevated mitochondrial reactive oxygen species (mtROS) levels, mitochondrial membrane potential, and mitochondrial DNA (mtDNA) copy number. Flow cytometry revealed a notable increase in both apoptosis and activation levels of CD4+ T cells in SLE patients. Gene transfection experiments showed that suppressing CMPK2 led to a significant improvement in these conditions. These findings suggest that CMPK2 may be involved in the pathogenesis of SLE by regulating mitochondrial dysfunction in CD4+ T cells and thus affecting CD4+ T cell activation and apoptosis. Our study may provide a new target for the treatment of SLE.
Collapse
Affiliation(s)
- Ya-Nan Tan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, PR China
- Division of Life Sciences and Medicine, Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, PR China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, PR China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, PR China
| | - Ge-Ge Jiang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, PR China
- Division of Life Sciences and Medicine, Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, PR China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, PR China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, PR China
| | - Xiang-Wen Meng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, PR China
- Division of Life Sciences and Medicine, Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, PR China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, PR China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, PR China
| | - Zhi-Yuan Lu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, PR China
- Division of Life Sciences and Medicine, Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, PR China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, PR China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, PR China
| | - Yan-Ma
- Division of Life Sciences and Medicine, Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, PR China
| | - Jin Li
- Division of Life Sciences and Medicine, Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, PR China
| | - Nan-Xiang
- Division of Life Sciences and Medicine, Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, PR China
| | - Xiao-Ge Sun
- Division of Life Sciences and Medicine, Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, PR China
| | - Qian Wang
- Division of Life Sciences and Medicine, Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, PR China
| | - Xue Wang
- Division of Life Sciences and Medicine, Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, PR China
| | - Xiao-Yi Jia
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, PR China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, PR China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, PR China
| | - Min Zhang
- Division of Life Sciences and Medicine, Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, PR China.
| |
Collapse
|
4
|
Nedel W, Strogulski NR, Kopczynski A, Portela LV. Assessment of mitochondrial function and its prognostic role in sepsis: a literature review. Intensive Care Med Exp 2024; 12:107. [PMID: 39585590 PMCID: PMC11589057 DOI: 10.1186/s40635-024-00694-9] [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: 08/01/2024] [Accepted: 11/08/2024] [Indexed: 11/26/2024] Open
Abstract
Sepsis is characterized by a dysregulated and excessive systemic inflammatory response to infection, associated with vascular and metabolic abnormalities that ultimately lead to organ dysfunction. In immune cells, both non-oxidative and oxidative metabolic rates are closely linked to inflammatory responses. Mitochondria play a central role in supporting these cellular processes by utilizing metabolic substrates and synthesizing ATP through oxygen consumption. To meet fluctuating cellular demands, mitochondria must exhibit adaptive plasticity underlying bioenergetic capacity, biogenesis, fusion, and fission. Given their role as a hub for various cellular functions, mitochondrial alterations induced by sepsis may hold significant pathophysiological implications and impact on clinical outcomes. In patients, mitochondrial DNA concentration, protein expression levels, and bioenergetic profiles can be accessed via tissue biopsies or isolated peripheral blood cells. Clinically, monocytes and lymphocytes serve as promising matrices for evaluating mitochondrial function. These mononuclear cells are highly oxidative, mitochondria-rich, routinely monitored in blood, easy to collect and process, and show a clinical association with immune status. Hence, mitochondrial assessments in immune cells could serve as biomarkers for clinical recovery, immunometabolic status, and responsiveness to oxygen and vasopressor therapies in sepsis. These characteristics underscore mitochondrial parameters in both tissues and immune cells as practical tools for exploring underlying mechanisms and monitoring septic patients in intensive care settings. In this article, we examine pathophysiological aspects, key methods for measuring mitochondrial function, and prominent studies in this field.
Collapse
Affiliation(s)
- Wagner Nedel
- Intensive Care Unit, Grupo Hospitalar Conceição (GHC), Porto Alegre, Brazil.
- Laboratory of Neurotrauma and Biomarkers, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.
- Unidade de Terapia Intensiva, Hospital Nossa Senhora da Conceição, Av Francisco Trein, 596-primeiro andar, Porto Alegre, RS, Brazil.
| | - Nathan Ryzewski Strogulski
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, University of Dublin, Dublin, Ireland
| | - Afonso Kopczynski
- Laboratory of Neurotrauma and Biomarkers, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Programa de Pós-Graduação Em Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Luis Valmor Portela
- Laboratory of Neurotrauma and Biomarkers, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Programa de Pós-Graduação Em Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| |
Collapse
|
5
|
Abad-Jiménez Z, López-Domènech S, Pelechá M, Perea-Galera L, Rovira-Llopis S, Bañuls C, Blas-García A, Apostolova N, Morillas C, Víctor VM, Rocha M. Calorie restriction modulates mitochondrial dynamics and autophagy in leukocytes of patients with obesity. Free Radic Biol Med 2024; 225:677-686. [PMID: 39447993 DOI: 10.1016/j.freeradbiomed.2024.10.295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/13/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Although it is established that caloric restriction offers metabolic and clinical benefits, the molecular mechanisms underlying these effects remain unclear. Thus, this study aimed to investigate whether caloric restriction can modulate mitochondrial function and remodeling and stimulate autophagic flux in the PBMCs of patients with obesity. METHODS This was an interventional study of 38 obese subjects (BMI >35 kg/m2) who underwent 6 months of dietary therapy, including a 6-week very-low-calorie diet (VLCD) followed by an 18-week low-calorie diet (LCD). We determined clinical variables, mitochondrial function parameters (by fluorescence imaging of mitochondrial ROS and membrane potential), and protein expression of markers of mitochondrial dynamics (MNF1, MFN2, OPA, DRP1 and FIS1) and autophagy (LC3, Beclin, BCL2 and NBR1) by Western blot. RESULTS Caloric restriction induced an improvement in metabolic outcomes that was accompanied by an increase in AMPK expression, a decrease of mitochondrial ROS and mitochondrial membrane potential, which was associated with increased markers of mitochondrial dynamics (MFN2, DRP1 and FIS1) and activation of autophagy as evidenced by augmented LC3 II/I, Beclin1 and NBR1, and a decrease in BCL2. CONCLUSION These findings shed light on the specific molecular mechanisms by which caloric restriction facilitates metabolic improvements, highlighting the relevance of pathways involving energy homeostasis and cell recovery, including mitochondrial function and dynamics and autophagy.
Collapse
Affiliation(s)
- Zaida Abad-Jiménez
- Department of Endocrinology and Nutrition University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research (FISABIO), 46017, Valencia, Spain
| | - Sandra López-Domènech
- Department of Endocrinology and Nutrition University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research (FISABIO), 46017, Valencia, Spain
| | - María Pelechá
- Department of Endocrinology and Nutrition University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research (FISABIO), 46017, Valencia, Spain
| | - Laura Perea-Galera
- Department of Endocrinology and Nutrition University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research (FISABIO), 46017, Valencia, Spain
| | - Susana Rovira-Llopis
- Department of Physiology, Faculty of Medicine & Dentistry, University of Valencia, 46010, Valencia, Spain
| | - Celia Bañuls
- Department of Endocrinology and Nutrition University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research (FISABIO), 46017, Valencia, Spain
| | - Ana Blas-García
- CIBEREHD (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), 28029, Madrid, Spain; Department of Pharmacology, Faculty of Medicine & Dentistry, University of Valencia, 46010, Valencia, Spain
| | - Nadezda Apostolova
- CIBEREHD (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), 28029, Madrid, Spain; Department of Pharmacology, Faculty of Medicine & Dentistry, University of Valencia, 46010, Valencia, Spain
| | - Carlos Morillas
- Department of Endocrinology and Nutrition University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research (FISABIO), 46017, Valencia, Spain
| | - Víctor Manuel Víctor
- Department of Physiology, Faculty of Medicine & Dentistry, University of Valencia, 46010, Valencia, Spain; CIBEREHD (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), 28029, Madrid, Spain.
| | - Milagros Rocha
- Department of Endocrinology and Nutrition University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research (FISABIO), 46017, Valencia, Spain; CIBEREHD (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), 28029, Madrid, Spain.
| |
Collapse
|
6
|
Wang G, Liu S, Fan X, Li J, Xue Q, Liu K, Li X, Yang Y, Wang X, Song M, Shao M, Li W, Han Y, Lv L, Su X. Mitochondrial Dysfunction and Cognitive Impairment in Schizophrenia: The Role of Inflammation. Schizophr Bull 2024:sbae196. [PMID: 39535935 DOI: 10.1093/schbul/sbae196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
BACKGROUND AND HYPOTHESIS The complex immune-brain interactions and the regulatory role of mitochondria in the immune response suggest that mitochondrial damage reported in schizophrenia (SZ) may be related to abnormalities observed in immune and brain functions. STUDY DESIGN Mitochondrial DNA copy number (mtDNA CN), a biomarker of mitochondrial function, was assessed in peripheral blood leukocytes (PBLs) of 121 healthy individuals and 118 SZ patients before and after 8 weeks of antipsychotic treatment, and a meta-analysis related to blood mtDNA CN was conducted. Plasma C-reactive protein (CRP) levels in SZ patients were obtained from the medical record system. Spearman correlation analysis and hierarchical linear regression were used to analyze the relationships among mtDNA CN, CRP levels, and cognitive function. A mediation model was constructed using the PROCESS program. STUDY RESULTS Our results revealed the decreased mtDNA CN in PBLs from SZ patients (P = .05). The meta-analysis supported the decreased blood mtDNA CN in SZ patients (P < .01). The mtDNA CN in PBL was positively correlated with working memory (P = .02) and negatively correlated with plasma CRP levels (P = .039). Furthermore, a lower mtDNA CN in PBL in SZ patients was a significant predictor of worse working memory (P = .006). CRP acted as a mediator with an 8.0% effect. CONCLUSIONS This study revealed an association between peripheral mitochondrial dysfunction and cognitive impairment in SZ, with inflammation acting as a mediating effect. Therefore, mitochondrial dysfunction might provide novel targets for new treatments for cognitive impairment in SZ.
Collapse
Affiliation(s)
- Guanyu Wang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang 453002, China
| | - Senqi Liu
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang 453002, China
| | - Xiaoyun Fan
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang 453002, China
| | - Jinming Li
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang 453002, China
| | - Qianzi Xue
- The Second Clinical College of Xinxiang Medical University, Xinxiang 453003, China
| | - Kang Liu
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang 453002, China
| | - Xue Li
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang 453002, China
| | - Yongfeng Yang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang 453002, China
| | - Xiujuan Wang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang 453002, China
| | - Meng Song
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang 453002, China
| | - Minglong Shao
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang 453002, China
| | - Wenqiang Li
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang 453002, China
| | - Yong Han
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang 453002, China
| | - Luxian Lv
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang 453002, China
| | - Xi Su
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang 453002, China
| |
Collapse
|
7
|
Li X, Ma Q, Huang Y, Cheng L, Liu Y, Li H, Zhan H, Zhang F, Liu Y, Li Y. Increase in Mitochondrial Mass of Lymphocyte Subsets in Anti-MDA5 and TIF1-γ-Positive Dermatomyositis Patients. Immunol Invest 2024; 53:1381-1396. [PMID: 39291775 DOI: 10.1080/08820139.2024.2402824] [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: 09/19/2024]
Abstract
OBJECTIVES The mitochondrial function in anti-MDA5 and TIF1-γ-positive dermatomyositis (DM) is relatively unknown. This study attempted to explore mitochondrial mass within the peripheral lymphocyte subsets of anti-MDA5 and TIF1-γ-positive DM. METHODS This cross-sectional study enrolled 109 DM patients and 32 healthy controls (HCs). The mitochondrial mass of peripheral lymphocyte subsets was analyzed via flow cytometry using median fluorescence intensity assessment. RESULTS Compared with HCs, there was an abnormal change in peripheral lymphocyte subsets in anti-MDA5 and anti-TIF1-γ-positive DM patients. Anti-MDA5 and anti-TIF1-γ-positive DM patients also exhibited a significantly elevated mitochondrial mass in peripheral lymphocyte subsets. Furthermore, anti-MDA5 antibody levels were positively associated with the mitochondrial mass of most lymphocyte subsets in anti-MDA5-positive DM patients. Univariate logistic regression analysis indicated that the increased mitochondrial mass in some peripheral lymphocyte subsets was related to the occurrence of anti-MDA5-positive DM and presence of anti-MDA5 antibodies. Similar results were obtained in anti-TIF1-γ-positive DM patients. CONCLUSIONS Abnormal lymphocyte subset counts and percentages as well as altered mitochondrial mass in anti-MDA5 and TIF1-γ-positive DM patients were associated with anti-MDA5 and TIF1-γ antibodies. We believe that these results may provide novel mitochondria-based insights into DM pathogenesis.
Collapse
Affiliation(s)
- Xiaomeng Li
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Qingqing Ma
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yuan Huang
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Linlin Cheng
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yongmei Liu
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Haolong Li
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Haoting Zhan
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Fengchun Zhang
- Department of Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yudong Liu
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
| | - Yongzhe Li
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
8
|
Lu M, Li W, Zhou J, Shang J, Lin L, Liu Y, Zhu X. Integrative bioinformatics analysis for identifying the mitochondrial-related gene signature associated with immune infiltration in premature ovarian insufficiency. BMC Med 2024; 22:444. [PMID: 39379953 PMCID: PMC11462806 DOI: 10.1186/s12916-024-03675-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/01/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Premature ovarian insufficiency (POI) is a reproductive disorder characterized by the cessation of ovarian function before the age of 40. Although mitochondrial dysfunction and immune disorders are believed to contribute to ovarian damage in POI, the interplay between these factors remains understudied. METHODS In this research, transcriptomic data related to POI were obtained from the NCBI GEO database. Hub biomarkers were identified through the construction of a protein‒protein interaction (PPI) network and further validated using RT‒qPCR and Western blot. Moreover, their expression across various cell types was elucidated via single-cell RNA sequencing analysis. A comprehensive investigation of the mitochondrial and immune profiles of POI was carried out through correlation analysis. Furthermore, potential therapeutic agents were predicted utilizing the cMap database. RESULTS A total of 119 mitochondria-related differentially expressed genes (MitoDEGs) were identified and shown to be significantly enriched in metabolic pathways. Among these genes, Hadhb, Cpt1a, Mrpl12, and Mrps7 were confirmed both in a POI model and in human granulosa cells (GCs), where they were found to accumulate in GCs and theca cells. Immune analysis revealed variations in macrophages, monocytes, and 15 other immune cell types between the POI and control groups. Notably, strong correlations were observed between seven hub-MitoDEGs (Hadhb, Cpt1a, Cpt2, Mrpl12, Mrps7, Mrpl51, and Eci1) and various functions, such as mitochondrial respiratory complexes, dynamics, mitophagy, mitochondrial metabolism, immune-related genes, and immunocytes. Additionally, nine potential drugs (calyculin, amodiaquine, eudesmic acid, cefotaxime, BX-912, prostratin, SCH-79797, HU-211, and pizotifen) targeting key genes were identified. CONCLUSIONS Our results highlight the crosstalk between mitochondrial function and the immune response in the development of POI. The identification of MitoDEGs could lead to reliable biomarkers for the early diagnosis, monitoring, and personalized treatment of POI.
Collapse
Affiliation(s)
- Minjun Lu
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
- Department of Central Laboratory, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
| | - Wenxin Li
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
- Department of Central Laboratory, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
| | - Jiamin Zhou
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
- Department of Central Laboratory, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
| | - Junyu Shang
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
- Department of Central Laboratory, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
| | - Li Lin
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
- Department of Central Laboratory, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
| | - Yueqin Liu
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
| | - Xiaolan Zhu
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China.
| |
Collapse
|
9
|
Zhang L, Liu K, Liu Z, Tao H, Fu X, Hou J, Jia G, Hou Y. In pre-clinical study fetal hypoxia caused autophagy and mitochondrial impairment in ovary granulosa cells mitigated by melatonin supplement. J Adv Res 2024; 64:15-30. [PMID: 37956860 PMCID: PMC11464463 DOI: 10.1016/j.jare.2023.11.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 11/15/2023] Open
Abstract
INTRODUCTION Fetal hypoxia has long-term effects on postnatal reproductive functions and the mitochondrial impairments of ovarian granulosa cells may be one of the causes. Melatonin applied to mitigate mitochondrial dysfunction and autophagy in mammalian cells has been reported. However, the potential mechanisms by which fetal hypoxia damages reproductive function in neonatal female mice and the melatonin effects on this problem remain unclear. OBJECTIVES This research aimed to explore the mechanism that fetal hypoxia damages reproductive function in neonatal female mice and attempt to improve the reproductive function by treating with melatonin in vivo and in vitro. METHODS We established a fetal hypoxia model and confirmed that fetal hypoxia affects ovarian function by inducing GC excessive autophagy. Transcriptomic analysis, gene interference, cell immunofluorescence, immunohistochemistry and western blot were conducted to explore and verify the underlying mechanisms in mice GCs and KGN cells. Finally, melatonin treatment was executed on hypoxia-treated mice GCs and KGN cells and melatonin injection to fetal-hypoxia-treated mice to determine its effect. RESULTS The results of in vitro experiments found that fetal hypoxia led to mitochondrial dysfunction in ovarian GCs causing autophagic cell death. And the PI3K/Akt/FoxO pathway mediated the occurrence of this process by transcriptome analysis of ovarian GCs from normal and fetal hypoxia mice, which was further verified in mice GCs and KGN cells. Additionally, melatonin administration prevented autophagic injuries and mitochondrial impairments in hypoxia-treated mice GCs and KGN cells. Meanwhile, in vivo experiments by melatonin injection ameliorated oxidative stress of ovary in fetal-hypoxia-treated mice and improved their low fertility. CONCLUSION Our data found that fetal hypoxia causes ovarian GCs excessive autophagy leading to low fertility in neonatal female mice and mitigated by melatonin. These results provide a potential therapy for hypoxic stress-related reproductive disorders.
Collapse
Affiliation(s)
- Luyao Zhang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China; Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, China
| | - Kexiong Liu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Zhiqiang Liu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Haiping Tao
- University of Chinese Academy of Sciences, Beijing 100049, China; Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, China
| | - Xiangwei Fu
- National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China; State Key Laboratory of Sheep Genetic Improvement and Healthy Breeding, Xinjiang Academy of Agricultural and Reclamation Sciences, Shihezi, China
| | - Jian Hou
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Gongxue Jia
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, China; University of Chinese Academy of Sciences, Beijing 100049, China; Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, China
| | - Yunpeng Hou
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China.
| |
Collapse
|
10
|
Ouyang W, Yan D, Hu J, Liu Z. Multifaceted mitochondrial as a novel therapeutic target in dry eye: insights and interventions. Cell Death Discov 2024; 10:398. [PMID: 39242592 PMCID: PMC11379830 DOI: 10.1038/s41420-024-02159-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/09/2024] Open
Abstract
Dry eye, recognized as the most prevalent ocular surface disorder, has risen to prominence as a significant public health issue, adversely impacting the quality of life for individuals across the globe. Despite decades of extensive research into the chronic inflammation that characterizes dry eye, the intricate mechanisms fueling this persistent inflammatory state remain incompletely understood. Among the various cellular components under investigation, mitochondria-essential for cellular energy production and homeostasis-have attracted increasing attention for their role in dry eye pathogenesis. This involvement points to mechanisms such as oxidative stress, apoptosis, and sustained inflammation, which are central to the progression of the disease. This review aims to provide a thorough exploration of mitochondrial dysfunction in dry eye, shedding light on the critical roles played by mitochondrial oxidative stress, apoptosis, and mitochondrial DNA damage. It delves into the mechanisms through which diverse pathogenic factors may trigger mitochondrial dysfunction, thereby contributing to the onset and exacerbation of dry eye. Furthermore, it lays the groundwork for an overview of current therapeutic strategies that specifically target mitochondrial dysfunction, underscoring their potential in managing this complex condition. By spotlighting this burgeoning area of research, our review seeks to catalyze the development of innovative drug discovery and therapeutic approaches. The ultimate goal is to unlock promising avenues for the future management of dry eye, potentially revolutionizing treatment paradigms and improving patient outcomes. Through this comprehensive examination, we endeavor to enrich the scientific community's understanding of dry eye and inspire novel interventions that address the underlying mitochondrial dysfunctions contributing to this widespread disorder.
Collapse
Affiliation(s)
- Weijie Ouyang
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine of Xiamen University, Xiamen, Fujian, China; Department of Ophthalmology, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Dan Yan
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine of Xiamen University, Xiamen, Fujian, China
| | - Jiaoyue Hu
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine of Xiamen University, Department of Ophthalmology of Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China.
| | - Zuguo Liu
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine of Xiamen University, Department of Ophthalmology of Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China; Department of Ophthalmology, the First Affiliated Hospital of University of South China, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
11
|
Gao Y, Liu H, Zhou Y, Cai S, Zhang J, Sun J, Duan M. Cold inducible RNA binding protein-regulated mitochondria associated endoplasmic reticulum membranes-mediated Ca 2+ transport play a critical role in hypothermia cerebral resuscitation. Exp Neurol 2024; 379:114883. [PMID: 38992825 DOI: 10.1016/j.expneurol.2024.114883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024]
Abstract
Cardiac arrest is a global health issue causing more deaths than many other diseases. Hypothermia therapy is commonly used to treat secondary brain injury resulting from cardiac arrest. Previous studies have shown that CIRP is induced in specific brain regions during hypothermia and inhibits mitochondrial apoptotic factors. However, the specific mechanisms by which hypothermia-induced CIRP exerts its anti-apoptotic effect are still unknown. This study aims to investigate the role of Cold-inducible RNA-binding protein (CIRP) in mitochondrial-associated endoplasmic reticulum membrane (MAM)-mediated Ca2+ transport during hypothermic brain resuscitation.We constructed a rat model of cardiac arrest and resuscitation and hippocampal neuron oxygen-glucose deprivation/reoxygenation model. We utilized shRNA transfection to interfere the expression of CIRP and observe the effect of CIRP on the structure and function of MAM.Hypothermia induced CIRP can reduce the apoptosis of hippocampal neurons, and improve the survival rate of rats. Hypothermia induced CIRP can reduce the expressions of calcium transporters IP3R and VDAC1 in MAM, reduce the concentration of calcium in mitochondria, decrease the expression of ROS, and stabilize the mitochondrial membrane potential. Immunofluorescence and immunocoprecipitation showed that CIRP could directly interact with IP3R-VDAC1 complex, thereby changing the structure of MAM, inhibiting calcium transportation and improving mitochondrial function in vivo and vitro.Both in vivo and in vitro experiments have confirmed that hypothermia induced CIRP can act on the calcium channel IP3R-VDAC1 in MAM, reduce the calcium overload in mitochondria, improve the energy metabolism of mitochondria, and thus play a role in neuron resuscitation. This study contributes to understanding hypothermia therapy and identifies potential targets for brain injury treatment.
Collapse
Affiliation(s)
- Yu Gao
- Department of Anesthesiology, Zhongda Hospital Southeast University, Nanjing 210000, Jiangsu, China
| | - Haoxin Liu
- Department of Anesthesiology, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, China
| | - Yaqing Zhou
- Department of Pain Management, Affiliated Hospital of Jiangnan University, Wuxi 214000, Jiangsu, China
| | - Shenquan Cai
- Department of Anesthesiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210000, Jiangsu, China
| | - Jie Zhang
- Department of Anesthesiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210000, Jiangsu, China
| | - Jie Sun
- Department of Anesthesiology, Zhongda Hospital Southeast University, Nanjing 210000, Jiangsu, China.
| | - Manlin Duan
- Department of Anesthesiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210000, Jiangsu, China; Department of Anesthesiology, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, China.
| |
Collapse
|
12
|
Soliman Wadan AH, Abdelsattar Ahmed M, Hussein Ahmed A, El-Sayed Ellakwa D, Hamed Elmoghazy N, Gawish A. The Interplay of Mitochondrial Dysfunction in Oral Diseases: Recent Updates in Pathogenesis and Therapeutic Implications. Mitochondrion 2024; 78:101942. [PMID: 39111357 DOI: 10.1016/j.mito.2024.101942] [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: 04/05/2024] [Revised: 07/12/2024] [Accepted: 08/03/2024] [Indexed: 08/15/2024]
Abstract
Mitochondrial dysfunction is linked to various systemic and localized diseases, including oral diseases like periodontitis, oral cancer, and temporomandibular joint disorders. This paper explores the intricate mechanisms underlying mitochondrial dysfunction in oral pathologies, encompassing oxidative stress, inflammation, and impaired energy metabolism. Furthermore, it elucidates the bidirectional relationship between mitochondrial dysfunction and oral diseases, wherein the compromised mitochondrial function exacerbates disease progression, while oral pathologies, in turn, exacerbate mitochondrial dysfunction. Understanding these intricate interactions offers insights into novel therapeutic strategies targeting mitochondrial function for managing oral diseases. This paper pertains to the mechanisms underlying mitochondrial dysfunction, its implications in various oral pathological and inflammatory conditions, and emerging versatile treatment approaches. It reviews current therapeutic strategies to mitigate mitochondrial dysfunction, including antioxidants, mitochondrial-targeted agents, and metabolic modulators.
Collapse
Affiliation(s)
- Al-Hassan Soliman Wadan
- Faculty of Dentistry, Sinai University, Arish Branch, North Sinai, Egypt; Sinai University Research Center (SURC), Sinai University, North Sinai, Egypt.
| | - Mohamed Abdelsattar Ahmed
- Faculty of Dentistry, Sinai University, Kantra Branch, Ismailia, Egypt; Sinai University Research Center (SURC), Sinai University, North Sinai, Egypt
| | - Abdelnaser Hussein Ahmed
- Faculty of Dentistry, Sinai University, Arish Branch, North Sinai, Egypt; Sinai University Research Center (SURC), Sinai University, North Sinai, Egypt
| | - Doha El-Sayed Ellakwa
- Department of Biochemistry & Molecular Biology, Faculty of Pharmacy for Girls, Al-Azhar University, Cairo, Egypt; Department of Biochemistry, Faculty of Pharmacy, Sinai University, Kantra Branch, Ismailia, Egypt
| | - Nourhan Hamed Elmoghazy
- Faculty of Dentistry, Sinai University, Arish Branch, North Sinai, Egypt; Sinai University Research Center (SURC), Sinai University, North Sinai, Egypt
| | - Abeer Gawish
- Faculty of Dentistry, Sinai University, Arish Branch, North Sinai, Egypt; Sinai University Research Center (SURC), Sinai University, North Sinai, Egypt; Faculty of Graduate Studies, Sinai University, Arish Branche, North Sinai, Egypt; Oral Medicine, Periodontology, Diagnosis and Radiology Department, Al Azhar University, Egypt
| |
Collapse
|
13
|
Qiao YS, Blackwell TL, Cawthon PM, Coen PM, Cummings SR, Distefano G, Farsijani S, Forman DE, Goodpaster BH, Kritchevsky SB, Mau T, Toledo FGS, Newman AB, Glynn NW. Associations of accelerometry-measured and self-reported physical activity and sedentary behavior with skeletal muscle energetics: The Study of Muscle, Mobility and Aging (SOMMA). JOURNAL OF SPORT AND HEALTH SCIENCE 2024; 13:621-630. [PMID: 38341136 PMCID: PMC11282341 DOI: 10.1016/j.jshs.2024.02.001] [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: 10/18/2023] [Revised: 11/25/2023] [Accepted: 01/08/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Skeletal muscle energetics decline with age, and physical activity (PA) has been shown to offset these declines in older adults. Yet, many studies reporting these effects were based on self-reported PA or structured exercise interventions. Therefore, we examined the associations of accelerometry-measured and self-reported PA and sedentary behavior (SB) with skeletal muscle energetics and explored the extent to which PA and sedentary behavior would attenuate the associations of age with muscle energetics. METHODS As part of the Study of Muscle, Mobility and Aging, enrolled older adults (n = 879), 810 (age = 76.4 ± 5.0 years old, mean ± SD; 58% women) had maximal muscle oxidative capacity measured ex vivo via high-resolution respirometry of permeabilized myofibers (maximal oxidative phosphorylation (maxOXPHOS)) and in vivo by 31phosphorus magnetic resonance spectroscopy (maximal adenosine triphosphate (ATPmax)). Accelerometry-measured sedentary behavior, light activity, and moderate-to-vigorous PA (MVPA) were assessed using a wrist-worn ActiGraph GT9X over 7 days. Self-reported sedentary behavior, MVPA, and all PA were assessed with the Community Healthy Activities Model Program for Seniors (CHAMPS) questionnaire. Linear regression models with progressive covariate adjustments evaluated the associations of sedentary behavior and PA with muscle energetics, as well as the attenuation of the age/muscle energetics association by MVPA and sedentary behavior. As a sensitivity analysis, we also examined activPAL-measured daily step count and time spent in sedentary behavior and their associations with muscle energetics. RESULTS Every 30 min/day more of ActiGraph-measured MVPA was associated with 0.65 pmol/(s × mg) higher maxOXPHOS and 0.012 mM/s higher ATPmax after adjusting for age, site/technician, and sex (p < 0.05). Light activity was not associated with maxOXPHOS or ATPmax. Meanwhile, every 30 min/day spent in ActiGraph-measured sedentary behavior was associated with 0.39 pmol/s × mg lower maxOXPHOS and 0.006 mM/s lower ATPmax (p < 0.05). Only associations with ATPmax held after further adjusting for socioeconomic status, body mass index, lifestyle factors, and multimorbidity. CHAMPS MVPA and all PA yielded similar associations with maxOXPHOS and ATPmax (p < 0.05), but sedentary behavior did not. Higher activPAL step count was associated with higher maxOXHPOS and ATPmax (p < 0.05), but time spent in sedentary behavior was not. Additionally, age was significantly associated with muscle energetics for men only (p < 0.05); adjusting for time spent in ActiGraph-measured MVPA attenuated the age association with ATPmax by 58% in men. CONCLUSION More time spent in accelerometry-measured or self-reported daily PA, especially MVPA, was associated with higher skeletal muscle energetics. Interventions aimed specifically at increasing higher intensity activity might offer potential therapeutic interventions to slow age-related decline in muscle energetics. Our work also emphasizes the importance of taking PA into consideration when evaluating associations related to skeletal muscle energetics.
Collapse
Affiliation(s)
- Yujia Susanna Qiao
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA; San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA 94143, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, CA 94143, USA
| | - Terri L Blackwell
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA 94143, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, CA 94143, USA
| | - Peggy M Cawthon
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA 94143, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, CA 94143, USA
| | - Paul M Coen
- Translational Research Institute, AdventHealth, Orlando, FL 32804, USA
| | - Steven R Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA 94143, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, CA 94143, USA
| | | | - Samaneh Farsijani
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Daniel E Forman
- Department of Medicine (Geriatrics and Cardiology), University of Pittsburgh; and Geriatrics, Research, Education, and Clinical Center (GRECC), VA Pittsburgh Healthcare System, Pittsburgh, PA 15261, USA
| | - Bret H Goodpaster
- Translational Research Institute, AdventHealth, Orlando, FL 32804, USA
| | - Stephen B Kritchevsky
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27109, USA
| | - Theresa Mau
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA 94143, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, CA 94143, USA
| | - Frederico G S Toledo
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Anne B Newman
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Nancy W Glynn
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
14
|
Narala VR, Narala SR, Aiya Subramani P, Panati K, Kolliputi N. Role of mitochondria in inflammatory lung diseases. Front Pharmacol 2024; 15:1433961. [PMID: 39228517 PMCID: PMC11368744 DOI: 10.3389/fphar.2024.1433961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/05/2024] [Indexed: 09/05/2024] Open
Abstract
Mitochondria play a significant and varied role in inflammatory lung disorders. Mitochondria, known as the powerhouse of the cell because of their role in producing energy, are now recognized as crucial regulators of inflammation and immunological responses. Asthma, chronic obstructive pulmonary disease, and acute respiratory distress syndrome are characterized by complex interactions between immune cells, inflammatory substances, and tissue damage. Dysfunctional mitochondria can increase the generation of reactive oxygen species (ROS), triggering inflammatory pathways. Moreover, mitochondrial failure impacts cellular signaling, which in turn affects the expression of molecules that promote inflammation. In addition, mitochondria have a crucial role in controlling the behavior of immune cells, such as their activation and differentiation, which is essential in the development of inflammatory lung diseases. Their dynamic behavior, encompassing fusion, fission, and mitophagy, also impacts cellular responses to inflammation and oxidative stress. Gaining a comprehensive understanding of the intricate correlation between mitochondria and lung inflammation is essential in order to develop accurate treatment strategies. Targeting ROS generation, dynamics, and mitochondrial function may offer novel approaches to treating inflammatory lung diseases while minimizing tissue damage. Additional investigation into the precise contributions of mitochondria to lung inflammation will provide significant knowledge regarding disease mechanisms and potential therapeutic approaches. This review will focus on how mitochondria in the lung regulate these processes and their involvement in acute and chronic lung diseases.
Collapse
Affiliation(s)
| | | | | | - Kalpana Panati
- Department of Biotechnology, Government College for Men, Kadapa, India
| | - Narasaiah Kolliputi
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
15
|
Sun X, Wu T, Yang Z, Chen S, Zhao Z, Hu C, Wu S, Wu J, Mao Y, Liu J, Guo C, Cao G, Xu X, Huang S, Liang G. Regulatory role of PDK1 via integrated gene analysis of mitochondria-immune response in periodontitis. Gene 2024; 918:148476. [PMID: 38657876 DOI: 10.1016/j.gene.2024.148476] [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: 11/12/2023] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 04/26/2024]
Abstract
AIMS To investigate the association between mitochondrial events and immune response in periodontitis and related regulatory genes. MAIN METHODS Gene expression profiles in gingival tissues were retrieved from the Gene Expression Omnibus. Mitochondria-immune response-related differentially expressed genes (MIR-DEGs) between the healthy and periodontitis samples were determined. WGCNA, GO, and KEGG were used to investigate the function and the enriched pathways of MIR-DEGs. The correlation between MIR-DEGs expression and clinical probing pocket depth was analyzed. The MIR-DEGs were further identified and verified in animal samples. A periodontitis model was established in C57BL/6 mice with silk ligation. Micro-computed tomography was used to assess alveolar bone loss. Western blot, quantitative real-time polymerase chain reaction, and immunohistochemical analyses further validated the differential expression of the MIR-DEGs. KEY FINDINGS A total of ten MIR-DEGs (CYP24A1, PRDX4, GLDC, PDK1, BCL2A1, CBR3, ARMCX3, BNIP3, IFI27, and UNG) were identified, the expression of which could effectively distinguish patients with periodontitis from the healthy controls. Enhanced immune response was detected in the periodontitis group with that in the healthy controls, especially in B cells. PDK1 was a critical MIR-DEG correlated with B cell immune response and clinical periodontal probing pocket depth. Both animal and clinical periodontal samples presented higher gene and protein expression of PDK1 than the control samples. Additionally, PDK1 colocalized with B cells in both animal and clinical periodontal tissues. SIGNIFICANCE Mitochondria participate in the regulation of the immune response in periodontitis. PDK1 may be the key mitochondria-related gene regulating B-cell immune response in periodontitis.
Collapse
Affiliation(s)
- Xiaoyu Sun
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310012, China; Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Periodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Tong Wu
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Zhan Yang
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Shuhong Chen
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Zheyu Zhao
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Chaoming Hu
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Shengzhuang Wu
- School and Hospital of Stomatology, Hangzhou Medical University, Wenzhou, China
| | - Jiayu Wu
- School of Medicine, Jiujiang University, 320 Xunyang East Road, Jiujiang City, Jiangxi Province 332000, China
| | - Yixin Mao
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Jiefan Liu
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Oral and Maxillofacial Surgery/Pathology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Chen Guo
- School/Hospital of Stomatology, Lanzhou University, Lanzhou 730000, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang 310053, China
| | - Xiangwei Xu
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310012, China.
| | - Shengbin Huang
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.
| | - Guang Liang
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310012, China.
| |
Collapse
|
16
|
Tao H, Zhu P, Xia W, Chu M, Chen K, Wang Q, Gu Y, Lu X, Bai J, Geng D. The Emerging Role of the Mitochondrial Respiratory Chain in Skeletal Aging. Aging Dis 2024; 15:1784-1812. [PMID: 37815897 PMCID: PMC11272194 DOI: 10.14336/ad.2023.0924] [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: 08/03/2023] [Accepted: 09/24/2023] [Indexed: 10/12/2023] Open
Abstract
Maintenance of mitochondrial homeostasis is crucial for ensuring healthy mitochondria and normal cellular function. This process is primarily responsible for regulating processes that include mitochondrial OXPHOS, which generates ATP, as well as mitochondrial oxidative stress, apoptosis, calcium homeostasis, and mitophagy. Bone mesenchymal stem cells express factors that aid in bone formation and vascular growth. Positive regulation of hematopoietic stem cells in the bone marrow affects the differentiation of osteoclasts. Furthermore, the metabolic regulation of cells that play fundamental roles in various regions of the bone, as well as interactions within the bone microenvironment, actively participates in regulating bone integrity and aging. The maintenance of cellular homeostasis is dependent on the regulation of intracellular organelles, thus understanding the impact of mitochondrial functional changes on overall bone metabolism is crucially important. Recent studies have revealed that mitochondrial homeostasis can lead to morphological and functional abnormalities in senescent cells, particularly in the context of bone diseases. Mitochondrial dysfunction in skeletal diseases results in abnormal metabolism of bone-associated cells and a secondary dysregulated microenvironment within bone tissue. This imbalance in the oxidative system and immune disruption in the bone microenvironment ultimately leads to bone dysplasia. In this review, we examine the latest developments in mitochondrial respiratory chain regulation and its impacts on maintenance of bone health. Specifically, we explored whether enhancing mitochondrial function can reduce the occurrence of bone cell deterioration and improve bone metabolism. These findings offer prospects for developing bone remodeling biology strategies to treat age-related degenerative diseases.
Collapse
Affiliation(s)
- Huaqiang Tao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| | - Pengfei Zhu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| | - Wenyu Xia
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| | - Miao Chu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| | - Kai Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| | - Qiufei Wang
- Department of Orthopedics, Changshu Hospital Affiliated to Soochow University, First People’s Hospital of Changshu City, Jiangsu, China.
| | - Ye Gu
- Department of Orthopedics, Changshu Hospital Affiliated to Soochow University, First People’s Hospital of Changshu City, Jiangsu, China.
| | - Xiaomin Lu
- Department of Oncology, Affiliated Haian Hospital of Nantong University, Jiangsu, China.
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui, China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| |
Collapse
|
17
|
Guevara-Cruz M, Hernández-Gómez KG, Condado-Huerta C, González-Salazar LE, Peña-Flores AK, Pichardo-Ontiveros E, Serralde-Zúñiga AE, Sánchez-Tapia M, Maya O, Medina-Vera I, Noriega LG, López-Barradas A, Rodríguez-Lima O, Mata I, Olin-Sandoval V, Torres N, Tovar AR, Velázquez-Villegas LA. Intermittent fasting, calorie restriction, and a ketogenic diet improve mitochondrial function by reducing lipopolysaccharide signaling in monocytes during obesity: A randomized clinical trial. Clin Nutr 2024; 43:1914-1928. [PMID: 39003957 DOI: 10.1016/j.clnu.2024.06.036] [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: 02/27/2024] [Revised: 06/24/2024] [Accepted: 06/30/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Mitochondrial dysfunction occurs in monocytes during obesity and contributes to a low-grade inflammatory state; therefore, maintaining good mitochondrial conditions is a key aspect of maintaining health. Dietary interventions are primary strategies for treating obesity, but little is known about their impact on monocyte bioenergetics. Thus, the aim of this study was to evaluate the effects of calorie restriction (CR), intermittent fasting (IF), a ketogenic diet (KD), and an ad libitum habitual diet (AL) on mitochondrial function in monocytes and its modulation by the gut microbiota. METHODS AND FINDINGS A randomized controlled clinical trial was conducted in which individuals with obesity were assigned to one of the 4 groups for 1 month. Subsequently, the subjects received rifaximin and continued with the assigned diet for another month. The oxygen consumption rate (OCR) was evaluated in isolated monocytes, as was the gut microbiota composition in feces and anthropometric and biochemical parameters. Forty-four subjects completed the study, and those who underwent CR, IF and KD interventions had an increase in the maximal respiration OCR (p = 0.025, n2p = 0.159 [0.05, 0.27] 95% confidence interval) in monocytes compared to that in the AL group. The improvement in mitochondrial function was associated with a decrease in monocyte dependence on glycolysis after the IF and KD interventions. Together, diet and rifaximin increased the gut microbiota diversity in the IF and KD groups (p = 0.0001), enriched the abundance of Phascolarctobacterium faecium (p = 0.019) in the CR group and Ruminococcus bromii (p = 0.020) in the CR and KD groups, and reduced the abundance of lipopolysaccharide (LPS)-producing bacteria after CR, IF and KD interventions compared to the AL group at the end of the study according to ANCOVA with covariate adjustment. Spearman's correlation between the variables measured highlighted LPS as a potential modulator of the observed effects. In line with this findings, serum LPS and intracellular signaling in monocytes decreased with the three interventions (CR, p = 0.002; IF, p = 0.001; and KD, p = 0.001) compared to those in the AL group at the end of the study. CONCLUSIONS We conclude that these dietary interventions positively regulate mitochondrial bioenergetic health and improve the metabolic profile of monocytes in individuals with obesity via modulation of the gut microbiota. Moreover, the evaluation of mitochondrial function in monocytes could be used as an indicator of metabolic and inflammatory status, with potential applications in future clinical trials. TRIAL REGISTRATION This trial was registered with ClinicalTrials.gov (NCT05200468).
Collapse
Affiliation(s)
- Martha Guevara-Cruz
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Karla G Hernández-Gómez
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Citlally Condado-Huerta
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Luis E González-Salazar
- Servicio de Nutriología Clínica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Ana Karen Peña-Flores
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Edgar Pichardo-Ontiveros
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Aurora E Serralde-Zúñiga
- Servicio de Nutriología Clínica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Mónica Sánchez-Tapia
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Otoniel Maya
- Chalmers e-Commons. Chalmers University of Technology, Gotemburg, Vastra Gotaland, Sweden
| | - Isabel Medina-Vera
- Departamento de Metodología de la Investigación, Instituto Nacional de Pediatría, Ciudad de México, Mexico
| | - Lilia G Noriega
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Adriana López-Barradas
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Oscar Rodríguez-Lima
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Irma Mata
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Viridiana Olin-Sandoval
- Laboratorio 43. Departamento de Biotecnología y Bioingeniería, Cinvestav-Zacatenco, Ciudad de México, Mexico
| | - Nimbe Torres
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Armando R Tovar
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Laura A Velázquez-Villegas
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico.
| |
Collapse
|
18
|
Wei YY, Ye JJ, Zhang DW, Hu L, Wu HM, Fei GH. Melatonin Rescues Influenza A Virus-Induced Cellular Energy Exhaustion via OMA1-OPA1-S in Acute Exacerbation of COPD. J Pineal Res 2024; 76:e12991. [PMID: 39039850 DOI: 10.1111/jpi.12991] [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: 01/23/2024] [Revised: 05/25/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024]
Abstract
Although rapid progression and a poor prognosis in influenza A virus (IAV) infection-induced acute exacerbation of chronic obstructive pulmonary disease (AECOPD) are frequently associated with metabolic energy disorders, the underlying mechanisms and rescue strategies remain unknown. We herein demonstrated that the level of resting energy expenditure increased significantly in IAV-induced AECOPD patients and that cellular energy exhaustion emerged earlier and more significantly in IAV-infected primary COPD bronchial epithelial (pDHBE) cells. The differentially expressed genes were enriched in the oxidative phosphorylation (OXPHOS) pathway; additionally, we consistently uncovered much earlier ATP exhaustion, more severe mitochondrial structural destruction and dysfunction, and OXPHOS impairment in IAV-inoculated pDHBE cells, and these changes were rescued by melatonin. The level of OMA1-dependent cleavage of OPA1 in the mitochondrial inner membrane and the shift in energy metabolism from OXPHOS to glycolysis were significantly increased in IAV-infected pDHBE cells; however, these changes were rescued by OMA1-siRNA or melatonin further treatment. Collectively, our data revealed that melatonin rescued IAV-induced cellular energy exhaustion via OMA1-OPA1-S to improve the clinical prognosis in COPD. This treatment may serve as a potential therapeutic agent for patients in which AECOPD is induced by IAV.
Collapse
Affiliation(s)
- Yuan-Yuan Wei
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, People's Republic of China
| | - Jing-Jing Ye
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, People's Republic of China
| | - Da-Wei Zhang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, People's Republic of China
| | - Lei Hu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, People's Republic of China
| | - Hui-Mei Wu
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, People's Republic of China
- Department of Geriatric Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Guang-He Fei
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, People's Republic of China
| |
Collapse
|
19
|
Wang L, Peng T, Deng J, Gao W, Wang H, Junhong Luo O, Huang L, Chen G. Nicotinamide riboside alleviates brain dysfunction induced by chronic cerebral hypoperfusion via protecting mitochondria. Biochem Pharmacol 2024; 225:116272. [PMID: 38723719 DOI: 10.1016/j.bcp.2024.116272] [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: 01/17/2024] [Revised: 04/16/2024] [Accepted: 05/06/2024] [Indexed: 05/14/2024]
Abstract
Chronic cerebral hypoperfusion (CCH) is an enduring inadequate blood flow to the brain, resulting in vascular dementia (VaD). However, the effective treatment strategies are lacking. Supplementing with nicotinamide adenine dinucleotide (NAD+) has shown neuroprotective benefits in other neurodegenerative disorders. Nicotinamide riboside (NR), as a precursor of NAD+, is believed to hold promise in improving mitochondrial health, autophagy, and cognitive function. Meanwhile, NR has unique oral bioavailability, good tolerability, and minimal side effects, and it is the most promising for clinical translation. However, the effectiveness of NR in treating CCH-related VaD is still uncertain. The present study examined the neuroprotective effects of NR supplementation and its underlying mechanisms in a CCH rat model. The rats with CCH were given NR at a daily dosage of 400 mg/kg for 3 months. NR supplementation increased blood and brain NAD+ levels and improved brain function in CCH rats, including cognitive function and oxygenation capacity. It also reduced hippocampal neuronal loss and abnormalities and mitigated the decrease in dendritic spine density. The analysis of RNA sequencing in hippocampal tissue supports these findings. Electron microscopy and protein detection results suggest that NR may maintain mitochondrial structural integrity and exert a protective role by attenuating mitochondrial fission and impaired autophagy flux caused by CCH. In conclusion, these findings offer evidence for the neuroprotective potential of NR supplementation in ameliorating cognitive impairment induced by CCH.
Collapse
Affiliation(s)
- Lina Wang
- Department of Neurology, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China; Department of Microbiology and Immunology, School of Medicine, Institute of Geriatric Immunology, Jinan University, Guangzhou 510632, China
| | - Tianchan Peng
- Department of Neurology, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China; Department of Microbiology and Immunology, School of Medicine, Institute of Geriatric Immunology, Jinan University, Guangzhou 510632, China
| | - Jieping Deng
- Department of Microbiology and Immunology, School of Medicine, Institute of Geriatric Immunology, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Guangzhou 510632, China
| | - Wen Gao
- Department of Microbiology and Immunology, School of Medicine, Institute of Geriatric Immunology, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Guangzhou 510632, China
| | - Haoyun Wang
- Department of Microbiology and Immunology, School of Medicine, Institute of Geriatric Immunology, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Guangzhou 510632, China
| | - Oscar Junhong Luo
- Department of Microbiology and Immunology, School of Medicine, Institute of Geriatric Immunology, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Guangzhou 510632, China
| | - Li'an Huang
- Department of Neurology, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China.
| | - Guobing Chen
- Department of Neurology, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China; Department of Microbiology and Immunology, School of Medicine, Institute of Geriatric Immunology, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Guangzhou 510632, China.
| |
Collapse
|
20
|
Ding SM, Shi LG, Cao ZP, Zhu NN, Liu YY, Wang MY, Cui SS, Cheng HR, Liang D, Cao YX, Liu YJ. HTR1B regulates mitochondrial homeostasis and mitophagy by activating the ERK/ MAPK signalling pathway during human embryonic arrest. Heliyon 2024; 10:e33132. [PMID: 39022094 PMCID: PMC11253063 DOI: 10.1016/j.heliyon.2024.e33132] [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: 03/08/2024] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 07/20/2024] Open
Abstract
Background Previous studies have shown that serotonin and its receptors are widely distributed in mammalian reproductive tisssues and play an important role in embryonic development. However, the specific effects of the serotonergic system on embryonic arrest (EA) and the underlying mechanism require further investigation. Methods Chorionic villi were collected from patients with EA and healthy pregnant women. Western blotting (WB) and immunohistochemistry (IHC) were used to detect serotonin receptor 1B (HTR1B) levels and evaluate mitochondrial function. Additionally, HTR-8/SVneo cells were transfected with an HTR1B overexpression plasmid. Quantitative real-time polymerase chain reaction(qRT-PCR), Cell Counting Kit-8 (CCK-8), and wound healing assays were utilized to evaluate mitophagy level, cell proliferation and cell migration, respectively. Results We discovered elevated HTR1B levels in the chorionic villi of the patients with EA compared to controls. Concurrently, we observed enhanced levels of nucleus-encoded proteins including mitofilin, succinate dehydrogenase complex subunit A (SDHA), and cytochrome c oxidase subunit 4 (COXIV), along with the mitochondrial fusion protein optic atrophy 1(OPA1), fission proteins mitochondrial fission protein 1(FIS1) and mitochondrial fission factor (MFF) in the EA group. Additionally, there was an excessive mitophagy levels in EA group. Furthermore, a notable activation of mitogen-activated protein kinase (MAPK) signaling pathway proteins including extracellular regulating kinase (ERK), c-Jun N-terminal kinase (JNK), and P38 was observed in the EA group. By overexpressing HTR1B in HTR-8/SVneo cells, we observed a significant reduction in cell proliferation and migration. HTR1B overexpression also caused an increase in levels of SDHA and FIS1, as well as an upregulation of mitophagy. Notably, the ERK inhibitor U0126 effectively mitigated these effects. Conclusion These findings show that HTR1B influences mitochondrial homeostasis, promoting excessive mitophagy and impairing cell proliferation and migration by activating the MAPK signalling pathway during post-implantation EA. Therefore, HTR1B may serve as a potential therapeutic target for patients with EA.
Collapse
Affiliation(s)
- Si-min Ding
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230022, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University),Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Province Key Laboratory of Reproductive Disorders and Obstetrics and Gynaecology Diseases, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Ling-ge Shi
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230022, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University),Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Province Key Laboratory of Reproductive Disorders and Obstetrics and Gynaecology Diseases, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Zhen-ping Cao
- The Third People's Hospital of Hefei, The Third Clinical Teaching Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Na-na Zhu
- The Second People's Hospital of Hefei, The Second Clinical Teaching Hospital of Anhui Medical University, Hefei, 230011, Anhui, China
| | - Yun-yun Liu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230022, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University),Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Meng-yao Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230022, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University),Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Shuang-shuang Cui
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230022, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University),Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Hui-ru Cheng
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230022, China
| | - Dan Liang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230022, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University),Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yun-xia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230022, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University),Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Province Key Laboratory of Reproductive Disorders and Obstetrics and Gynaecology Diseases, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Institute of Translational Medicine,No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Ya-jing Liu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230022, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University),Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Province Key Laboratory of Reproductive Disorders and Obstetrics and Gynaecology Diseases, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Institute of Translational Medicine,No 81 Meishan Road, Hefei, 230032, Anhui, China
| |
Collapse
|
21
|
Nakamura J, Shiohama Y, Röth D, Haruta T, Yamashita Y, Mitsuzono T, Mochizuki C, Kalkum M, Nakamura M. Size and Surface Properties of Functionalized Organosilica Particles Impact Cell-Particle Interactions Including Mitochondrial Activity. ACS APPLIED MATERIALS & INTERFACES 2024; 16:30980-30996. [PMID: 38857433 DOI: 10.1021/acsami.4c06455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Understanding of the interactions between macrophages and multifunctional nanoparticles is important for development of novel macrophage-based immunotherapies. Here, we investigated the effects of fluorescent thiol-organosilica particle size and surface properties on cell-particle interactions, including mitochondrial activity, using the mouse macrophage cell line J774A.1. Three different sizes of thiol-organosilica particles (150, 400, and 680 nm in diameter) containing fluorescein (OS/F150, OS/F400, and OS/F680) and particles surface functionalized with polyethylenimine (PEI) (OS/F150PEI, OS/F400PEI, and OS/F680PEI) were prepared. Flow cytometric analysis, time-lapse imaging, and single-cell analysis of particle uptake and mitochondrial activity of J774A.1 cells demonstrated variations in uptake and kinetics depending on the particle size and surface as well as on each individual cell. Cells treated with OS/F150 and OS/F150PEI showed higher uptake and mitochondrial activity than those treated with other particles. The interaction between endosomes and mitochondria was observed using 3D fluorescent imaging and was characterized by the involvement of iron transport into mitochondria by iron-containing proteins adsorbed on the particle surface. Scanning electron microscopy of the cells treated with the particles revealed alterations in cell membrane morphology, depending on particle size and surface. We performed correlative light and electron microscopy combined with time-lapse and 3D imaging to develop an integrated correlation analysis of particle uptake, mitochondrial activity, and cell membrane morphology in single macrophages. These cell-specific characteristics of macrophages against functional particles and their evaluation methods are crucial for understanding the immunological functions of individual macrophages and developing novel immunotherapies.
Collapse
Affiliation(s)
- Junna Nakamura
- Department of Organ Anatomy and Nanomedicine, Yamaguchi University Graduate School of Medicine, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
- Core Clusters for Research Initiatives of Yamaguchi University, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
- Research Institute for Cell Design Medical Science, Yamaguchi University, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Yasuo Shiohama
- Department of Organ Anatomy and Nanomedicine, Yamaguchi University Graduate School of Medicine, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
- Core Clusters for Research Initiatives of Yamaguchi University, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
- Research Institute for Cell Design Medical Science, Yamaguchi University, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Daniel Röth
- Department of Department of Immunology & Theranostics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, California 91010, United States
| | - Tomohiro Haruta
- EM application group, EM business unit, JEOL Ltd., Akishima, Tokyo JP 196-8558, Japan
| | - Yukari Yamashita
- Department of Organ Anatomy and Nanomedicine, School of Medicine, Facuelty of Medicine and Health Sciences, Yamaguchi University, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Tomohiro Mitsuzono
- Department of Organ Anatomy and Nanomedicine, School of Medicine, Facuelty of Medicine and Health Sciences, Yamaguchi University, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Chihiro Mochizuki
- Department of Organ Anatomy and Nanomedicine, Yamaguchi University Graduate School of Medicine, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
- Core Clusters for Research Initiatives of Yamaguchi University, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
- Research Institute for Cell Design Medical Science, Yamaguchi University, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Markus Kalkum
- Department of Department of Immunology & Theranostics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, California 91010, United States
| | - Michihiro Nakamura
- Department of Organ Anatomy and Nanomedicine, Yamaguchi University Graduate School of Medicine, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
- Core Clusters for Research Initiatives of Yamaguchi University, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
- Research Institute for Cell Design Medical Science, Yamaguchi University, 1-1-1 minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| |
Collapse
|
22
|
Fang B, Zhao L, Huo B, Chen F, Yuan P, Lai S, Wu A, Zhuo Y. Maternal consumption of fish oil protected breast-fed piglets against Escherichia coli lipopolysaccharide-induced damage through reshaping of intestinal fatty acids profile. Front Vet Sci 2024; 11:1417078. [PMID: 38952807 PMCID: PMC11215148 DOI: 10.3389/fvets.2024.1417078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 06/05/2024] [Indexed: 07/03/2024] Open
Abstract
It has been well documented that n-3 polyunsaturated fatty acids (n-3 PUFA) can alleviate inflammation caused by Escherichia coli (E. coli) lipopolysaccharides (LPS), the etiologic agents that causing yellow or white dysentery in young pigs. However, it remains unclear whether the increase in n-3 PUFA availability could enhance the ability of nursery pigs to resist invasion by E. coli. LPS. Twenty-four 21-day-old female piglets, each two of them from the same sow fed the beef tallow (BT) or fish oil (FO) diets, were allocated into four treatment groups: BT-CON, piglets from the BT-fed sows and intraperitoneally injected with saline (9 g/L); BT-LPS, piglets from the BT-fed sows and injected with LPS (100 μg/kg body weight); FO-CON, piglets from the FO-fed sows and injected with saline; FO-LPS, piglets from the FO-fed sows and injected with LPS. Following 2 h of LPS challenge, the magnitudes of increase in body temperature approached to a marked (p < 0.01) difference between the BT-CON and BT-LPS piglets, whereas the dramatic (p < 0.01) difference between the FO-CON and FO-LPS piglets was only observed at 4 h post LPS challenge. The body temperature averaged across the time points evaluated was about 0.2°C lower (p < 0.05) in the FO group than in the BT group. The FO group had lower (p < 0.05) mean corpuscular hemoglobin concentration, lower increase in serum interleukin (IL)-1β (p < 0.10) and IL-8 (p < 0.05) levels, higher (p < 0.01) serum albumin concentration, and higher (p = 0.10) ratios of jejunum villus height to crypt depth than the BT group. The FO group had much higher (p < 0.0001) ileal content of C20:5n3, C24:0, and C22:6n3, which were 2-4 times the content of the BT group. LPS challenge resulted in decreased (p < 0.05) intestinal C20:1 and C20:5n3 content, and the decrease (p < 0.05) in intestinal C20:3n6 and C24:1 content was observed in the BT-LPS piglets rather than in the FO-LPS piglets. Taken together, this study indicated that maternal consumption of fish oil protected breast-fed piglets against E. coli LPS-induced damage through reshaping of intestinal fatty acids profile, which sheds new light on the development of nutritional strategies to enhance the ability of young pigs to resist E. coli invasion.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yong Zhuo
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
23
|
Qiu H, Xiong H, Zheng J, Peng Y, Wang C, Hu Q, Zhao F, Chen K. Sr-Incorporated Bioactive Glass Remodels the Immunological Microenvironment by Enhancing the Mitochondrial Function of Macrophage via the PI3K/AKT/mTOR Signaling Pathway. ACS Biomater Sci Eng 2024; 10:3923-3934. [PMID: 38766805 DOI: 10.1021/acsbiomaterials.4c00228] [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: 05/22/2024]
Abstract
The repair of critical-sized bone defects continues to pose a challenge in clinics. Strontium (Sr), recognized for its function in bone metabolism regulation, has shown potential in bone repair. However, the underlying mechanism through which Sr2+ guided favorable osteogenesis by modulating macrophages remains unclear, limiting their application in the design of bone biomaterials. Herein, Sr-incorporated bioactive glass (SrBG) was synthesized for further investigation. The release of Sr ions enhanced the immunomodulatory properties and osteogenic potential by modulating the polarization of macrophages toward the M2 phenotype. In vivo, a 3D-printed SrBG scaffold was fabricated and showed consistently improved bone regeneration by creating a prohealing immunological microenvironment. RNA sequencing was performed to explore the underlying mechanisms. It was found that Sr ions might enhance the mitochondrial function of macrophage by activating PI3K/AKT/mTOR signaling, thereby favoring osteogenesis. Our findings demonstrate the relationship between the immunomodulatory role of Sr ions and the mitochondrial function of macrophages. By focusing on the mitochondrial function of macrophages, Sr2+-mediated immunomodulation sheds light on the future design of biomaterials for tissue regenerative engineering.
Collapse
Affiliation(s)
- Huanhuan Qiu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Huacui Xiong
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Jiafu Zheng
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, China
| | - Yuqi Peng
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Chunhui Wang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Qing Hu
- School of Materials Science and Engineering, Jingdezhen Ceramic University, Jingdezhen 333001, China
| | - Fujian Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Ke Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
24
|
Mukherjee A, Ghosh KK, Chakrabortty S, Gulyás B, Padmanabhan P, Ball WB. Mitochondrial Reactive Oxygen Species in Infection and Immunity. Biomolecules 2024; 14:670. [PMID: 38927073 PMCID: PMC11202257 DOI: 10.3390/biom14060670] [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: 05/15/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Reactive oxygen species (ROS) contain at least one oxygen atom and one or more unpaired electrons and include singlet oxygen, superoxide anion radical, hydroxyl radical, hydroperoxyl radical, and free nitrogen radicals. Intracellular ROS can be formed as a consequence of several factors, including ultra-violet (UV) radiation, electron leakage during aerobic respiration, inflammatory responses mediated by macrophages, and other external stimuli or stress. The enhanced production of ROS is termed oxidative stress and this leads to cellular damage, such as protein carbonylation, lipid peroxidation, deoxyribonucleic acid (DNA) damage, and base modifications. This damage may manifest in various pathological states, including ageing, cancer, neurological diseases, and metabolic disorders like diabetes. On the other hand, the optimum levels of ROS have been implicated in the regulation of many important physiological processes. For example, the ROS generated in the mitochondria (mitochondrial ROS or mt-ROS), as a byproduct of the electron transport chain (ETC), participate in a plethora of physiological functions, which include ageing, cell growth, cell proliferation, and immune response and regulation. In this current review, we will focus on the mechanisms by which mt-ROS regulate different pathways of host immune responses in the context of infection by bacteria, protozoan parasites, viruses, and fungi. We will also discuss how these pathogens, in turn, modulate mt-ROS to evade host immunity. We will conclude by briefly giving an overview of the potential therapeutic approaches involving mt-ROS in infectious diseases.
Collapse
Affiliation(s)
- Arunima Mukherjee
- Department of Biological Sciences, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur 522502, Andhra Pradesh, India;
| | - Krishna Kanta Ghosh
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (B.G.)
| | - Sabyasachi Chakrabortty
- Department of Chemistry, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur 522502, Andhra Pradesh, India;
| | - Balázs Gulyás
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (B.G.)
- Cognitive Neuroimaging Centre, 59 Nanyang Drive, Nanyang Technological University, Singapore 636921, Singapore
- Department of Clinical Neuroscience, Karolinska Institute, 17176 Stockholm, Sweden
| | - Parasuraman Padmanabhan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (B.G.)
- Cognitive Neuroimaging Centre, 59 Nanyang Drive, Nanyang Technological University, Singapore 636921, Singapore
| | - Writoban Basu Ball
- Department of Biological Sciences, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur 522502, Andhra Pradesh, India;
| |
Collapse
|
25
|
Lin L, Ren R, Xiong Q, Zheng C, Yang B, Wang H. Remodeling of T-cell mitochondrial metabolism to treat autoimmune diseases. Autoimmun Rev 2024; 23:103583. [PMID: 39084278 DOI: 10.1016/j.autrev.2024.103583] [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: 05/06/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
T cells are key drivers of the pathogenesis of autoimmune diseases by producing cytokines, stimulating the generation of autoantibodies, and mediating tissue and cell damage. Distinct mitochondrial metabolic pathways govern the direction of T-cell differentiation and function and rely on specific nutrients and metabolic enzymes. Metabolic substrate uptake and mitochondrial metabolism form the foundational elements for T-cell activation, proliferation, differentiation, and effector function, contributing to the dynamic interplay between immunological signals and mitochondrial metabolism in coordinating adaptive immunity. Perturbations in substrate availability and enzyme activity may impair T-cell immunosuppressive function, fostering autoreactive responses and disrupting immune homeostasis, ultimately contributing to autoimmune disease pathogenesis. A growing body of studies has explored how metabolic processes regulate the function of diverse T-cell subsets in autoimmune diseases such as systemic lupus erythematosus (SLE), multiple sclerosis (MS), autoimmune hepatitis (AIH), inflammatory bowel disease (IBD), and psoriasis. This review describes the coordination of T-cell biology by mitochondrial metabolism, including the electron transport chain (ETC), oxidative phosphorylation, amino acid metabolism, fatty acid metabolism, and one‑carbon metabolism. This study elucidated the intricate crosstalk between mitochondrial metabolic programs, signal transduction pathways, and transcription factors. This review summarizes potential therapeutic targets for T-cell mitochondrial metabolism and signaling in autoimmune diseases, providing insights for future studies.
Collapse
Affiliation(s)
- Liyan Lin
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu 610041, China; Laboratory Medicine Research Center of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ruyu Ren
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu 610041, China; Laboratory Medicine Research Center of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiao Xiong
- Department of Infectious Disease, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chunfu Zheng
- Department of Microbiology, Immunology & Infection Diseases, University of Calgary, Calgary, Alberta, Canada.
| | - Bin Yang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu 610041, China; Laboratory Medicine Research Center of West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Huiqing Wang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
26
|
Zhou R, Song Y, Xu C, Zhang Y, Wu X, Zhang L, Luo X, Zhao H, Liu M, Xu J, Wang L, Chen Z, Han Q. Altered counts and mitochondrial mass of peripheral blood leucocytes in patients with chronic hepatitis B virus infection. J Cell Mol Med 2024; 28:e18440. [PMID: 38890792 PMCID: PMC11187856 DOI: 10.1111/jcmm.18440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 04/24/2024] [Accepted: 05/11/2024] [Indexed: 06/20/2024] Open
Abstract
Hepatitis B virus (HBV) damages liver cells through abnormal immune responses. Mitochondrial metabolism is necessary for effector functions of white blood cells (WBCs). The aim was to investigate the altered counts and mitochondrial mass (MM) of WBCs by two novel indicators of mitochondrial mass, MM and percentage of low mitochondrial membrane potential, MMPlow%, due to chronic HBV infection. The counts of lymphocytes, neutrophils and monocytes in the HBV infection group were in decline, especially for lymphocyte (p = 0.034) and monocyte counts (p = 0.003). The degraded MM (p = 0.003) and MMPlow% (p = 0.002) of lymphocytes and MM (p = 0.005) of monocytes suggested mitochondrial dysfunction of WBCs. HBV DNA within WBCs showed an extensive effect on mitochondria metabolic potential of lymphocytes, neutrophils and monocytes indicated by MM; hepatitis B e antigen was associated with instant mitochondrial energy supply indicated by MMPlow% of neutrophils; hepatitis B surface antigen, antiviral therapy by nucleos(t)ide analogues and prolonged infection were also vital factors contributing to WBC alterations. Moreover, degraded neutrophils and monocytes could be used to monitor immune responses reflecting chronic liver fibrosis and inflammatory damage. In conclusion, MM combined with cell counts of WBCs could profoundly reflect WBC alterations for monitoring chronic HBV infection. Moreover, HBV DNA within WBCs may be a vital factor in injuring mitochondria metabolic potential.
Collapse
Affiliation(s)
- Ruo‐Ran Zhou
- Medical Center of Soochow UniversitySuzhou Medical College of Soochow UniversitySuzhouPeople's Republic of China
| | - Ya‐Hui Song
- Center of Clinical Laboratory and Translational MedicineThe Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake HospitalSuzhouPeople's Republic of China
| | - Cheng‐Yu Xu
- Center of Clinical Laboratory and Translational MedicineThe Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake HospitalSuzhouPeople's Republic of China
| | - Ying‐Ying Zhang
- Infectious Disease DepartmentThe Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake HospitalSuzhouPeople's Republic of China
| | - Xiang‐Wei Wu
- Center of Clinical Laboratory and Translational MedicineThe Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake HospitalSuzhouPeople's Republic of China
| | - Lu Zhang
- Center of Clinical Laboratory and Translational MedicineThe Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake HospitalSuzhouPeople's Republic of China
| | - Xi‐Ni Luo
- Medical Center of Soochow UniversitySuzhou Medical College of Soochow UniversitySuzhouPeople's Republic of China
| | - Han Zhao
- Medical Center of Soochow UniversitySuzhou Medical College of Soochow UniversitySuzhouPeople's Republic of China
| | - Ming‐Ming Liu
- Infectious Disease DepartmentThe Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake HospitalSuzhouPeople's Republic of China
| | - Jun‐Chi Xu
- The Fifth People's Hospital of SuzhouSuzhouPeople's Republic of China
| | - Lin Wang
- Center of Clinical Laboratory and Translational MedicineThe Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake HospitalSuzhouPeople's Republic of China
| | - Zu‐Tao Chen
- Center of Clinical Laboratory and Translational MedicineThe Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake HospitalSuzhouPeople's Republic of China
- Infectious Disease DepartmentThe First Affiliated Hospital of Soochow UniversitySuzhouPeople's Republic of China
| | - Qing‐Zhen Han
- Medical Center of Soochow UniversitySuzhou Medical College of Soochow UniversitySuzhouPeople's Republic of China
- Center of Clinical Laboratory and Translational MedicineThe Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake HospitalSuzhouPeople's Republic of China
| |
Collapse
|
27
|
Qian F, Zhong Q, Chen Z. Role of mitochondrial dysfunction in acute traumatic brain injury: Evidence from bioinformatics analysis. Heliyon 2024; 10:e31121. [PMID: 38803920 PMCID: PMC11128910 DOI: 10.1016/j.heliyon.2024.e31121] [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: 12/06/2023] [Revised: 04/23/2024] [Accepted: 05/10/2024] [Indexed: 05/29/2024] Open
Abstract
Background The intricate regulatory relationship between mitochondrial dysfunction, apoptosis, and immune cells remains largely elusive following traumatic brain injury (TBI). Methods The GSE45997 dataset from the Gene Expression Omnibus database and utilized GEO2R to screen for differentially expressed genes (DEGs). Functional enrichment analyses were performed. Mitochondrial gene data from the MitoCarta3.0 database were combined with the DEGs to identify mitochondria-related DEGs (MitoDEGs). The hub MitoDEGs related to apoptosis were further screened. Animal models of TBI were established to investigate the mechanisms underlying mitochondrial dysfunction regulation of apoptosis. Furthermore, we explored the relationship between MitoDEGs/hub MitoDEGs and immune cells using the Spearman correlation method. Results Fifty-seven MitoDEGs were significantly enriched in pathways related to fatty acid degradation and metabolism. We identified three upregulated hub MitoDEGs, namely Dnm1l, Mcl1 and Casp3, were associated with apoptosis. In the animal experiments, we observed significant expression levels of microtubule-associated protein 1 light chain 3 beta (LC3B) surrounding the injury site. Most LC3B-expressing cells exhibited positive staining for Beclin 1 and colocalization analysis revealed the simultaneous presence of Beclin 1 and caspase-3. The Western blot analysis further unveiled a significant upregulation of cleaved caspase-3 levels and LC3B II/LC3B I ratio after TBI. Moreover, the quantity of myeloid cell leukaemia-1 immunoreactive cells was notably higher than that in the control group. Spearman correlation analysis demonstrated strong associations between plasma cells, marginal zone B cells, native CD4 T cells, monocytes, and MitoDEGs/hub MitoDEGs. Conclusions This study sheds light on enhanced fatty acid metabolism following mitochondrial dysfunction and its potential association with apoptosis and immune cell activation, thereby providing new mechanistic insights into the acute phase of TBI.
Collapse
Affiliation(s)
- Fangfang Qian
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Qi Zhong
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zhuoming Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
28
|
Halász H, Tárnai V, Matkó J, Nyitrai M, Szabó-Meleg E. Cooperation of Various Cytoskeletal Components Orchestrates Intercellular Spread of Mitochondria between B-Lymphoma Cells through Tunnelling Nanotubes. Cells 2024; 13:607. [PMID: 38607046 PMCID: PMC11011538 DOI: 10.3390/cells13070607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 03/28/2024] [Indexed: 04/13/2024] Open
Abstract
Membrane nanotubes (NTs) are dynamic communication channels connecting spatially separated cells even over long distances and promoting the transport of different cellular cargos. NTs are also involved in the intercellular spread of different pathogens and the deterioration of some neurological disorders. Transport processes via NTs may be controlled by cytoskeletal elements. NTs are frequently observed membrane projections in numerous mammalian cell lines, including various immune cells, but their functional significance in the 'antibody factory' B cells is poorly elucidated. Here, we report that as active channels, NTs of B-lymphoma cells can mediate bidirectional mitochondrial transport, promoted by the cooperation of two different cytoskeletal motor proteins, kinesin along microtubules and myosin VI along actin, and bidirectional transport processes are also supported by the heterogeneous arrangement of the main cytoskeletal filament systems of the NTs. We revealed that despite NTs and axons being different cell extensions, the mitochondrial transport they mediate may exhibit significant similarities. Furthermore, we found that microtubules may improve the stability and lifespan of B-lymphoma-cell NTs, while F-actin strengthens NTs by providing a structural framework for them. Our results may contribute to a better understanding of the regulation of the major cells of humoral immune response to infections.
Collapse
Affiliation(s)
- Henriett Halász
- Department of Biophysics, Medical School, University of Pécs, H-7624 Pécs, Hungary
| | - Viktória Tárnai
- Department of Biophysics, Medical School, University of Pécs, H-7624 Pécs, Hungary
| | - János Matkó
- Department of Immunology, Faculty of Science, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Miklós Nyitrai
- Department of Biophysics, Medical School, University of Pécs, H-7624 Pécs, Hungary
| | - Edina Szabó-Meleg
- Department of Biophysics, Medical School, University of Pécs, H-7624 Pécs, Hungary
| |
Collapse
|
29
|
Zhang M, Wei J, Sun Y, He C, Ma S, Pan X, Zhu X. The efferocytosis process in aging: Supporting evidence, mechanisms, and therapeutic prospects for age-related diseases. J Adv Res 2024:S2090-1232(24)00109-7. [PMID: 38499245 DOI: 10.1016/j.jare.2024.03.008] [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: 12/16/2023] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Aging is characterized by an ongoing struggle between the buildup of damage caused by a combination of external and internal factors. Aging has different effects on phagocytes, including impaired efferocytosis. A deficiency in efferocytosis can cause chronic inflammation, aging, and several other clinical disorders. AIM OF REVIEW Our review underscores the possible feasibility and extensive scope of employing dual targets in various age-related diseases to reduce the occurrence and progression of age-related diseases, ultimately fostering healthy aging and increasing lifespan. Key scientific concepts of review Hence, the concurrent implementation of strategies aimed at augmenting efferocytic mechanisms and anti-aging treatments has the potential to serve as a potent intervention for extending the duration of a healthy lifespan. In this review, we comprehensively discuss the concept and physiological effects of efferocytosis. Subsequently, we investigated the association between efferocytosis and the hallmarks of aging. Finally, we discuss growing evidence regarding therapeutic interventions for age-related disorders, focusing on the physiological processes of aging and efferocytosis.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Jin Wei
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Yu Sun
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Chang He
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Shiyin Ma
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| |
Collapse
|
30
|
Gerard A, Mizerik E, Mohila CA, AlAwami S, Hunter JV, Kearney DL, Lalani SR, Scaglia F. Intracranial calcifications simulating Aicardi-Goutières syndrome in PARS2-related mitochondrial disease. Am J Med Genet A 2024:e63589. [PMID: 38469956 DOI: 10.1002/ajmg.a.63589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/05/2024] [Accepted: 02/28/2024] [Indexed: 03/13/2024]
Abstract
PARS2 encodes an aminoacyl-tRNA synthetase that catalyzes the ligation of proline to mitochondrial prolyl-tRNA molecules. Diseases associated with PARS2 primarily affect the central nervous system, causing early infantile developmental epileptic encephalopathies (EIDEE; DEE75; MIM #618437) with infantile-onset neurodegeneration. Dilated cardiomyopathy has also been reported in the affected individuals. About 10 individuals to date have been described with pathogenic biallelic variants in PARS2. While many of the reported individuals succumbed to the disease in the first two decades of life, autopsy findings have not yet been reported. Here, we describe neuropathological findings in a deceased male with evidence of intracranial calcifications in the basal ganglia, thalamus, cerebellum, and white matter, similar to Aicardi-Goutières syndrome. This report describes detailed autopsy findings in a child with PARS2-related mitochondrial disease and provides plausible evidence that intracranial calcifications may be a previously unrecognized feature of this disorder.
Collapse
Affiliation(s)
- Amanda Gerard
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children's Hospital, Houston, Texas, USA
| | - Elizabeth Mizerik
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children's Hospital, Houston, Texas, USA
| | - Carrie A Mohila
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Sarah AlAwami
- King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Jill V Hunter
- Department of Radiology, Baylor College of Medicine, Houston, Texas, USA
- E.B. Singleton Department of Pediatric Radiology, Texas Children's Hospital, Houston, Texas, USA
| | - Debra L Kearney
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Seema R Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children's Hospital, Houston, Texas, USA
| | - Fernando Scaglia
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children's Hospital, Houston, Texas, USA
- Joint BCM-CUHK Center of Medical Genetics, Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
31
|
Pandey R, Chiu CC, Wang LF. Immunotherapy Study on Non-small-Cell Lung Cancer (NSCLC) Combined with Cytotoxic T Cells and miRNA34a. Mol Pharm 2024; 21:1364-1381. [PMID: 38291993 PMCID: PMC10915804 DOI: 10.1021/acs.molpharmaceut.3c01040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/18/2024] [Accepted: 01/18/2024] [Indexed: 02/01/2024]
Abstract
Immunotherapy has emerged as a promising approach for cancer treatment, and the use of microRNAs (miRNAs) as therapeutic agents has gained significant attention. In this study, we investigated the effectiveness of immunotherapy utilizing miRNA34a and Jurkat T cells in inducing cell death in non-small-cell lung cancer cells, specifically A549 cells. Moreover, we explored the impact of Jurkat T cell activation and miRNA34a delivery using iron oxide nanorods (IONRs) on the killing of cancer cells. A549 cells were cocultured with both activated and inactivated Jurkat T cells, both before and after the delivery of miRNA34a. Surprisingly, our results revealed that even inactive Jurkat T cells were capable of inducing cell death in cancer cells. This unexpected observation suggested the presence of alternative mechanisms by which Jurkat T cells can exert cytotoxic effects on cancer cells. We stimulated Jurkat T cells using anti-CD3/CD28 and analyzed their efficacy in killing A549 compared to that of the inactive Jurkat T cells in conjunction with miRNA34a. Our findings indicated that the activation of Jurkat T cells significantly enhanced their cytotoxic potential against cancer cells compared to their inactive counterparts. The combined treatment of A549 cells with activated Jurkat T cells and miRNA34a demonstrated the highest level of cancer cell death, suggesting a synergistic effect between Jurkat T cell activation and miRNA therapy. Besides the apoptosis mechanism for the Jurkat T cells' cytotoxic effects on A549 cells, we furthermore investigated the ferroptosis pathway, which was found to have an impact on the cancer cell killing due to the presence of miRNA34a and IONRs as the delivery agent inside the cancer cells.
Collapse
Affiliation(s)
- Richa Pandey
- Department
of Medicinal and Applied Chemistry, Kaohsiung
Medical University, No. 100 Shih-Chuan first Road, Kaohsiung 80708, Taiwan
| | - Chien-Chih Chiu
- Department
of Biotechnology, Kaohsiung Medical University, No. 100 Shih-Chuan first Road, Kaohsiung 80708, Taiwan
- Department
of Medical Research, Kaohsiung Medical University
Hospital, No.100 Tzyou
first Road, Kaohsiung 80708, Taiwan
| | - Li-Fang Wang
- Department
of Medicinal and Applied Chemistry, Kaohsiung
Medical University, No. 100 Shih-Chuan first Road, Kaohsiung 80708, Taiwan
- Department
of Medical Research, Kaohsiung Medical University
Hospital, No.100 Tzyou
first Road, Kaohsiung 80708, Taiwan
- Institute
of Medical Science and Technology, National
Sun Yat-Sen University, No.70 Lien-Hai Road, Kaohsiung 804201, Taiwan
| |
Collapse
|
32
|
Hartsoe P, Holguin F, Chu HW. Mitochondrial Dysfunction and Metabolic Reprogramming in Obesity and Asthma. Int J Mol Sci 2024; 25:2944. [PMID: 38474191 PMCID: PMC10931700 DOI: 10.3390/ijms25052944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 02/23/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Mitochondrial dysfunction and metabolic reprogramming have been extensively studied in many disorders ranging from cardiovascular to neurodegenerative disease. Obesity has previously been associated with mitochondrial fragmentation, dysregulated glycolysis, and oxidative phosphorylation, as well as increased reactive oxygen species production. Current treatments focus on reducing cellular stress to restore homeostasis through the use of antioxidants or alterations of mitochondrial dynamics. This review focuses on the role of mitochondrial dysfunction in obesity particularly for those suffering from asthma and examines mitochondrial transfer from mesenchymal stem cells to restore function as a potential therapy. Mitochondrial targeted therapy to restore healthy metabolism may provide a unique approach to alleviate dysregulation in individuals with this unique endotype.
Collapse
Affiliation(s)
- Paige Hartsoe
- Department of Medicine, National Jewish Health, Denver, CO 80222, USA
| | - Fernando Holguin
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Hong Wei Chu
- Department of Medicine, National Jewish Health, Denver, CO 80222, USA
| |
Collapse
|
33
|
Kisielewska M, Rakoczy K, Skowron I, Górczyńska J, Kacer J, Bocheńska A, Choromańska A. Utilizing Extracellular Vesicles for Eliminating 'Unwanted Molecules': Harnessing Nature's Structures in Modern Therapeutic Strategies. Molecules 2024; 29:948. [PMID: 38474460 DOI: 10.3390/molecules29050948] [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: 01/14/2024] [Revised: 02/09/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Extracellular vesicles (EVs) are small phospholipid bilayer-bond structures released by diverse cell types into the extracellular environment, maintaining homeostasis of the cell by balancing cellular stress. This article provides a comprehensive overview of extracellular vesicles, their heterogeneity, and diversified roles in cellular processes, emphasizing their importance in the elimination of unwanted molecules. They play a role in regulating oxidative stress, particularly by discarding oxidized toxic molecules. Furthermore, endoplasmic reticulum stress induces the release of EVs, contributing to distinct results, including autophagy or ER stress transmission to following cells. ER stress-induced autophagy is a part of unfolded protein response (UPR) and protects cells from ER stress-related apoptosis. Mitochondrial-derived vesicles (MDVs) also play a role in maintaining homeostasis, as they carry damaged mitochondrial components, thereby preventing inflammation. Moreover, EVs partake in regulating aging-related processes, and therefore they can potentially play a crucial role in anti-aging therapies, including the treatment of age-related diseases such as Alzheimer's disease or cardiovascular conditions. Overall, the purpose of this article is to provide a better understanding of EVs as significant mediators in both physiological and pathological processes, and to shed light on their potential for therapeutic interventions targeting EV-mediated pathways in various pathological conditions, with an emphasis on age-related diseases.
Collapse
Affiliation(s)
| | - Katarzyna Rakoczy
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Izabela Skowron
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Julia Górczyńska
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Julia Kacer
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Agata Bocheńska
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Anna Choromańska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| |
Collapse
|
34
|
Koos B, Unterberg M, Rahmel T, Adamzik M. [Immunometabolism in Sepsis]. Anasthesiol Intensivmed Notfallmed Schmerzther 2024; 59:78-94. [PMID: 38354729 DOI: 10.1055/a-2070-3170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
Immunometabolism is a fascinating field of research that investigates the interactions between metabolic processes and the immune response. This intricate connection plays a pivotal role in regulating inflammatory reactions and consequently exerts a significant impact on the course of sepsis. The proinflammatory response during an immune reaction is closely tied to a high energy demand in immune cells. As a result, proinflammatory immune cells rapidly require substantial amounts of energy in the form of ATP, necessitating a fundamental and swift shift in their metabolism, i.e., their means of generating energy. This entails a marked increase in glycolysis within the proinflammatory response, thereby promptly meeting the energy requirements and providing essential metabolic building blocks for the biosynthesis of macromolecules. Alongside glycolysis, there is heightened activity in the pentose phosphate pathway (PPP). The PPP significantly contributes to NADPH production within the cell, thus maintaining redox equilibrium. Elevated PPP activity consequently leads to an increased NADPH level, resulting in enhanced production of reactive oxygen species (ROS) and nitric oxide (NO). While these molecules are crucial for pathogen elimination, an excess can also induce tissue damage. Simultaneously, there are dual interruptions in the citric acid cycle. In the cellular resting state, the citric acid cycle acts as a sort of "universal processor", where metabolic byproducts of glycolysis, fatty acid breakdown, and amino acid degradation are initially transformed into NADH and FADH2, subsequently yielding ATP. While the citric acid cycle and its connected oxidative phosphorylation predominantly generate energy at rest, it becomes downregulated in the proinflammatory phase of sepsis. The two interruptions lead to an accumulation of citrate and succinate within cells, reflecting mitochondrial dysfunction. Additionally, the significantly heightened glycolysis through fermentation yields lactate, a pivotal metabolite for sepsis diagnosis and prognosis. Conversely, cells in an anti-inflammatory state revert to a metabolic profile akin to the resting state: Glycolysis is attenuated, PPP is suppressed, and the citric acid cycle is reactivated. Of particular interest is that not only does the immune reaction influence metabolic pathways, but this connection also operates in reverse. Thus, modulation of metabolic pathways also modulates the immunity of the corresponding cell and thereby the state of the immune system itself. This could potentially serve as an intriguing avenue in sepsis therapy.
Collapse
|
35
|
Sciacchitano S, Carola V, Nicolais G, Sciacchitano S, Napoli C, Mancini R, Rocco M, Coluzzi F. To Be Frail or Not to Be Frail: This Is the Question-A Critical Narrative Review of Frailty. J Clin Med 2024; 13:721. [PMID: 38337415 PMCID: PMC10856357 DOI: 10.3390/jcm13030721] [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: 11/15/2023] [Revised: 01/07/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Many factors have contributed to rendering frailty an emerging, relevant, and very popular concept. First, many pandemics that have affected humanity in history, including COVID-19, most recently, have had more severe effects on frail people compared to non-frail ones. Second, the increase in human life expectancy observed in many developed countries, including Italy has led to a rise in the percentage of the older population that is more likely to be frail, which is why frailty is much a more common concern among geriatricians compared to other the various health-care professionals. Third, the stratification of people according to the occurrence and the degree of frailty allows healthcare decision makers to adequately plan for the allocation of available human professional and economic resources. Since frailty is considered to be fully preventable, there are relevant consequences in terms of potential benefits both in terms of the clinical outcome and healthcare costs. Frailty is becoming a popular, pervasive, and almost omnipresent concept in many different contexts, including clinical medicine, physical health, lifestyle behavior, mental health, health policy, and socio-economic planning sciences. The emergence of the new "science of frailty" has been recently acknowledged. However, there is still debate on the exact definition of frailty, the pathogenic mechanisms involved, the most appropriate method to assess frailty, and consequently, who should be considered frail. This narrative review aims to analyze frailty from many different aspects and points of view, with a special focus on the proposed pathogenic mechanisms, the various factors that have been considered in the assessment of frailty, and the emerging role of biomarkers in the early recognition of frailty, particularly on the role of mitochondria. According to the extensive literature on this topic, it is clear that frailty is a very complex syndrome, involving many different domains and affecting multiple physiological systems. Therefore, its management should be directed towards a comprehensive and multifaceted holistic approach and a personalized intervention strategy to slow down its progression or even to completely reverse the course of this condition.
Collapse
Affiliation(s)
- Salvatore Sciacchitano
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy;
- Unit of Anaesthesia, Intensive Care and Pain Medicine, Sant’Andrea University Hospital, 00189 Rome, Italy; (M.R.); (F.C.)
- Department of Life Sciences, Health and Health Professions, Link Campus University, 00165 Rome, Italy
| | - Valeria Carola
- Department of Dynamic and Clinical Psychology and Health Studies, Sapienza University of Rome, 00189 Rome, Italy; (V.C.); (G.N.)
| | - Giampaolo Nicolais
- Department of Dynamic and Clinical Psychology and Health Studies, Sapienza University of Rome, 00189 Rome, Italy; (V.C.); (G.N.)
| | - Simona Sciacchitano
- Department of Psychiatry, La Princesa University Hospital, 28006 Madrid, Spain;
| | - Christian Napoli
- Department of Surgical and Medical Science and Translational Medicine, Sapienza University of Rome, 00189 Rome, Italy;
| | - Rita Mancini
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy;
| | - Monica Rocco
- Unit of Anaesthesia, Intensive Care and Pain Medicine, Sant’Andrea University Hospital, 00189 Rome, Italy; (M.R.); (F.C.)
- Department of Surgical and Medical Science and Translational Medicine, Sapienza University of Rome, 00189 Rome, Italy;
| | - Flaminia Coluzzi
- Unit of Anaesthesia, Intensive Care and Pain Medicine, Sant’Andrea University Hospital, 00189 Rome, Italy; (M.R.); (F.C.)
- Department Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy
| |
Collapse
|
36
|
Soto M, Filbert EL, Yang H, Starzinski S, Starzinski A, Gin M, Chen B, Le P, Li T, Bol B, Cheung A, Zhang L, Hsu FJ, Ko A, Fong L, Keenan BP. Neoadjuvant CD40 Agonism Remodels the Tumor Immune Microenvironment in Locally Advanced Esophageal/Gastroesophageal Junction Cancer. CANCER RESEARCH COMMUNICATIONS 2024; 4:200-212. [PMID: 38181044 PMCID: PMC10809910 DOI: 10.1158/2767-9764.crc-23-0550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/26/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024]
Abstract
Sotigalimab is an agonistic anti-CD40 mAb that can modulate antitumor immune responses. In a phase II clinical trial of sotigalimab combined with neoadjuvant chemoradiation (CRT) in locally advanced esophageal/gastroesophageal junction (E/GEJ) cancer with the primary outcome of efficacy as measured by pathologic complete response (pCR) rate, the combination induced pCR in 38% of treated patients. We investigated the mechanism of action of sotigalimab in samples obtained from this clinical trial. Tumor biopsies and peripheral blood samples were collected at baseline, following an initial dose of sotigalimab, and at the time of surgery after CRT completion from six patients. High dimensional single-cell techniques were used, including combined single-cell RNA-sequencing and proteomics (CITEseq) and multiplexed ion beam imaging, to analyze immune responses. Sotigalimab dramatically remodeled the immune compartment in the periphery and within the tumor microenvironment (TME), increasing expression of molecules related to antigen processing and presentation and altering metabolic pathways in myeloid cells. Concomitant with these changes in myeloid cells, sotigalimab treatment primed new T cell clonotypes and increased the density and activation of T cells with enhanced cytotoxic function. Sotigalimab treatment also induced a decrease in the frequency of Tregs in the TME. These findings indicate that a single dose of sotigalimab leads to enhanced antigen presentation that can activate T cells and induce new T cell clones. This restructuring of the TME provides elements which are critical to the development of effective antitumor immune responses and improved clinical outcomes.
Collapse
Affiliation(s)
- Maira Soto
- Pyxis Oncology, Inc., Boston, Massachusetts
- Apexigen America, Inc, San Carlos, California (now a fully owned subsidiary of Pyxis Oncology, Inc.)
| | - Erin L. Filbert
- Apexigen America, Inc, San Carlos, California (now a fully owned subsidiary of Pyxis Oncology, Inc.)
| | - Hai Yang
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Stephanie Starzinski
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Alec Starzinski
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Marissa Gin
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Brandon Chen
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Phi Le
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Tony Li
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Brandon Bol
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Alexander Cheung
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Li Zhang
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California
- Division of Hematology/Oncology, University of California, San Francisco, California
| | - Frank J. Hsu
- Pyxis Oncology, Inc., Boston, Massachusetts
- Apexigen America, Inc, San Carlos, California (now a fully owned subsidiary of Pyxis Oncology, Inc.)
| | - Andrew Ko
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Division of Hematology/Oncology, University of California, San Francisco, California
| | - Lawrence Fong
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Division of Hematology/Oncology, University of California, San Francisco, California
| | - Bridget P. Keenan
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Division of Hematology/Oncology, University of California, San Francisco, California
| |
Collapse
|
37
|
Liang Y, Chen Q, Chang Y, Han J, Yan J, Chen Z, Zhou J. Critical role of FGF21 in diabetic kidney disease: from energy metabolism to innate immunity. Front Immunol 2024; 15:1333429. [PMID: 38312833 PMCID: PMC10834771 DOI: 10.3389/fimmu.2024.1333429] [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: 11/05/2023] [Accepted: 01/08/2024] [Indexed: 02/06/2024] Open
Abstract
Diabetic kidney disease (DKD) stands as the predominant cause of chronic kidney disease (CKD) on a global scale, with its incidence witnessing a consistent annual rise, thereby imposing a substantial burden on public health. The pathogenesis of DKD is primarily rooted in metabolic disorders and inflammation. Recent years have seen a surge in studies highlighting the regulatory impact of energy metabolism on innate immunity, forging a significant area of research interest. Within this context, fibroblast growth factor 21 (FGF21), recognized as an energy metabolism regulator, assumes a pivotal role. Beyond its role in maintaining glucose and lipid metabolism homeostasis, FGF21 exerts regulatory influence on innate immunity, concurrently inhibiting inflammation and fibrosis. Serving as a nexus between energy metabolism and innate immunity, FGF21 has evolved into a therapeutic target for diabetes, nonalcoholic steatohepatitis, and cardiovascular diseases. While the relationship between FGF21 and DKD has garnered increased attention in recent studies, a comprehensive exploration of this association has yet to be systematically addressed. This paper seeks to fill this gap by summarizing the mechanisms through which FGF21 operates in DKD, encompassing facets of energy metabolism and innate immunity. Additionally, we aim to assess the diagnostic and prognostic value of FGF21 in DKD and explore its potential role as a treatment modality for the condition.
Collapse
Affiliation(s)
- Yingnan Liang
- Department of Nephrology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qi Chen
- Department of Nephrology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yue Chang
- Department of Nephrology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Junsong Han
- Department of Nephrology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jiaxin Yan
- Department of Nephrology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zhenjie Chen
- Department of Nephrology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jingwei Zhou
- Department of Nephrology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
38
|
Szabó-Meleg E. Intercellular Highways in Transport Processes. Results Probl Cell Differ 2024; 73:173-201. [PMID: 39242380 DOI: 10.1007/978-3-031-62036-2_9] [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: 09/09/2024]
Abstract
Communication among cells is vital in multicellular organisms. Various structures and mechanisms have evolved over time to achieve the intricate flow of material and information during this process. One such way of communication is through tunnelling membrane nanotubes (TNTs), which were initially described in 2004. These TNTs are membrane-bounded actin-rich cellular extensions, facilitating direct communication between distant cells. They exhibit remarkable diversity in terms of structure, morphology, and function, in which cytoskeletal proteins play an essential role. Biologically, TNTs play a crucial role in transporting membrane components, cell organelles, and nucleic acids, and they also present opportunities for the efficient transmission of bacteria and viruses, furthermore, may contribute to the dissemination of misfolded proteins in certain neurodegenerative diseases. Convincing results of studies conducted both in vitro and in vivo indicate that TNTs play roles in various biomedical processes, including cell differentiation, tissue regeneration, neurodegenerative diseases, immune response and function, as well as tumorigenesis.
Collapse
Affiliation(s)
- Edina Szabó-Meleg
- Department of Biophysics, Medical School, University of Pécs, Pécs, Hungary.
| |
Collapse
|
39
|
Barbero Mazzucca C, Cappellano G, Chiocchetti A. Nutrition, Immunity and Aging: Current Scenario and Future Perspectives in Neurodegenerative Diseases. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:573-587. [PMID: 37138438 DOI: 10.2174/1871527322666230502123255] [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: 07/28/2022] [Revised: 01/17/2023] [Accepted: 02/14/2023] [Indexed: 05/05/2023]
Abstract
Aging is a gradual decline of physiological function and tissue homeostasis and, in many instances, is related to increased (neuro)-degeneration, together with inflammation, becoming one of the most important risks for developing neurodegenerative diseases. Certain individual nutrients or foods in combination may counteract aging and associated neurodegenerative diseases by promoting a balance between the pro- and anti-inflammatory responses. Thus, nutrition could represent a powerful modulator of this fine balance, other than a modifiable risk factor to contrast inflammaging. This narrative review explores from a broad perspective the impact of nutrition on the hallmarks of aging and inflammation in Alzheimer's disease (AD), Parkinson's disease (PD) and Amyotrophic Lateral Sclerosis Syndrome (ALS), starting from nutrients up to single foods and complex dietary patterns.
Collapse
Affiliation(s)
- Camilla Barbero Mazzucca
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Università del Piemonte Orientale, Novara, Italy
| | - Giuseppe Cappellano
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Università del Piemonte Orientale, Novara, Italy
| | - Annalisa Chiocchetti
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease-CAAD, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
40
|
Bogatyreva AI, Gerasimova EV, Kirichenko TV, Markina YV, Tolstik TV, Kiseleva DG, Popkova TV, Markin AM. Mitochondrial DNA copy number in patients with systemic sclerosis. Front Mol Biosci 2023; 10:1313426. [PMID: 38161383 PMCID: PMC10755920 DOI: 10.3389/fmolb.2023.1313426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/04/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction: Systemic scleroderma (SSc) is a chronic autoimmune disease of inflammatory origin. Mitochondrial dysfunction is considered as an important mechanism in the pathogenesis of SSc. Currently mitochondrial DNA (mtDNA) copy number is used as a surrogate marker of mitochondrial dysfunction. Previous studies demonstrate that innate immune cells are important participants in inflammatory and fibrotic processes in SSc. The aim of the study was to evaluate the number of mtDNA copies in CD14+ monocytes and whole blood of patients with SSc in comparison with healthy individuals. Methods: Absolute mtDNA copy number was measured using digital PCR. It was found that the number of mtDNA copies in CD14+ monocytes was significantly higher in patients with SSc compared to control, while the number of mtDNA copies in the whole blood did not have significant differences. Results: The correlation analysis revealed an inverse association of mtDNA copy number with disease duration and the relationship between pro-inflammatory activation of CD14+ monocytes in terms of LPS-stimulated IL-6 secretion and mtDNA copy number. At the same time, basal and LPS-stimulated secretion of IL-6 by cultured CD+ monocytes were significantly higher in SSc group in comparison with control. Discussion: The study results suggest that increase of mtDNA copy number in CD14+ monocytes is a possible mechanism to maintain the reduced function of defective mitochondria in monocytes from patients with SSc associated with the development and progression of SSc.
Collapse
Affiliation(s)
- Anastasia I. Bogatyreva
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Avtsyn Research Institute of Human Morphology of FSBSI “Petrovsky National Research Centre of Surgery”, Moscow, Russia
| | - Elena V. Gerasimova
- Department of Systemic Rheumatic Diseases, VA Nasonova Research Institute of Rheumatology, Moscow, Russia
| | - Tatiana V. Kirichenko
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Avtsyn Research Institute of Human Morphology of FSBSI “Petrovsky National Research Centre of Surgery”, Moscow, Russia
| | - Yuliya V. Markina
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Avtsyn Research Institute of Human Morphology of FSBSI “Petrovsky National Research Centre of Surgery”, Moscow, Russia
| | - Taisiya V. Tolstik
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Avtsyn Research Institute of Human Morphology of FSBSI “Petrovsky National Research Centre of Surgery”, Moscow, Russia
| | - Diana G. Kiseleva
- Faculty of Biology, Department of Biophysics, Lomonosov Moscow State University, Moscow, Russia
| | - Tatiana V. Popkova
- Department of Systemic Rheumatic Diseases, VA Nasonova Research Institute of Rheumatology, Moscow, Russia
| | - Alexander M. Markin
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Avtsyn Research Institute of Human Morphology of FSBSI “Petrovsky National Research Centre of Surgery”, Moscow, Russia
- Medical Institute, Peoples’ Friendship University of Russia named after Patrice Lumumba (RUDN University), Moscow, Russia
| |
Collapse
|
41
|
Tang X, Huang Z, Wang F, Chen J, Qin D, Peng D, Yu B. Macrophage-specific deletion of MIC26 (APOO) mitigates advanced atherosclerosis by increasing efferocytosis. Atherosclerosis 2023; 386:117374. [PMID: 37995600 DOI: 10.1016/j.atherosclerosis.2023.117374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/01/2023] [Accepted: 11/01/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND AND AIMS Recent studies have suggested that MIC26 (apolipoprotein O, APOO), a novel mitochondrial inner membrane protein, is involved in inflammation. Thus, the role of macrophage MIC26 in acute inflammation and chronic inflammatory disease atherosclerosis was investigated. METHODS Macrophage-specific MIC26 knockout mice (MIC26LysM) were generated by crossing Apooflox/flox and LysMcre+/- mice. An endotoxemia mouse model was generated to explore the effects of macrophage MIC26 deficiency on acute inflammation, while an atherosclerosis mouse model was constructed by crossing MIC26LysM mice with Apoe-/- mice and challenged with a Western diet. Atherosclerotic plaques, primary macrophage function, and mitochondrial structure and function were analyzed. RESULTS MIC26 knockout did not affect the median survival time and post-injection serum interleukin 1β concentrations in mice with endotoxemia. Mice with MIC26 deficiency in an Apoe-/- background had smaller atherosclerotic lesions and necrotic core than the control group. In vitro studies found that the loss of MIC26 did not affect macrophage polarization, apoptosis, or lipid handling capacity, but increased efferocytosis (the ability to clear apoptotic cells). An in situ efferocytosis assay of plaques also showed that the ratio of macrophage-associated apoptotic cells to free apoptotic cells was higher in the MIC26-deficient group than in the control group, indicating increased efferocytosis. In addition, an in vivo thymus efferocytosis assay indicated that MIC26 deletion promoted efferocytosis. Mechanistically, the loss of MIC26 resulted in an abnormal mitochondrial inner membrane structure, increased mitochondrial fission, and decreased mitochondrial membrane potential. Loss of MIC26 reduced mitochondria optic atrophy type 1 (OPA1) protein, and OPA1 silencing in macrophages promoted efferocytosis. Overexpression of OPA1 abolished the increase in efferocytosis produced by MIC26 deficiency. CONCLUSIONS Macrophage MIC26 deletion alleviated advanced atherosclerosis and necrotic core expansion by promoting efferocytosis. This mechanism may be related to the increased mitochondrial fission caused by reduced mitochondrial OPA1.
Collapse
Affiliation(s)
- Xiaoyu Tang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Systemic Autoimmune Diseases in Hunan Province, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Zhijie Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Fengjiao Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Jin Chen
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Donglu Qin
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Daoquan Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China.
| | - Bilian Yu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; FuRong Laboratory, Changsha, 410078, Hunan, China.
| |
Collapse
|
42
|
Issara-Amphorn J, Sjoelund VH, Smelkinson M, Montalvo S, Yoon SH, Manes NP, Nita-Lazar A. Myristoylated, alanine-rich C-kinase substrate (MARCKS) regulates toll-like receptor 4 signaling in macrophages. Sci Rep 2023; 13:19562. [PMID: 37949888 PMCID: PMC10638260 DOI: 10.1038/s41598-023-46266-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023] Open
Abstract
MARCKS (myristoylated alanine-rich C-kinase substrate) is a membrane-associated protein expressed in many cell types, including macrophages. MARCKS is functionally implicated in cell adhesion, phagocytosis, and inflammation. LPS (lipopolysaccharide) triggers inflammation via TLR4 (toll-like receptor 4).The presence of MARCKS and the formation of phospho-MARCKS in various cell types have been described, but the role(s) of MARCKS in regulating macrophage functions remain unclear. We investigated the role of MARCKS in inflammation. Confocal microscopy revealed that MARCKS and phospho-MARCKS increased localization to endosomes and the Golgi apparatus upon LPS stimulation.CRISPR-CAS9 mediated knockout of MARCKS in macrophages downregulated the production of TNF and IL6, suggesting a role for MARCKS in inflammatory responses. Our comprehensive proteomics analysis together with real-time metabolic assays comparing LPS-stimulation of WT and MARCKS knock-out macrophages provided insights into the involvement of MARCKS in specific biological processes including innate immune response, inflammatory response, cytokine production, and molecular functions such as extracellularly ATP-gated cation channel activity, electron transfer activity and oxidoreductase activity, uncovering specific proteins involved in regulating MARCKS activity upon LPS stimulation. MARCKS appears to be a key regulator of inflammation whose inhibition might be beneficial for therapeutic intervention in inflammatory diseases.
Collapse
Affiliation(s)
- Jiraphorn Issara-Amphorn
- Functional Cellular Networks Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892-1892, USA
| | - Virginie H Sjoelund
- Functional Cellular Networks Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892-1892, USA
- Barnett Institute, Northeastern University, Boston, MA, 02115, USA
| | - Margery Smelkinson
- Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sebastian Montalvo
- Functional Cellular Networks Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892-1892, USA
| | - Sung Hwan Yoon
- Functional Cellular Networks Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892-1892, USA
| | - Nathan P Manes
- Functional Cellular Networks Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892-1892, USA
| | - Aleksandra Nita-Lazar
- Functional Cellular Networks Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892-1892, USA.
| |
Collapse
|
43
|
Qiao YS, Blackwell TL, Cawthon PM, Coen PM, Cummings SR, Distefano G, Farsijani S, Forman DE, Goodpaster BH, Kritchevsky SB, Mau T, Toledo FGS, Newman AB, Glynn NW. Associations of Objective and Self-Reported Physical Activity and Sedentary Behavior with Skeletal Muscle Energetics: The Study of Muscle, Mobility and Aging (SOMMA). MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.05.23298134. [PMID: 37986749 PMCID: PMC10659463 DOI: 10.1101/2023.11.05.23298134] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Background Skeletal muscle energetics decline with age, and physical activity (PA) has been shown to counteract these declines in older adults. Yet, many studies were based on self-reported PA or structured exercise interventions. We examined the associations of objective daily PA and sedentary behavior (SB) with skeletal muscle energetics and also compared with self-reported PA and SB. We also explored the extent to which PA would attenuate the associations of age with muscle energetics. Methods Among the Study of Muscle, Mobility and Aging (SOMMA) enrolled older adults, 810 (mean age=76±5, 58% women) had maximal muscle oxidative capacity measured ex vivo via high-resolution respirometry of permeabilized myofibers (maxOXPHOS) and in vivo by 31 Phosphorus magnetic resonance spectroscopy (ATP max ). Objective PA was measured using the wrist-worn ActiGraph GT9X over 7-days to capture sedentary behavior (SB), light, and moderate-to-vigorous PA (MVPA). Self-reported SB, MVPA, and all exercise-related PA were assessed with The Community Healthy Activities Model Program for Seniors questionnaire. Linear regression models with progressive covariate adjustments evaluated the associations between SB, PA and muscle energetics, and the attenuation of the age / muscle energetic association by PA. Results Every 30 minutes more objective MVPA was associated with 0.65 pmol/s*mg higher maxOXPHOS and 0.012 mM/sec higher ATP max , after adjustment for age, site/technician and sex. More time spent in objective light+MVPA was significantly associated with higher ATP max , but not maxOXPHOS. In contrast, every 30 minutes spent in objective SB was associated with 0.43 pmol/s*mg lower maxOXPHOS and 0.004 mM/sec lower ATP max . Only associations with ATP max held after further adjusting for socioeconomic status, body mass index, lifestyle factors and multimorbidities. Self-reported MVPA and all exercise-related activities, but not SB, yielded similar associations with maxOXPHOS and ATP max . Lastly, age was only significantly associated with muscle energetics in men. Adjusting for objective time spent in MVPA attenuated the age association with ATP max by nearly 60% in men. Conclusion More time spent in daily PA, especially MVPA, were associated with higher muscle energetics. Interventions that increase higher intensity activity might offer potential therapeutic interventions to slow the age-related decline in muscle energetics. Our work also emphasizes the importance of taking PA into consideration when evaluating associations related to skeletal muscle energetics.
Collapse
|
44
|
Li AL, Lian L, Chen XN, Cai WH, Fan XB, Fan YJ, Li TT, Xie YY, Zhang JP. The role of mitochondria in myocardial damage caused by energy metabolism disorders: From mechanisms to therapeutics. Free Radic Biol Med 2023; 208:236-251. [PMID: 37567516 DOI: 10.1016/j.freeradbiomed.2023.08.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/24/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023]
Abstract
Myocardial damage is the most serious pathological consequence of cardiovascular diseases and an important reason for their high mortality. In recent years, because of the high prevalence of systemic energy metabolism disorders (e.g., obesity, diabetes mellitus, and metabolic syndrome), complications of myocardial damage caused by these disorders have attracted widespread attention. Energy metabolism disorders are independent of traditional injury-related risk factors, such as ischemia, hypoxia, trauma, and infection. An imbalance of myocardial metabolic flexibility and myocardial energy depletion are usually the initial changes of myocardial injury caused by energy metabolism disorders, and abnormal morphology and functional destruction of the mitochondria are their important features. Specifically, mitochondria are the centers of energy metabolism, and recent evidence has shown that decreased mitochondrial function, caused by an imbalance in mitochondrial quality control, may play a key role in myocardial injury caused by energy metabolism disorders. Under chronic energy stress, mitochondria undergo pathological fission, while mitophagy, mitochondrial fusion, and biogenesis are inhibited, and mitochondrial protein balance and transfer are disturbed, resulting in the accumulation of nonfunctional and damaged mitochondria. Consequently, damaged mitochondria lead to myocardial energy depletion and the accumulation of large amounts of reactive oxygen species, further aggravating the imbalance in mitochondrial quality control and forming a vicious cycle. In addition, impaired mitochondria coordinate calcium homeostasis imbalance, and epigenetic alterations participate in the pathogenesis of myocardial damage. These pathological changes induce rapid progression of myocardial damage, eventually leading to heart failure or sudden cardiac death. To intervene more specifically in the myocardial damage caused by metabolic disorders, we need to understand the specific role of mitochondria in this context in detail. Accordingly, promising therapeutic strategies have been proposed. We also summarize the existing therapeutic strategies to provide a reference for clinical treatment and developing new therapies.
Collapse
Affiliation(s)
- Ao-Lin Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Lu Lian
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Xin-Nong Chen
- Department of Traditional Chinese Medicine, Tianjin First Central Hospital, Tianjin, 300190, China
| | - Wen-Hui Cai
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Xin-Biao Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Ya-Jie Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Ting-Ting Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Ying-Yu Xie
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Jun-Ping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China.
| |
Collapse
|
45
|
Dong Z, Wu L, Hong H. Mitochondrial Dysfunction in the Pathogenesis and Treatment of Oral Inflammatory Diseases. Int J Mol Sci 2023; 24:15483. [PMID: 37895162 PMCID: PMC10607498 DOI: 10.3390/ijms242015483] [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: 09/09/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
Oral inflammatory diseases (OIDs) include many common diseases such as periodontitis and pulpitis. The causes of OIDs consist microorganism, trauma, occlusal factors, autoimmune dis-eases and radiation therapy. When treated unproperly, such diseases not only affect oral health but also pose threat to people's overall health condition. Therefore, identifying OIDs at an early stage and exploring new therapeutic strategies are important tasks for oral-related research. Mitochondria are crucial organelles for many cellular activities and disruptions of mitochondrial function not only affect cellular metabolism but also indirectly influence people's health and life span. Mitochondrial dysfunction has been implicated in many common polygenic diseases, including cardiovascular and neurodegenerative diseases. Recently, increasing evidence suggests that mitochondrial dysfunction plays a critical role in the development and progression of OIDs and its associated systemic diseases. In this review, we elucidated the critical insights into mitochondrial dysfunction and its involvement in the inflammatory responses in OIDs. We also summarized recent research progresses on the treatment of OIDs targeting mitochondrial dysfunction and discussed the underlying mechanisms.
Collapse
Affiliation(s)
- Zhili Dong
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; (Z.D.); (L.W.)
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Liping Wu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; (Z.D.); (L.W.)
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Hong Hong
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; (Z.D.); (L.W.)
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| |
Collapse
|
46
|
Borja-Magno AI, Furuzawa-Carballeda J, Guevara-Cruz M, Arias C, Granados J, Bourges H, Tovar AR, Sears B, Noriega LG, Gómez FE. Supplementation with EPA and DHA omega-3 fatty acids improves peripheral immune cell mitochondrial dysfunction and inflammation in subjects with obesity. J Nutr Biochem 2023; 120:109415. [PMID: 37437746 DOI: 10.1016/j.jnutbio.2023.109415] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/01/2023] [Accepted: 07/07/2023] [Indexed: 07/14/2023]
Abstract
Omega-3 fatty acids (w-3 FA) have anti-inflammatory effects and improve mitochondrial function. Nonetheless, little is known about their effect on mitochondrial bioenergetics of peripheral blood mononuclear cells (PBMCs) in individuals with obesity. Thus, this study aimed to determine the mitochondrial bioenergetics status and cell subset composition of PBMCs during obesity, before and after 1 month supplementation with w-3 FA. We performed a case-control study with twelve women with normal BMI (lean group) and 19 with grade 2 obesity (obese group), followed by a before-after prospective study where twelve subjects with obesity received a 1 month intervention with 5.25 g of w-3 FA (3.5 g eicosapentaenoic (EPA) and 1.75 g docosahexaenoic (DHA) acids), and obtained PBMCs from all participants. Mitochondrial bioenergetic markers, including basal and ATP-production associated respiration, proton leak, and nonmitochondrial respiration, were higher in PBMCs from the obese group vs. the lean group. The bioenergetic health index (BHI), a marker of mitochondrial function, was lower in the obese vs. the lean group. In addition, Th1, Th2, Th17, CD4+ Tregs, CD8+ Tregs, and Bregs, M1 monocytes and pDCreg cells were higher in PBMCs from the obese group vs. the lean group. The w-3 FA intervention improved mitochondrial function, mainly by decreasing nonmitochondrial respiration and increasing the reserve respiratory capacity and BHI. The intervention also reduced circulating pro-inflammatory and anti-inflammatory lymphocyte and monocytes subsets in individuals with obesity. The mitochondrial dysfunction of PBMCs and the higher proportion of peripheral pro-inflammatory and anti-inflammatory immune cells in subjects with obesity, improved with 1 month supplementation with EPA and DHA.
Collapse
Affiliation(s)
- Angélica I Borja-Magno
- Department of Nutritional Physiology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México, Mexico
| | - Janette Furuzawa-Carballeda
- Department of Experimental Surgery, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México, Mexico
| | - Martha Guevara-Cruz
- Department of Nutritional Physiology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México, Mexico
| | - Clorinda Arias
- Department of Genomics Medicine and Environmental Toxicology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Coyoacan, Ciudad de México, Mexico
| | - Julio Granados
- Department of Transplants, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México, Mexico
| | - Hector Bourges
- Divission of Nutrition, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México, Mexico
| | - Armando R Tovar
- Department of Nutritional Physiology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México, Mexico
| | - Barry Sears
- Inflammation Research Foundation, Peabody, Massachusetts, USA
| | - Lilia G Noriega
- Department of Nutritional Physiology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México, Mexico.
| | - Francisco Enrique Gómez
- Department of Nutritional Physiology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México, Mexico.
| |
Collapse
|
47
|
Prabhu SS, Nair AS, Nirmala SV. Multifaceted roles of mitochondrial dysfunction in diseases: from powerhouses to saboteurs. Arch Pharm Res 2023; 46:723-743. [PMID: 37751031 DOI: 10.1007/s12272-023-01465-y] [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: 02/05/2023] [Accepted: 09/19/2023] [Indexed: 09/27/2023]
Abstract
The fact that mitochondria play a crucial part in energy generation has led to the nickname "powerhouses" of the cell being applied to them. They also play a significant role in many other cellular functions, including calcium signalling, apoptosis, and the creation of vital biomolecules. As a result, cellular function and health as a whole can be significantly impacted by mitochondrial malfunction. Indeed, malignancies frequently have increased levels of mitochondrial biogenesis and quality control. Adverse selection exists for harmful mitochondrial genome mutations, even though certain malignancies include modifications in the nuclear-encoded tricarboxylic acid cycle enzymes that generate carcinogenic metabolites. Since rare human cancers with mutated mitochondrial genomes are often benign, removing mitochondrial DNA reduces carcinogenesis. Therefore, targeting mitochondria offers therapeutic options since they serve several functions and are crucial to developing malignant tumors. Here, we discuss the various steps involved in the mechanism of cancer for which mitochondria plays a significant role, as well as the role of mitochondria in diseases other than cancer. It is crucial to understand mitochondrial malfunction to target these organelles for therapeutic reasons. This highlights the significance of investigating mitochondrial dysfunction in cancer and other disease research.
Collapse
Affiliation(s)
- Surapriya Surendranath Prabhu
- Department of Pharmaceutical Chemistry and Analysis, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - Aathira Sujathan Nair
- Department of Pharmaceutical Chemistry and Analysis, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - Saiprabha Vijayakumar Nirmala
- Department of Pharmaceutical Chemistry and Analysis, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India.
| |
Collapse
|
48
|
Kang EY, Kim DY, Kim HK, Shin WS, Park YS, Kim TH, Kim W, Cao L, Lee SG, Gang G, Shin M, Kim JM, Go GW. Modified Korean MIND Diet: A Nutritional Intervention for Improved Cognitive Function in Elderly Women through Mitochondrial Respiration, Inflammation Suppression, and Amino Acid Metabolism Regulation. Mol Nutr Food Res 2023; 67:e2300329. [PMID: 37650267 DOI: 10.1002/mnfr.202300329] [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: 05/21/2023] [Revised: 07/19/2023] [Indexed: 09/01/2023]
Abstract
SCOPE Mild cognitive impairment is associated with a high prevalence of dementia. The study examines the benefits of a modified Korean MIND (K-MIND) diet and explores biomarkers using multi-omics analysis. METHODS AND RESULTS The K-MIND diet, tailored to the elderly Korean population, includes perilla oil, milk, or fermented milk, and avoids alcohol consumption. As a result, the K-MIND diet significantly improves subjects "orientation to place" in the Korean version of the Mini-Mental State Examination, 2nd edition test. According to multi-omics analysis, the K-MIND diet upregulates genes associated with mitochondrial respiration, including ubiquinone oxidoreductase, cytochrome C oxidase, and ATP synthase, and immune system processes, and downregulates genes related to nuclear factor kappa B activity and inflammatory responses. In addition, K-MIND affects the metabolic pathways of glycine, serine, threonine, tryptophan, and sphingolipids, which are closely linked to cognitive function through synthesis of neurotransmitters and structures of brain cell membranes. CONCLUSION The findings imply that the K-MIND diet improves cognitive function by upregulating key genes involved in oxidative phosphorylation and downregulating pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Eun Young Kang
- Department of Food and Nutrition, Hanyang University, Seoul, 04763, Republic of Korea
| | - Do-Young Kim
- Department of Animal Science and Technology, Chung-Ang University, Gyeonggi-do, 17546, Republic of Korea
| | - Hyun Kyung Kim
- Department of Food and Nutrition, Hanyang University, Seoul, 04763, Republic of Korea
| | - Weon-Sun Shin
- Department of Food and Nutrition, Hanyang University, Seoul, 04763, Republic of Korea
| | - Young-Sook Park
- Department of Food and Nutrition, Hanyang University, Seoul, 04763, Republic of Korea
| | - Tae Hoon Kim
- Department of Food and Nutrition, Hanyang University, Seoul, 04763, Republic of Korea
| | - Wooki Kim
- Department of Food Science and Biotechnology, Kyung Hee University, Gyeonggi-do, 17104, Republic of Korea
| | - Lei Cao
- Department of Food Science and Biotechnology, Gachon University, Seongnam, 13120, Republic of Korea
| | - Sang-Gil Lee
- Department of Food Science and Nutrition, Pukyong National University, Busan, 48513, Republic of Korea
- Department of Smart Green Technology Engineering, Pukyong National University, Busan, 48513, Republic of Korea
| | - Gyoungok Gang
- Department of Food Science and Nutrition, Pukyong National University, Busan, 48513, Republic of Korea
| | - Minhye Shin
- Department of Microbiology, Inha University, Incheon, 22212, Republic of Korea
| | - Jun-Mo Kim
- Department of Animal Science and Technology, Chung-Ang University, Gyeonggi-do, 17546, Republic of Korea
| | - Gwang-Woong Go
- Department of Food and Nutrition, Hanyang University, Seoul, 04763, Republic of Korea
| |
Collapse
|
49
|
Lyu Y, Wang T, Huang S, Zhang Z. Mitochondrial Damage-Associated Molecular Patterns and Metabolism in the Regulation of Innate Immunity. J Innate Immun 2023; 15:665-679. [PMID: 37666239 PMCID: PMC10601681 DOI: 10.1159/000533602] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/10/2023] [Indexed: 09/06/2023] Open
Abstract
The innate immune system, as the host's first line of defense against intruders, plays a critical role in recognizing, identifying, and reacting to a wide range of microbial intruders. There is increasing evidence that mitochondrial stress is a major initiator of innate immune responses. When mitochondria's integrity is disrupted or dysfunction occurs, the mitochondria's contents are released into the cytosol. These contents, like reactive oxygen species, mitochondrial DNA, and double-stranded RNA, among others, act as damage-related molecular patterns (DAMPs) that can bind to multiple innate immune sensors, particularly pattern recognition receptors, thereby leading to inflammation. To avoid the production of DAMPs, in addition to safeguarding organelles integrity and functionality, mitochondria may activate mitophagy or apoptosis. Moreover, mitochondrial components and specific metabolic regulations modify properties of innate immune cells. These include macrophages, dendritic cells, innate lymphoid cells, and so on, in steady state or in stimulation that are involved in processes ranging from the tricarboxylic acid cycle to oxidative phosphorylation and fatty acid metabolism. Here we provide a brief summary of mitochondrial DAMPs' initiated and potentiated inflammatory response in the innate immune system. We also provide insights into how the state of activation, differentiation, and functional polarization of innate immune cells can be influenced by alteration to the metabolic pathways in mitochondria.
Collapse
Affiliation(s)
- Yanmin Lyu
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Tianyu Wang
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shuhong Huang
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhaoqiang Zhang
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
50
|
Zhao M, Zuo C, Hao H, Xing X, Zhao L, Li N. A patient with MELAS syndrome combined with autoimmune abnormalities: a case report. Front Neurol 2023; 14:1239664. [PMID: 37693769 PMCID: PMC10484216 DOI: 10.3389/fneur.2023.1239664] [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: 06/13/2023] [Accepted: 08/02/2023] [Indexed: 09/12/2023] Open
Abstract
Background Mitochondrial encephalomyopathy with lactic acidosis and stroke-like episodes (MELAS) is a group of maternally inherited disorders caused by mutations or deletions in mitochondrial genes with mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes as the main clinical manifestations. Case presentation We reported a 20-year-old female patient with MELAS syndrome combined with autoimmune abnormalities. She suffered from an intermittent headache in the right temporal region with no obvious cause, and then, after strenuous exercise in dance class, the headache became aggravated, accompanied by unresponsiveness, blurred vision, and diplopia. Her blood lactate levels were elevated, her antinuclear antibodies were positive, and the antimetabolic glutamate receptors 5 in her serum were positive. Brain DWI showed a hypertensive signal in the right temporo-parietal-occipital cortex and subcortical area. Brain MRS showed decreased NAA peak and increased Lac peak. Muscle biopsy showed myogenic damage, and the modified Gomori trichrome (MGT) staining showed ragged red fibers (RRF). A genetic study revealed a mitochondrial DNA A3243G mutation. Conclusion Mitochondrial encephalomyopathy is a rare clinical condition; however, the association with autoimmune diseases is not yet clear and still needs further research and analysis.
Collapse
Affiliation(s)
- Mingmin Zhao
- Graduate School of Hebei North University, Zhangjiakou, Hebei, China
- Neurological Intensive Care Unit, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Chun Zuo
- Graduate School of Hebei North University, Zhangjiakou, Hebei, China
- Neurological Intensive Care Unit, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Hongyu Hao
- Neurological Intensive Care Unit, Hebei General Hospital, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Shijiazhuang, Hebei, China
| | - Xing Xing
- Neurological Intensive Care Unit, Hebei General Hospital, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Shijiazhuang, Hebei, China
| | - Lei Zhao
- Neurological Intensive Care Unit, Hebei General Hospital, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Shijiazhuang, Hebei, China
| | - Na Li
- Neurological Intensive Care Unit, Hebei General Hospital, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Shijiazhuang, Hebei, China
| |
Collapse
|