1
|
Zhao X, Zhang J, Li C, Kuang W, Deng J, Tan X, Li C, Li S. Mitochondrial mechanisms in Treg cell regulation: Implications for immunotherapy and disease treatment. Mitochondrion 2024:101975. [PMID: 39491776 DOI: 10.1016/j.mito.2024.101975] [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: 06/11/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
Regulatory T cells (Tregs) play a critical role in maintaining immune homeostasis and preventing autoimmune diseases. Recent advances in immunometabolism have revealed the pivotal role of mitochondrial dynamics and metabolism in shaping Treg functionality. Tregs depend on oxidative phosphorylation (OXPHOS) and fatty acid oxidation (FAO) to support their suppressive functions and long-term survival. Mitochondrial processes such as fusion and fission significantly influence Treg activity, with mitochondrial fusion enhancing bioenergetic efficiency and reducing reactive oxygen species (ROS) production, thereby promoting Treg stability. In contrast, excessive mitochondrial fission disrupts ATP synthesis and elevates ROS levels, impairing Treg suppressive capacity. Furthermore, mitochondrial ROS act as critical signaling molecules in Treg regulation, where controlled levels stabilize FoxP3 expression, but excessive ROS leads to mitochondrial dysfunction and immune dysregulation. Mitophagy, as part of mitochondrial quality control, also plays an essential role in preserving Treg function. Understanding the intricate interplay between mitochondrial dynamics and Treg metabolism provides valuable insights for developing novel therapeutic strategies to treat autoimmune disorders and enhance immunotherapy in cancer.
Collapse
Affiliation(s)
- Xiaozhen Zhao
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Junmei Zhang
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Caifeng Li
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China.
| | - Weiying Kuang
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Jianghong Deng
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Xiaohua Tan
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Chao Li
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Shipeng Li
- Department of Rheumatology, National Centre for Children's Health Beijing Children's Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Wang H, Kim SJ, Lei Y, Wang S, Wang H, Huang H, Zhang H, Tsung A. Neutrophil extracellular traps in homeostasis and disease. Signal Transduct Target Ther 2024; 9:235. [PMID: 39300084 DOI: 10.1038/s41392-024-01933-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 06/25/2024] [Accepted: 07/16/2024] [Indexed: 09/22/2024] Open
Abstract
Neutrophil extracellular traps (NETs), crucial in immune defense mechanisms, are renowned for their propensity to expel decondensed chromatin embedded with inflammatory proteins. Our comprehension of NETs in pathogen clearance, immune regulation and disease pathogenesis, has grown significantly in recent years. NETs are not only pivotal in the context of infections but also exhibit significant involvement in sterile inflammation. Evidence suggests that excessive accumulation of NETs can result in vessel occlusion, tissue damage, and prolonged inflammatory responses, thereby contributing to the progression and exacerbation of various pathological states. Nevertheless, NETs exhibit dual functionalities in certain pathological contexts. While NETs may act as autoantigens, aggregated NET complexes can function as inflammatory mediators by degrading proinflammatory cytokines and chemokines. The delineation of molecules and signaling pathways governing NET formation aids in refining our appreciation of NETs' role in immune homeostasis, inflammation, autoimmune diseases, metabolic dysregulation, and cancer. In this comprehensive review, we delve into the multifaceted roles of NETs in both homeostasis and disease, whilst discussing their potential as therapeutic targets. Our aim is to enhance the understanding of the intricate functions of NETs across the spectrum from physiology to pathology.
Collapse
Affiliation(s)
- Han Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Susan J Kim
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Yu Lei
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuhui Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui Wang
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hai Huang
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Hongji Zhang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Allan Tsung
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
3
|
Wei J, Wang A, Li B, Li X, Yu R, Li H, Wang X, Wang Y, Zhu M. Pathological mechanisms and crosstalk among various cell death pathways in cardiac involvement of systemic lupus erythematosus. Front Immunol 2024; 15:1452678. [PMID: 39301029 PMCID: PMC11410571 DOI: 10.3389/fimmu.2024.1452678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/19/2024] [Indexed: 09/22/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a prevalent autoimmune disease primarily characterized by the involvement of multiple systems and organs. Cardiovascular disease is the primary cause of mortality in patients with SLE, though the mechanisms underlying the increased cardiovascular risk in SLE patients remain unclear. Recent studies indicate that abnormal activation of programmed cell death (PCD) signaling and the crosstalk among various forms of cell death are critical in the immunopathogenesis of SLE. Furthermore, apoptosis, necroptosis, pyroptosis, NETosis, and ferroptosis are recognized as key cellular processes in the pathogenesis of SLE and are closely linked to cardiac involvement. This review uniquely explores the intricate crosstalk between apoptosis, necroptosis, and other cell death pathways, discussing their roles and interactions in the pathogenesis of cardiac involvement in SLE. Investigating the interplay between PCD signaling and cardiac involvement in SLE in understanding the disease's underlying mechanisms and offers opportunities for new therapeutic interventions. The integration of precision medicine and innovative strategies targeting these complex pathways holds promise for enhancing the treatment prospects of SLE with cardiac involvement.
Collapse
Affiliation(s)
- Jingjing Wei
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Aolong Wang
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Bin Li
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Henan Evidence-based Medicine Center of Chinese Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xingyuan Li
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Rui Yu
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Haitao Li
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xinlu Wang
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Yongxia Wang
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Mingjun Zhu
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
4
|
Danieli MG, Antonelli E, Longhi E, Gangemi S, Allegra A. The role of microbiota and oxidative stress axis and the impact of intravenous immunoglobulin in systemic lupus erythematosus. Autoimmun Rev 2024; 23:103607. [PMID: 39187222 DOI: 10.1016/j.autrev.2024.103607] [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: 05/25/2024] [Accepted: 05/30/2024] [Indexed: 08/28/2024]
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease characterized by widespread inflammation affecting various organs. This review discusses the role of oxidative stress and gut microbiota in the pathogenesis of SLE and evaluates the therapeutic potential of intravenous immunoglobulins (IVIg). Oxidative stress contributes to SLE by causing impairment in the function of mitochondria, resulting in reactive oxygen species production, which triggers autoantigenicity and proinflammatory cytokines. Gut microbiota also plays a significant role in SLE. Dysbiosis has been associated to disease's onset and progression. Moreover, dysbiosis exacerbates SLE symptoms and influences systemic immunity, leading to a breakdown in bacterial tolerance and an increase in inflammatory responses. High-dose IVIg has emerged as a promising treatment for refractory cases of SLE. The beneficial effects of IVIg are partly due to its antioxidant property, reducing oxidative stress markers and modulating the immune responses. Additionally, IVIg can normalize the gut flora, as demonstrated in a case of severe intestinal pseudo-obstruction. In summary, both oxidative stress and dysregulation of microbiota are pivotal in the pathogenesis of SLE. The use of IVIg may improve the disease's outcome. Future research should be directed to elucidating the precise mechanisms by which oxidative stress and microbiota are linked with autoimmunity in SLE in developing targeted therapies.
Collapse
Affiliation(s)
- Maria Giovanna Danieli
- Postgraduate School of Allergy and Clinical Immunology, Università Politecnica delle Marche, 60126 Ancona, Italy; SOS Immunologia delle Malattie rare e dei Trapianti, AOU delle Marche, Ancona, Italy.
| | - Eleonora Antonelli
- Postgraduate School of Internal Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy
| | - Eleonora Longhi
- Postgraduate School in Clinical Pathology and Clinical Biochemistry, Università G. D'Annunzio Chieti -Pescara, 66100 Chieti, Italy.
| | - Sebastiano Gangemi
- Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy.
| | - Alessandro Allegra
- Division of Haematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy.
| |
Collapse
|
5
|
Akhil A, Bansal R, Ankita A, Kaur H, Monika M, Bhatnagar A. Disturbance in communication between mitochondrial redox processes and the AMPK/PGC-1α/SIRT-1 axis influences diverse organ symptoms in lupus-affected mice. Mitochondrion 2024; 78:101930. [PMID: 39025320 DOI: 10.1016/j.mito.2024.101930] [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: 02/28/2024] [Revised: 06/28/2024] [Accepted: 07/07/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Mechanisms behind multiple organ involvement in lupus, is still an enigma for researchers. Mitochondrial dysfunction and oxidative stress are known to be important aspects in lupus etiology however, their role in lupus organ manifestation is yet to be understood. The present study is based on the understanding of interplay between AMPK/PGC-1α/SIRT-1 axis, mitochondrial complexes, and anti-oxidants levels, which might be involved in lupus organ pathology. METHODOLOGY Pristane-induced Balb/c mice lupus model (PIL) was utilised and evaluation of anti-oxidants, mitochondrial complexes, pro-inflammatory cytokines levels, biochemical parameters were performed by standard procedures. Tissues were studied by haematoxylin and eosin staining followed by immunohistochemistry. The AMPK/PGC-1α/SIRT-1 expression was analysed by using qPCR and flowcytometry. Analysis of reactive oxygen species (ROS) among WBCs was performed by using various dyes (DCFDA, Mitosox, JC-1) on flowcytometry. RESULT Significant presence of immune complexes (Tissue sections), ANA (Serum), and pro-inflammatory cytokines (plasma), diminished anti-oxidants and altered biochemical parameters depict the altered pathology in PIL which was accompanied by dysregulated mitochondrial complex activity. Differential expression of the AMPK/PGC-1α/SIRT-1 axis was detected in tissue and correlation with mitochondrial and antioxidant activity emerged as negative in PIL group while positive in controls. Close association was observed between ROS, mitochondrial membrane potential, and AMPK/PGC-1α/SIRT-1 axis in WBCs. CONCLUSION This study concludes that mitochondria play a dual role in lupus organ pathology, contributing to organ damage while also potentially protecting against damage through the regulation of interactions between antioxidants and the AMPK axis expression.
Collapse
Affiliation(s)
- Akhil Akhil
- Department of Biochemistry, BMS-Block II, South Campus, Panjab University, Chandigarh 160014 India
| | - Rohit Bansal
- Department of Biochemistry, BMS-Block II, South Campus, Panjab University, Chandigarh 160014 India
| | - Ankita Ankita
- Department of Biochemistry, BMS-Block II, South Campus, Panjab University, Chandigarh 160014 India
| | - Harsimran Kaur
- Department of Biochemistry, BMS-Block II, South Campus, Panjab University, Chandigarh 160014 India
| | - Monika Monika
- Department of Biochemistry, BMS-Block II, South Campus, Panjab University, Chandigarh 160014 India
| | - Archana Bhatnagar
- Department of Biochemistry, BMS-Block II, South Campus, Panjab University, Chandigarh 160014 India.
| |
Collapse
|
6
|
Patiño-Martinez E, Nakabo S, Jiang K, Carmona-Rivera C, Tsai WL, Claybaugh D, Yu ZX, Romero A, Bohrnsen E, Schwarz B, Solís-Barbosa MA, Blanco LP, Naqi M, Temesgen-Oyelakin Y, Davis M, Manna Z, Gupta S, Mehta N, Naz F, dell'Orso S, Hasni S, Kaplan MJ. The Aconitate Decarboxylase 1/Itaconate Pathway Modulates Immune Dysregulation and Associates with Cardiovascular Disease Markers and Disease Activity in Systemic Lupus Erythematosus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:419-434. [PMID: 38949522 DOI: 10.4049/jimmunol.2400241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/10/2024] [Indexed: 07/02/2024]
Abstract
The Krebs cycle enzyme aconitate decarboxylase 1 (ACOD1) mediates itaconate synthesis in monocytes and macrophages. Previously, we reported that administration of 4-octyl itaconate to lupus-prone mice abrogated immune dysregulation and clinical features. In this study, we explore the role of the endogenous ACOD1/itaconate pathway in the development of TLR7-induced lupus (imiquimod [IMQ] model). We found that, in vitro, ACOD1 was induced in mouse bone marrow-derived macrophages and human monocyte-derived macrophages following TLR7 stimulation. This induction was partially dependent on type I IFN receptor signaling and on specific intracellular pathways. In the IMQ-induced mouse model of lupus, ACOD1 knockout (Acod1-/-) displayed disruptions of the splenic architecture, increased serum levels of anti-dsDNA and proinflammatory cytokines, and enhanced kidney immune complex deposition and proteinuria, when compared with the IMQ-treated wild-type mice. Consistent with these results, Acod1-/- bone marrow-derived macrophages treated in vitro with IMQ showed higher proinflammatory features. Furthermore, itaconate serum levels in systemic lupus erythematosus patients were decreased compared with healthy individuals, in association with disease activity and specific perturbed cardiometabolic parameters. These findings suggest that the ACOD1/itaconate pathway plays important immunomodulatory and vasculoprotective roles in systemic lupus erythematosus, supporting the potential therapeutic role of itaconate analogs in autoimmune diseases.
Collapse
Affiliation(s)
- Eduardo Patiño-Martinez
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Shuichiro Nakabo
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Kan Jiang
- Biodata Mining and Discovery Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Carmelo Carmona-Rivera
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Wanxia Li Tsai
- Translational Immunology Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Dillon Claybaugh
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Zu-Xi Yu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Aracely Romero
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Eric Bohrnsen
- Protein & Chemistry Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT
| | - Benjamin Schwarz
- Protein & Chemistry Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT
| | - Miguel A Solís-Barbosa
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Luz P Blanco
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Mohammad Naqi
- Lupus Clinical Trials Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Yenealem Temesgen-Oyelakin
- Lupus Clinical Trials Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Michael Davis
- Lupus Clinical Trials Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Zerai Manna
- Lupus Clinical Trials Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Sarthak Gupta
- Lupus Clinical Trials Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Nehal Mehta
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Faiza Naz
- Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Stefania dell'Orso
- Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Sarfaraz Hasni
- Lupus Clinical Trials Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
7
|
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 2024:10.1007/s12013-024-01443-1. [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] [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
|
8
|
Halfon M, Tankeu AT, Ribi C. Mitochondrial Dysfunction in Systemic Lupus Erythematosus with a Focus on Lupus Nephritis. Int J Mol Sci 2024; 25:6162. [PMID: 38892349 PMCID: PMC11173067 DOI: 10.3390/ijms25116162] [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: 05/01/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease affecting mostly women of child-bearing age. Immune dysfunction in SLE results from disrupted apoptosis which lead to an unregulated interferon (IFN) stimulation and the production of autoantibodies, leading to immune complex formation, complement activation, and organ damage. Lupus nephritis (LN) is a common and severe complication of SLE, impacting approximately 30% to 40% of SLE patients. Recent studies have demonstrated an alteration in mitochondrial homeostasis in SLE patients. Mitochondrial dysfunction contributes significantly to SLE pathogenesis by enhancing type 1 IFN production through various pathways involving neutrophils, platelets, and T cells. Defective mitophagy, the process of clearing damaged mitochondria, exacerbates this cycle, leading to increased immune dysregulation. In this review, we aim to detail the physiopathological link between mitochondrial dysfunction and disease activity in SLE. Additionally, we will explore the potential role of mitochondria as biomarkers and therapeutic targets in SLE, with a specific focus on LN. In LN, mitochondrial abnormalities are observed in renal cells, correlating with disease progression and renal fibrosis. Studies exploring cell-free mitochondrial DNA as a biomarker in SLE and LN have shown promising but preliminary results, necessitating further validation and standardization. Therapeutically targeting mitochondrial dysfunction in SLE, using drugs like metformin or mTOR inhibitors, shows potential in modulating immune responses and improving clinical outcomes. The interplay between mitochondria, immune dysregulation, and renal involvement in SLE and LN underscores the need for comprehensive research and innovative therapeutic strategies. Understanding mitochondrial dynamics and their impact on immune responses offers promising avenues for developing personalized treatments and non-invasive biomarkers, ultimately improving outcomes for LN patients.
Collapse
Affiliation(s)
- Matthieu Halfon
- Transplantation Center, Lausanne University Hospital, Rue du Bugnon 44, CH-1010 Lausanne, Switzerland;
| | - Aurel T. Tankeu
- Transplantation Center, Lausanne University Hospital, Rue du Bugnon 44, CH-1010 Lausanne, Switzerland;
| | - Camillo Ribi
- Division of Immunology and Allergy, Lausanne University Hospital, CH-1010 Lausanne, Switzerland;
| |
Collapse
|
9
|
Hisada R, Kono M. Potential therapies targeting metabolic pathways in systemic lupus erythematosus. Clin Immunol 2024; 263:110224. [PMID: 38648959 DOI: 10.1016/j.clim.2024.110224] [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: 01/12/2024] [Revised: 03/15/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
The pathophysiology of systemic lupus erythematosus (SLE) is multifactorial and involves alterations in metabolic pathways, including glycolysis, lipid metabolism, amino acid metabolism, and mitochondrial dysfunction. Increased glycolysis in SLE T cells, which is associated with elevated glucose transporter 1 expression, suggests targeting glucose transporters and hexokinase as potential treatments. Abnormalities in lipid metabolism, particularly in lipid rafts and enzymes, present new therapeutic targets. This review discusses how changes in glutaminolysis and tryptophan metabolism affect T-cell function, suggesting new therapeutic interventions, as well as mitochondrial dysfunction in SLE, which increases reactive oxygen species. The review also emphasizes that modulating metabolic pathways in immune cells is a promising approach for SLE treatment, and can facilitate personalized therapies based on individual metabolic profiles of patients with SLE. The review provides novel insights into strategies for managing SLE.
Collapse
Affiliation(s)
- Ryo Hisada
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan.
| | - Michihito Kono
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan.
| |
Collapse
|
10
|
Laniak OT, Winans T, Patel A, Park J, Perl A. Redox Pathogenesis in Rheumatic Diseases. ACR Open Rheumatol 2024; 6:334-346. [PMID: 38664977 PMCID: PMC11168917 DOI: 10.1002/acr2.11668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 06/14/2024] Open
Abstract
Despite being some of the most anecdotally well-known roads to pathogenesis, the mechanisms governing autoimmune rheumatic diseases are not yet fully understood. The overactivation of the cellular immune system and the characteristic development of autoantibodies have been linked to oxidative stress. Typical clinical manifestations, such as joint swelling and deformities and inflammation of the skin and internal organs, have also been connected directly or indirectly to redox mechanisms. The differences in generation and restraint of oxidative stress provide compelling evidence for the broad variety in pathology among rheumatic diseases and explain some of the common triggers and discordant manifestations in these diseases. Growing evidence of redox mechanisms in pathogenesis has provided a broad array of new potential therapeutic targets. Here, we explore the mechanisms by which oxidative stress is generated, explore its roles in autoimmunity and end-organ damage, and discuss how individual rheumatic diseases exhibit unique features that offer targets for therapeutic interventions.
Collapse
Affiliation(s)
- Olivia T. Laniak
- Norton College of MedicineState University of New York Upstate Medical UniversitySyracuse
| | - Thomas Winans
- Norton College of MedicineState University of New York Upstate Medical UniversitySyracuse
| | - Akshay Patel
- Norton College of MedicineState University of New York Upstate Medical UniversitySyracuse
| | - Joy Park
- Norton College of MedicineState University of New York Upstate Medical UniversitySyracuse
| | - Andras Perl
- Norton College of MedicineState University of New York Upstate Medical UniversitySyracuse
| |
Collapse
|
11
|
Morel L, Scindia Y. Functional consequence of Iron dyshomeostasis and ferroptosis in systemic lupus erythematosus and lupus nephritis. Clin Immunol 2024; 262:110181. [PMID: 38458303 DOI: 10.1016/j.clim.2024.110181] [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: 01/06/2024] [Accepted: 03/04/2024] [Indexed: 03/10/2024]
Abstract
Systemic lupus erythematosus (SLE) and its renal manifestation Lupus nephritis (LN) are characterized by a dysregulated immune system, autoantibodies, and injury to the renal parenchyma. Iron accumulation and ferroptosis in the immune effectors and renal tubules are recently identified pathological features in SLE and LN. Ferroptosis is an iron dependent non-apoptotic form of regulated cell death and ferroptosis inhibitors have improved disease outcomes in murine models of SLE, identifying it as a novel druggable target. In this review, we discuss novel mechanisms by which iron accumulation and ferroptosis perpetuate immune cell mediated pathology in SLE/LN. We highlight intra-renal dysregulation of iron metabolism and ferroptosis as an underlying pathogenic mechanism of renal tubular injury. The basic concepts of iron biology and ferroptosis are also discussed to expose the links between iron, cell metabolism and ferroptosis, that identify intracellular pro-ferroptotic enzymes and their protein conjugates as potential targets to improve SLE/LN outcomes.
Collapse
Affiliation(s)
- Laurence Morel
- Department of Microbiology, Immunology, and Molecular Genetics, UT Health San Antonio, San Antonio, TX, USA
| | - Yogesh Scindia
- Department of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
12
|
Mantle D, Hargreaves IP. Coenzyme Q10 and Autoimmune Disorders: An Overview. Int J Mol Sci 2024; 25:4576. [PMID: 38674161 PMCID: PMC11049925 DOI: 10.3390/ijms25084576] [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: 03/15/2024] [Revised: 04/13/2024] [Accepted: 04/20/2024] [Indexed: 04/28/2024] Open
Abstract
Some 90 autoimmune disorders have been described in medical literature, affecting most of the tissues within the body. Autoimmune disorders may be difficult to treat, and there is a need to develop novel therapeutic strategies for these disorders. Autoimmune disorders are characterised by mitochondrial dysfunction, oxidative stress, and inflammation; there is therefore a rationale for a role for coenzyme Q10 in the management of these disorders, on the basis of its key role in normal mitochondrial function, as an antioxidant, and as an anti-inflammatory agent. In this article, we have therefore reviewed the potential role of CoQ10, in terms of both deficiency and/or supplementation, in a range of autoimmune disorders.
Collapse
Affiliation(s)
| | - Iain P. Hargreaves
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool L3 3AF, UK
| |
Collapse
|
13
|
Furment MM, Perl A. Immmunometabolism of systemic lupus erythematosus. Clin Immunol 2024; 261:109939. [PMID: 38382658 DOI: 10.1016/j.clim.2024.109939] [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: 01/08/2024] [Revised: 01/26/2024] [Accepted: 02/09/2024] [Indexed: 02/23/2024]
Abstract
Systemic lupus erythematosus (SLE) is a potentially fatal chronic autoimmune disease which is underlain by complex dysfunction of the innate and adaptive immune systems. Although a series of well-defined genetic and environmental factors have been implicated in disease etiology, neither the development nor the persistence of SLE is well understood. Given that several disease susceptibility genes and environmental factors interact and influence inflammatory lineage specification through metabolism, the field of immunometabolism has become a forefront of cutting edge research. Along these lines, metabolic checkpoints of pathogenesis have been identified as targets of effective therapeutic interventions in mouse models and validated in clinical trials. Ongoing studies focus on mitochondrial oxidative stress, activation of the mechanistic target of rapamycin, calcium signaling, glucose utilization, tryptophan degradation, and metabolic cross-talk between gut microbiota and the host immune system.
Collapse
Affiliation(s)
- Marlene Marte Furment
- Departments of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York 13210, United States of America
| | - Andras Perl
- Departments of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York 13210, United States of America; Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York 13210, United States of America; Microbiology and Immunology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York 13210, United States of America.
| |
Collapse
|
14
|
Linder BA, Stute NL, Hutchison ZJ, Barnett AM, Tharpe MA, Kavazis AN, Kirkman DL, Gutierrez OM, Robinson AT. Acute high-dose MitoQ does not increase urinary kidney injury markers in healthy adults: a randomized crossover trial. Am J Physiol Renal Physiol 2024; 326:F135-F142. [PMID: 37942539 PMCID: PMC11198989 DOI: 10.1152/ajprenal.00186.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 10/23/2023] [Accepted: 11/06/2023] [Indexed: 11/10/2023] Open
Abstract
Several human studies have used the mitochondrial antioxidant MitoQ. Recent in vitro data indicating that MitoQ may induce nephrotoxicity caused concern regarding the safety of MitoQ on the kidneys, but the doses were supraphysiological. Therefore, we sought to determine whether acute MitoQ elicits changes in urinary biomarkers associated with tubular injury in healthy adults with our hypothesis being there would be no changes. Using a randomized crossover design, 32 healthy adults (16 females and 16 males, 29 ± 11 yr old) consumed MitoQ (100-160 mg based on body mass) or placebo capsules. We obtained serum samples and a 4- to 6-h postcapsule consumption urine sample. We assessed creatinine clearance and urine kidney injury biomarkers including the chitinase 3-like-1 gene product YKL-40, kidney-injury marker-1, monocyte chemoattractant protein-1, epidermal growth factor, neutrophil gelatinase-associated lipocalin, interleukin-18, and uromodulin using multiplex assays. We used t tests, Wilcoxon tests, and Hotelling's T2 to assess global differences in urinary kidney injury markers between conditions. Acute MitoQ supplementation did not influence urine flow rate (P = 0.086, rrb = 0.39), creatinine clearance (P = 0.085, rrb = 0.42), or urinary kidney injury markers (T22,8 = 30.6, P = 0.121, univariate ps > 0.064). Using exploratory univariate analysis, MitoQ did not alter individual injury markers compared with placebo (e.g., placebo vs. MitoQ: YKL-40, 507 ± 241 vs. 442 ± 236 pg/min, P = 0.241; kidney injury molecule-1, 84.1 ± 43.2 vs. 76.2 ± 51.2 pg/min, P = 0.890; and neutrophil gelatinase-associated lipocalin, 10.8 ± 10.1 vs. 9.83 ± 8.06 ng/min, P = 0.609). In conclusion, although longer-term surveillance and data are needed in clinical populations, our findings suggest that acute high-dose MitoQ had no effect on urinary kidney injury markers in healthy adults.NEW & NOTEWORTHY We found acute high-dose mitochondria-targeted antioxidant (MitoQ) supplementation was not nephrotoxic and had no effect on markers of acute kidney injury in healthy adults. These findings can help bolster further confidence in the safety of MitoQ, particularly for future investigations seeking to examine the role of mitochondrial oxidative stress, via acute MitoQ supplementation, on various physiological outcomes.
Collapse
Affiliation(s)
- Braxton A Linder
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Nina L Stute
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Zach J Hutchison
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Alex M Barnett
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - McKenna A Tharpe
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Andreas N Kavazis
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Danielle L Kirkman
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Orlando M Gutierrez
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Austin T Robinson
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| |
Collapse
|
15
|
Zhang Z, Jin L, Liu L, Zhou M, Zhang X, Zhang L. The intricate relationship between autoimmunity disease and neutrophils death patterns: a love-hate story. Apoptosis 2023; 28:1259-1284. [PMID: 37486407 DOI: 10.1007/s10495-023-01874-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2023] [Indexed: 07/25/2023]
Abstract
Autoimmune diseases are pathological conditions that result from the misidentification of self-antigens in immune system, leading to host tissue damage and destruction. These diseases can affect different organs and systems, including the blood, joints, skin, and muscles. Despite the significant progress made in comprehending the underlying pathogenesis, the complete mechanism of autoimmune disease is still not entirely understood. In autoimmune diseases, the innate immunocytes are not functioning properly: they are either abnormally activated or physically disabled. As a vital member of innate immunocyte, neutrophils and their modes of death are influenced by the microenvironment of different autoimmune diseases due to their short lifespan and diverse death modes. Related to neutrophil death pathways, apoptosis is the most frequent cell death form of neutrophil non-lytic morphology, delayed or aberrant apoptosis may contribute to the development anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitis (AAV). In addition, NETosis, necroptosis and pyroptosis which are parts of lytic morphology exacerbate disease progression through various mechanisms in autoimmune diseases. This review aims to summarize recent advancements in understanding neutrophil death modes in various autoimmune diseases and provide insights into the development of novel therapeutic approaches for autoimmune diseases.
Collapse
Affiliation(s)
- Ziwei Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China
- Anti-Inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui Province, China
| | - Lin Jin
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China
- Anti-Inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui Province, China
| | - Lianghu Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China
- Anti-Inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui Province, China
| | - Mengqi Zhou
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China
- Anti-Inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui Province, China
| | - Xianzheng Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China.
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China.
- Anti-Inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui Province, China.
| | - Lingling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China.
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China.
- Anti-Inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui Province, China.
| |
Collapse
|
16
|
Reshetnyak T, Nurbaeva K. The Role of Neutrophil Extracellular Traps (NETs) in the Pathogenesis of Systemic Lupus Erythematosus and Antiphospholipid Syndrome. Int J Mol Sci 2023; 24:13581. [PMID: 37686381 PMCID: PMC10487763 DOI: 10.3390/ijms241713581] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/10/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disease of unknown aetiology [...].
Collapse
Affiliation(s)
- Tatiana Reshetnyak
- Department of Thromboinflammation, V.A. Nasonova Research Institute of Rheumatology, 115522 Moscow, Russia;
| | | |
Collapse
|
17
|
Cheng G, Karoui H, Hardy M, Kalyanaraman B. Redox-crippled MitoQ potently inhibits breast cancer and glioma cell proliferation: A negative control for verifying the antioxidant mechanism of MitoQ in cancer and other oxidative pathologies. Free Radic Biol Med 2023; 205:175-187. [PMID: 37321281 PMCID: PMC11129726 DOI: 10.1016/j.freeradbiomed.2023.06.009] [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: 04/13/2023] [Revised: 06/01/2023] [Accepted: 06/13/2023] [Indexed: 06/17/2023]
Abstract
Mitochondria-targeted coenzyme Q10 (Mito-ubiquinone, Mito-quinone mesylate, or MitoQ) was shown to be an effective antimetastatic drug in patients with triple-negative breast cancer. MitoQ, sold as a nutritional supplement, prevents breast cancer recurrence. It potently inhibited tumor growth and tumor cell proliferation in preclinical xenograft models and in vitro breast cancer cells. The proposed mechanism of action involves the inhibition of reactive oxygen species by MitoQ via a redox-cycling mechanism between the oxidized form, MitoQ, and the fully reduced form, MitoQH2 (also called Mito-ubiquinol). To fully corroborate this antioxidant mechanism, we substituted the hydroquinone group (-OH) with the methoxy group (-OCH3). Unlike MitoQ, the modified form, dimethoxy MitoQ (DM-MitoQ), lacks redox-cycling between the quinone and hydroquinone forms. DM-MitoQ was not converted to MitoQ in MDA-MB-231 cells. We tested the antiproliferative effects of both MitoQ and DM-MitoQ in human breast cancer (MDA-MB-231), brain-homing cancer (MDA-MB-231BR), and glioma (U87MG) cells. Surprisingly, DM-MitoQ was slightly more potent than MitoQ (IC50 = 0.26 μM versus 0.38 μM) at inhibiting proliferation of these cells. Both MitoQ and DM-MitoQ potently inhibited mitochondrial complex I-dependent oxygen consumption (IC50 = 0.52 μM and 0.17 μM, respectively). This study also suggests that DM-MitoQ, which is a more hydrophobic analog of MitoQ (logP: 10.1 and 8.7) devoid of antioxidant function and reactive oxygen species scavenging ability, can inhibit cancer cell proliferation. We conclude that inhibition of mitochondrial oxidative phosphorylation by MitoQ is responsible for inhibition of breast cancer and glioma proliferation and metastasis. Blunting the antioxidant effect using the redox-crippled DM-MitoQ can serve as a useful negative control in corroborating the involvement of free radical-mediated processes (e.g., ferroptosis, protein oxidation/nitration) using MitoQ in other oxidative pathologies.
Collapse
Affiliation(s)
- Gang Cheng
- Department of Biophysics, 8701 Watertown Plank Road, Milwaukee, WI, 53226, United States
| | - Hakim Karoui
- Aix Marseille Univ, CNRS, ICR, UMR, 7273, Marseille, 13013, France
| | - Micael Hardy
- Aix Marseille Univ, CNRS, ICR, UMR, 7273, Marseille, 13013, France
| | - Balaraman Kalyanaraman
- Department of Biophysics, 8701 Watertown Plank Road, Milwaukee, WI, 53226, United States.
| |
Collapse
|
18
|
Lock MC, Botting KJ, Allison BJ, Niu Y, Ford SG, Murphy MP, Orgeig S, Giussani DA, Morrison JL. MitoQ as an antenatal antioxidant treatment improves markers of lung maturation in healthy and hypoxic pregnancy. J Physiol 2023; 601:3647-3665. [PMID: 37467062 PMCID: PMC10952154 DOI: 10.1113/jp284786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/04/2023] [Indexed: 07/21/2023] Open
Abstract
Chronic fetal hypoxaemia is a common pregnancy complication that increases the risk of infants experiencing respiratory complications at birth. In turn, chronic fetal hypoxaemia promotes oxidative stress, and maternal antioxidant therapy in animal models of hypoxic pregnancy has proven to be protective with regards to fetal growth and cardiovascular development. However, whether antenatal antioxidant therapy confers any benefit on lung development in complicated pregnancies has not yet been investigated. Here, we tested the hypothesis that maternal antenatal treatment with MitoQ will protect the developing lung in hypoxic pregnancy in sheep, a species with similar fetal lung developmental milestones as humans. Maternal treatment with MitoQ during late gestation promoted fetal pulmonary surfactant maturation and an increase in the expression of lung mitochondrial complexes III and V independent of oxygenation. Maternal treatment with MitoQ in hypoxic pregnancy also increased the expression of genes regulating liquid reabsorption in the fetal lung. These data support the hypothesis tested and suggest that MitoQ as an antenatal targeted antioxidant treatment may improve lung maturation in the late gestation fetus. KEY POINTS: Chronic fetal hypoxaemia promotes oxidative stress, and maternal antioxidant therapy in hypoxic pregnancy has proven to be protective with regards to fetal growth and cardiovascular development. MitoQ is a targeted antioxidant that uses the cell and the mitochondrial membrane potential to accumulate within the mitochondria. Treatment of healthy or hypoxic pregnancy with MitoQ, increases the expression of key molecules involved in surfactant maturation, lung liquid reabsorption and in mitochondrial proteins driving ATP synthesis in the fetal sheep lung. There were no detrimental effects of MitoQ treatment alone on the molecular components measured in the present study, suggesting that maternal antioxidant treatment has no effect on other components of normal maturation of the surfactant system.
Collapse
Affiliation(s)
- Mitchell C. Lock
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health ScienceUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Kimberley J. Botting
- Department of Physiology, Development & NeuroscienceUniversity of CambridgeCambridgeUK
| | - Beth J. Allison
- Department of Physiology, Development & NeuroscienceUniversity of CambridgeCambridgeUK
| | - Youguo Niu
- Department of Physiology, Development & NeuroscienceUniversity of CambridgeCambridgeUK
| | - Sage G. Ford
- Department of Physiology, Development & NeuroscienceUniversity of CambridgeCambridgeUK
| | | | - Sandra Orgeig
- UniSA: Clinical and Health ScienceUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Dino A. Giussani
- Department of Physiology, Development & NeuroscienceUniversity of CambridgeCambridgeUK
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health ScienceUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| |
Collapse
|
19
|
Mohan C, Zhang T, Putterman C. Pathogenic cellular and molecular mediators in lupus nephritis. Nat Rev Nephrol 2023:10.1038/s41581-023-00722-z. [PMID: 37225921 DOI: 10.1038/s41581-023-00722-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2023] [Indexed: 05/26/2023]
Abstract
Kidney involvement in patients with systemic lupus erythematosus - lupus nephritis (LN) - is one of the most important and common clinical manifestations of this disease and occurs in 40-60% of patients. Current treatment regimens achieve a complete kidney response in only a minority of affected individuals, and 10-15% of patients with LN develop kidney failure, with its attendant morbidity and considerable prognostic implications. Moreover, the medications most often used to treat LN - corticosteroids in combination with immunosuppressive or cytotoxic drugs - are associated with substantial side effects. Advances in proteomics, flow cytometry and RNA sequencing have led to important new insights into immune cells, molecules and mechanistic pathways that are instrumental in the pathogenesis of LN. These insights, together with a renewed focus on the study of human LN kidney tissue, suggest new therapeutic targets that are already being tested in lupus animal models and early-phase clinical trials and, as such, are hoped to eventually lead to meaningful improvements in the care of patients with systemic lupus erythematosus-associated kidney disease.
Collapse
Affiliation(s)
- Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA.
| | - Ting Zhang
- Division of Rheumatology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chaim Putterman
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.
- Division of Rheumatology and Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
20
|
Jiao Y, Yan Z, Yang A. Mitochondria in innate immunity signaling and its therapeutic implications in autoimmune diseases. Front Immunol 2023; 14:1160035. [PMID: 37122709 PMCID: PMC10130412 DOI: 10.3389/fimmu.2023.1160035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/28/2023] [Indexed: 05/02/2023] Open
Abstract
Autoimmune diseases are characterized by vast alterations in immune responses, but the pathogenesis remains sophisticated and yet to be fully elucidated. Multiple mechanisms regulating cell differentiation, maturation, and death are critical, among which mitochondria-related cellular organelle functions have recently gained accumulating attention. Mitochondria, as a highly preserved organelle in eukaryotes, have crucial roles in the cellular response to both exogenous and endogenous stress beyond their fundamental functions in chemical energy conversion. In this review, we aim to summarize recent findings on the function of mitochondria in the innate immune response and its aberrancy in autoimmune diseases such as rheumatoid arthritis, systemic lupus erythematosus, etc., mainly focusing on its direct impact on cellular metabolism and its machinery on regulating immune response signaling pathways. More importantly, we summarize the status quo of potential therapeutic targets found in the mitochondrial regulation in the setting of autoimmune diseases and wish to shed light on future studies.
Collapse
Affiliation(s)
- Yuhao Jiao
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhiyu Yan
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- 4+4 Medical Doctor Program, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Aiming Yang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
21
|
Zhang X, Meng J, Shi X, Quinet RJ, Davis W, Zakem J, Keshavamurthy C, Patel R, Lobo G, Hellmers L, Ray AN, Rivers LE, Ali H, Posas-Mendoza T, Hille C, You Z. Lupus pathogenesis and autoimmunity are exacerbated by high fat diet-induced obesity in MRL/lpr mice. Lupus Sci Med 2023; 10:10/1/e000898. [PMID: 37041033 PMCID: PMC10106072 DOI: 10.1136/lupus-2023-000898] [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: 01/11/2023] [Accepted: 03/18/2023] [Indexed: 04/13/2023]
Abstract
OBJECTIVE SLE is an autoimmune disease characterised by persistent inflammation and autoantibody production. Genetic predisposition and environmental factors such as a high-fat diet (HFD) may contribute to lupus development. However, the immune cell profile and gender difference in response to HFD in lupus have not been reported. Here we investigated the impact of HFD on lupus pathogenesis and autoimmunity using lupus-prone mice. METHODS Thirty male and 30 female MRL/lymphoproliferation (lpr) mice were fed with regular diet (RD) or HFD. Body weights were recorded weekly. SLE progression was monitored by skin lesion, urine protein, titres of antidouble-strand DNA (dsDNA) and ANA. At week 14, kidney and skin tissue sections were stained with H&E and periodic acid-Schiff to detect histological kidney index and skin score. Splenocytes were identified by immunofluorescence staining and flow cytometry. RESULTS HFD significantly increased body weight and lipid levels compared with RD (p<0.01). Skin lesions were observed in 55.6% of the HFD group compared with 11.1% of the RD group, with greater histopathological skin scores in the female HFD group (p<0.01). Although both male and female mice had higher serum IgG in the HFD group than in the RD group, only the male HFD group showed an increased trend in anti-dsDNA Ab and ANA titres. Kidney pathological changes in the HFD group were more severe in male mice than in female mice (p<0.05), detected by proteinuria, kidney index and glomerular cell proliferation. Significant increases of germinal centre B cells and T follicular helper cells were observed in the spleens of HFD mice (p<0.05). CONCLUSION HFD induced an accelerated and exacerbated lupus development and autoimmunity in MRL/lpr mice. Our results parallel many known clinical lupus phenotypes and sexual dimorphism in which male patients are likelier to have a severe disease (nephritis) than female lupus patients who may have a broader range of lupus symptoms.
Collapse
Affiliation(s)
- Xin Zhang
- Institute of Translational Research, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
| | - Juan Meng
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Rheumatology and Immunology, Beijing Chao-Yang Hospital Capital Medical University, Beijing, China
| | - Xuhua Shi
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Rheumatology and Immunology, Beijing Chao-Yang Hospital Capital Medical University, Beijing, China
| | - Robert James Quinet
- Department of Rheumatology, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
| | - William Davis
- Department of Rheumatology, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
- Department of Rheumatology, The University of Queensland Medical School, Ochsner Clinical School, New Orleans, Louisiana, USA
| | - Jerald Zakem
- Department of Rheumatology, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
| | - Chandana Keshavamurthy
- Department of Rheumatology, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
- Department of Rheumatology, The University of Queensland Medical School, Ochsner Clinical School, New Orleans, Louisiana, USA
| | - Ronak Patel
- Department of Rheumatology, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
| | - Gitanjali Lobo
- Department of Rheumatology, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
| | - Linh Hellmers
- Institute of Translational Research, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
| | - Alicia Nicole Ray
- Institute of Translational Research, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
| | - Laura E Rivers
- Department of Rheumatology, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
| | - Hiba Ali
- Department of Rheumatology, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
| | | | - Chad Hille
- Department of Rheumatology, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
| | - Zongbing You
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Research Service, Southeast Louisiana Veterans Health Care System, New Orleans, Louisiana, USA
| |
Collapse
|
22
|
Blanco LP, Kaplan MJ. Metabolic alterations of the immune system in the pathogenesis of autoimmune diseases. PLoS Biol 2023; 21:e3002084. [PMID: 37098006 PMCID: PMC10128981 DOI: 10.1371/journal.pbio.3002084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023] Open
Abstract
Systemic autoimmune diseases are characteristically associated with aberrant autoreactive innate and adaptive immune responses that lead to tissue damage and increased morbidity and mortality. Autoimmunity has been linked to alterations in the metabolic functions of immune cells (immunometabolism) and, more specifically, to mitochondrial dysfunction. Much has been written about immunometabolism in autoimmunity in general, so this Essay focuses on recent research into the role of mitochondrial dysfunction in the dysregulation of innate and adaptive immunity that is characteristic of systemic autoimmune diseases such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Enhancing the understanding of mitochondrial dysregulation in autoimmunity will hopefully contribute to accelerating the development of immunomodulatory treatments for these challenging diseases.
Collapse
Affiliation(s)
- Luz P Blanco
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
23
|
Daamen AR, Wang H, Bachali P, Shen N, Kingsmore KM, Robl RD, Grammer AC, Fu SM, Lipsky PE. Molecular mechanisms governing the progression of nephritis in lupus prone mice and human lupus patients. Front Immunol 2023; 14:1147526. [PMID: 36936908 PMCID: PMC10016352 DOI: 10.3389/fimmu.2023.1147526] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 02/15/2023] [Indexed: 03/04/2023] Open
Abstract
Introduction Pathologic inflammation is a major driver of kidney damage in lupus nephritis (LN), but the immune mechanisms of disease progression and risk factors for end organ damage are poorly understood. Methods To characterize molecular profiles through the development of LN, we carried out gene expression analysis of microdissected kidneys from lupus-prone NZM2328 mice. We examined male mice and the congenic NZM2328.R27 strain as a means to define mechanisms associated with resistance to chronic nephritis. Gene expression profiles in lupus mice were compared with those in human LN. Results NZM2328 mice exhibited progress from acute to transitional and then to chronic glomerulonephritis (GN). Each stage manifested a unique molecular profile. Neither male mice nor R27 mice progressed past the acute GN stage, with the former exhibiting minimal immune infiltration and the latter enrichment of immunoregulatory gene signatures in conjunction with robust kidney tubule cell profiles indicative of resistance to cellular damage. The gene expression profiles of human LN were similar to those noted in the NZM2328 mouse suggesting comparable stages of LN progression. Conclusions Overall, this work provides a comprehensive examination of the immune processes involved in progression of murine LN and thus contributes to our understanding of the risk factors for end-stage renal disease. In addition, this work presents a foundation for improved classification of LN and illustrates the applicability of murine models to identify the stages of human disease.
Collapse
Affiliation(s)
| | - Hongyang Wang
- Center for Immunity, Inflammation, and Regenerative Medicine, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
- Division of Rheumatology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| | | | - Nan Shen
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Robert D. Robl
- AMPEL BioSolutions LLC, Charlottesville, VA, United States
| | | | - Shu Man Fu
- Center for Immunity, Inflammation, and Regenerative Medicine, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
- Division of Rheumatology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| | | |
Collapse
|
24
|
Yan Z, Chen Q, Xia Y. Oxidative Stress Contributes to Inflammatory and Cellular Damage in Systemic Lupus Erythematosus: Cellular Markers and Molecular Mechanism. J Inflamm Res 2023; 16:453-465. [PMID: 36761905 PMCID: PMC9907008 DOI: 10.2147/jir.s399284] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a multifactorial autoimmune disease with complex pathogenesis, the treatment of which relies exclusively on the use of immunosuppressants. Increased oxidative stress is involved in causing inflammatory and cellular defects in the pathogenesis of SLE. Various inflammatory and cellular markers including oxidative modifications of proteins, lipids, and DNA contribute to immune system dysregulation and trigger an aggressive autoimmune attack through molecular mechanisms like enhanced NETosis, mTOR pathway activation, and imbalanced T-cell differentiation. Accordingly, the detection of inflammatory and cellular markers is important for providing an accurate assessment of the extent of oxidative stress. Oxidative stress also reduces DNA methylation, thus allowing the increased expression of affected genes. As a result, pharmacological approaches targeting oxidative stress yield promising results in treating patients with SLE. The purpose of this review is to examine the involvement of oxidative stress in the pathogenesis and management of SLE.
Collapse
Affiliation(s)
- Zhu Yan
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
| | - Qin Chen
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China,Correspondence: Yumin Xia, Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, 157 Xiwu Road, Xi’an, 710004, People’s Republic of China, Tel/Fax +86-29-87679969, Email
| |
Collapse
|
25
|
Wincup C, Fasano S. 'What is fuelling the immune response in systemic lupus erythematosus?' Evaluating the key metabolites driving plasmablast differentiation. Rheumatology (Oxford) 2023; 62:492-494. [PMID: 35861392 PMCID: PMC9891419 DOI: 10.1093/rheumatology/keac418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 07/13/2022] [Indexed: 02/04/2023] Open
Affiliation(s)
- Chris Wincup
- Department of Rheumatology, Division of Medicine, Rayne Building, University College London.,Department of Rheumatology, King's College Hospital, London, UK
| | - Serena Fasano
- Rheumatology Unit, Department of Precision Medicine, University of Campania, 'Luigi Vanvitelli', Naples, Italy
| |
Collapse
|
26
|
Coenzyme Q10: Role in Less Common Age-Related Disorders. Antioxidants (Basel) 2022; 11:antiox11112293. [DOI: 10.3390/antiox11112293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022] Open
Abstract
In this article we have reviewed the potential role of coenzyme Q10 (CoQ10) in the pathogenesis and treatment of a number of less common age-related disorders, for many of which effective therapies are not currently available. For most of these disorders, mitochondrial dysfunction, oxidative stress and inflammation have been implicated in the disease process, providing a rationale for the potential therapeutic use of CoQ10, because of its key roles in mitochondrial function, as an antioxidant, and as an anti-inflammatory agent. Disorders reviewed in the article include multi system atrophy, progressive supranuclear palsy, sporadic adult onset ataxia, and pulmonary fibrosis, together with late onset versions of Huntington’s disease, Alexander disease, lupus, anti-phospholipid syndrome, lysosomal storage disorders, fibromyalgia, Machado-Joseph disease, acyl-CoA dehydrogenase deficiency, and Leber’s optic neuropathy.
Collapse
|
27
|
Wigerblad G, Kaplan MJ. Neutrophil extracellular traps in systemic autoimmune and autoinflammatory diseases. Nat Rev Immunol 2022; 23:274-288. [PMID: 36257987 PMCID: PMC9579530 DOI: 10.1038/s41577-022-00787-0] [Citation(s) in RCA: 121] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2022] [Indexed: 11/09/2022]
Abstract
Systemic autoimmune diseases are characterized by the failure of the immune system to differentiate self from non-self. These conditions are associated with significant morbidity and mortality, and they can affect many organs and systems, having significant clinical heterogeneity. Recent discoveries have highlighted that neutrophils, and in particular the neutrophil extracellular traps that they can release upon activation, can have central roles in the initiation and perpetuation of systemic autoimmune disorders and orchestrate complex inflammatory responses that lead to organ damage. Dysregulation of neutrophil cell death can lead to the modification of autoantigens and their presentation to the adaptive immune system. Furthermore, subsets of neutrophils that seem to be more prevalent in patients with systemic autoimmune disorders can promote vascular damage and increased oxidative stress. With the emergence of new technologies allowing for improved assessments of neutrophils, the complexity of neutrophil biology and its dysregulation is now starting to be understood. In this Review, we provide an overview of the roles of neutrophils in systemic autoimmune and autoinflammatory diseases and address putative therapeutic targets that may be explored based on this new knowledge.
Collapse
|
28
|
Ma L, Roach T, Morel L. Immunometabolic alterations in lupus: where do they come from and where do we go from there? Curr Opin Immunol 2022; 78:102245. [PMID: 36122544 PMCID: PMC10161929 DOI: 10.1016/j.coi.2022.102245] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 08/23/2022] [Indexed: 01/28/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease in which the overactivation of the immune system has been associated with metabolic alterations. Targeting the altered immunometabolism has been proposed to treat SLE patients based on their results obtained and mouse models of the disease. Here, we review the recent literature to discuss the possible origins of the alterations in the metabolism of immune cells in lupus, the dominant role of mitochondrial defects, technological advances that may move the field forward, as well as how targeting lupus immunometabolism may have therapeutic potential.
Collapse
Affiliation(s)
- Longhuan Ma
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, 7703 Floyd Curl Dr., MC7758, San Antonio, TX, 78229-3900, USA
| | - Tracoyia Roach
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, 7703 Floyd Curl Dr., MC7758, San Antonio, TX, 78229-3900, USA
| | - Laurence Morel
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, 7703 Floyd Curl Dr., MC7758, San Antonio, TX, 78229-3900, USA.
| |
Collapse
|
29
|
Rizvi ZA, Babele P, Sadhu S, Madan U, Tripathy MR, Goswami S, Mani S, Kumar S, Awasthi A, Dikshit M. Prophylactic treatment of Glycyrrhiza glabra mitigates COVID-19 pathology through inhibition of pro-inflammatory cytokines in the hamster model and NETosis. Front Immunol 2022; 13:945583. [PMID: 36238303 PMCID: PMC9550929 DOI: 10.3389/fimmu.2022.945583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 09/05/2022] [Indexed: 01/08/2023] Open
Abstract
Severe coronavirus disease (COVID-19) is accompanied by acute respiratory distress syndrome and pulmonary pathology, and is presented mostly with an inflammatory cytokine release, a dysregulated immune response, a skewed neutrophil/lymphocyte ratio, and a hypercoagulable state. Though vaccinations have proved effective in reducing the COVID-19-related mortality, the limitation of the use of vaccine against immunocompromised individuals, those with comorbidity, and emerging variants remains a concern. In the current study, we investigate for the first time the efficacy of the Glycyrrhiza glabra (GG) extract, a potent immunomodulator, against SARS-CoV-2 infection in hamsters. Prophylactic treatment with GG showed protection against loss in body weight and a 35%–40% decrease in lung viral load along with reduced lung pathology in the hamster model. Remarkably, GG reduced the mRNA expression of pro-inflammatory cytokines and plasminogen activator inhibitor-1 (PAI-1). In vitro, GG acted as a potent immunomodulator by reducing Th2 and Th17 differentiation and IL-4 and IL-17A cytokine production. In addition, GG also showed robust potential to suppress ROS, mtROS, and NET generation in a concentration-dependent manner in both human polymorphonuclear neutrophils (PMNs) and murine bone marrow-derived neutrophils (BMDNs). Taken together, we provide evidence for the protective efficacy of GG against COVID-19 and its putative mechanistic insight through its immunomodulatory properties. Our study provides the proof of concept for GG efficacy against SARS-CoV-2 using a hamster model and opens the path for further studies aimed at identifying the active ingredients of GG and its efficacy in COVID-19 clinical cases.
Collapse
Affiliation(s)
- Zaigham Abbas Rizvi
- Immuno-Biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Prabhakar Babele
- Noncommunicable Disease Centre, Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
| | - Srikanth Sadhu
- Immuno-Biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Upasna Madan
- Immuno-Biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Manas Ranjan Tripathy
- Immuno-Biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Sandeep Goswami
- Immuno-Biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Shailendra Mani
- Noncommunicable Disease Centre, Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
| | - Sachin Kumar
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Amit Awasthi
- Immuno-Biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, Faridabad, Haryana, India
- *Correspondence: Madhu Dikshit, ; ; Amit Awasthi,
| | - Madhu Dikshit
- Noncommunicable Disease Centre, Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- *Correspondence: Madhu Dikshit, ; ; Amit Awasthi,
| |
Collapse
|
30
|
Quintero-González DC, Muñoz-Urbano M, Vásquez G. Mitochondria as a key player in systemic lupus erythematosus. Autoimmunity 2022; 55:497-505. [PMID: 35978536 DOI: 10.1080/08916934.2022.2112181] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous, multisystemic autoimmune disease with a broad clinical spectrum. Loss of self-tolerance and chronic inflammation are critical markers of SLE pathogenesis. Although alterations in adaptive immunity are widely recognized, increasing reports indicate the role of mitochondrial dysfunction in activating pathogenic pathways involving the innate immune system. Among these, disarrangements in mitochondrial DNA copy number and heteroplasmy percentage are related to SLE activity. Furthermore, increased oxidative stress contributes to post-translational changes in different molecules (proteins, nucleic acids, and lipids), release of oxidized mitochondrial DNA through a pore of voltage-dependent anion channel oligomers, and spontaneous mitochondrial antiviral signaling protein oligomerization. Finally, a reduction in mitophagy, apoptosis induction, and NETosis has been reported in SLE. Most of these pathways lead to persistent and inappropriate exposure to oxidized mitochondrial DNA, which can stimulate plasmacytoid dendritic cells, enhance autoreactive lymphocyte activation, and release increased amounts of interferons through stimulation of toll-like receptors and cytosolic DNA sensors. Likewise, abnormal T-cell receptor activation, decreased regulatory T cells, enhanced Th17 phenotypes, and increased monocyte maturation to dendritic cells have also been observed in SLE. Targeting the players involved in mitochondrial damage can ultimately help.
Collapse
Affiliation(s)
| | - Marcela Muñoz-Urbano
- Rheumatology Section, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - G Vásquez
- Rheumatology Section, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia.,Grupo de Inmunología Celular e Inmunogenética (GICIC), Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
31
|
Xu Y, Li P, Li K, Li N, Liu H, Zhang X, Liu W, Liu Y. Pathological mechanisms and crosstalk among different forms of cell death in systemic lupus erythematosus. J Autoimmun 2022; 132:102890. [PMID: 35963809 DOI: 10.1016/j.jaut.2022.102890] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 07/29/2022] [Indexed: 10/15/2022]
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disorder characterized by a profound immune dysregulation and the presence of a variety of autoantibodies. Aberrant activation of programmed cell death (PCD) signaling and accelerated cell death is critical in the immunopathogenesis of SLE. Accumulating cellular components from the dead cells and ineffective clearance of the dead cell debris, in particular the nucleic acids and nucleic acids-protein complexes, provide a stable source of self-antigens, which potently activate auto-reactive B cells and promote IFN-I responses in SLE. Different cell types display distinct susceptibility and characteristics to a certain type of cell death, while different PCDs in various cells have mutual and intricate connections to promote immune dysregulation and contribute to the development of SLE. In this review, we discuss the role of various cell death pathways and their interactions in the pathogenesis of SLE. An in depth understanding of the interconnections among various forms cell death in SLE will lead to a better understanding of disease pathogenesis, shedding light on the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Yue Xu
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Pengchong Li
- Department of Gastroenterology, Beijing Friendship Hospital, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Diseases, Capital Medical University, Beijing, China
| | - Ketian Li
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Nannan Li
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Huazhen Liu
- Peking Union Medical College Hospital, Beijing, China
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Liu
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.
| | - Yudong Liu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
32
|
Tubular Mitochondrial Dysfunction, Oxidative Stress, and Progression of Chronic Kidney Disease. Antioxidants (Basel) 2022; 11:antiox11071356. [PMID: 35883847 PMCID: PMC9311633 DOI: 10.3390/antiox11071356] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 12/23/2022] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are interconnected conditions, and CKD is projected to become the fifth leading global cause of death by 2040. New therapeutic approaches are needed. Mitochondrial dysfunction and oxidative stress have emerged as drivers of kidney injury in acute and chronic settings, promoting the AKI-to-CKD transition. In this work, we review the role of mitochondrial dysfunction and oxidative stress in AKI and CKD progression and discuss novel therapeutic approaches. Specifically, evidence for mitochondrial dysfunction in diverse models of AKI (nephrotoxicity, cytokine storm, and ischemia-reperfusion injury) and CKD (diabetic kidney disease, glomerulopathies) is discussed; the clinical implications of novel information on the key role of mitochondria-related transcriptional regulators peroxisome proliferator-activated receptor gamma coactivator 1-alpha, transcription factor EB (PGC-1α, TFEB), and carnitine palmitoyl-transferase 1A (CPT1A) in kidney disease are addressed; the current status of the clinical development of therapeutic approaches targeting mitochondria are updated; and barriers to the clinical development of mitochondria-targeted interventions are discussed, including the lack of clinical diagnostic tests that allow us to categorize the baseline renal mitochondrial dysfunction/mitochondrial oxidative stress and to monitor its response to therapeutic intervention. Finally, key milestones for further research are proposed.
Collapse
|
33
|
Davuluri S, Duvvuri B, Lood C, Faghihi-Kashani S, Chung L. Calcinosis in dermatomyositis: Origins and possible therapeutic avenues. Best Pract Res Clin Rheumatol 2022; 36:101768. [PMID: 35803868 DOI: 10.1016/j.berh.2022.101768] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Calcinosis, insoluble calcium compounds deposited in skin and other tissues, is a crippling sequela of dermatomyositis. Prolonged disease associated with ongoing inflammation, ischemia, repetitive trauma, and certain autoantibodies are associated with calcinosis. Herein, we describe potential pathogenic mechanisms including the role of mitochondrial calcification. There are no widely effective treatments for calcinosis. We review available pharmacologic therapies for calcinosis including those targeting calcium and phosphorus metabolism; immunosuppressive/anti-inflammatory therapies; and vasodilators. Mounting evidence supports the use of various formulations of sodium thiosulfate in the treatment of calcinosis. Although the early institution of aggressive immunosuppression may prevent calcinosis in juvenile dermatomyositis, only limited data support improvement once it has developed. Minocycline can be useful particularly for lesions associated with surrounding inflammation. Powerful vasodilators, such as prostacyclin analogs, may have promise in the treatment of calcinosis, but further studies are necessary. Surgical removal of lesions when amenable is our treatment of choice.
Collapse
Affiliation(s)
- Srijana Davuluri
- Stanford School of Medicine, Division of Immunology &Rheumatology, 1000 Welch Road, Suite 204, Palo Alto, 94304, California, USA.
| | - Bhargavi Duvvuri
- University of Washington, Department of Medicine, Division of Rheumatology, 750 Republican Street, Seattle, WA, 98109, USA.
| | - Christian Lood
- University of Washington, Division of Rheumatology, 750 Republican Street, Room E-545, Seattle, WA, 98109, USA.
| | - Sara Faghihi-Kashani
- Stanford School of Medicine, Division of Immunology &Rheumatology, 1000 Welch Road, Suite 204, Palo Alto, 94304, California, USA.
| | - Lorinda Chung
- Stanford School of Medicine & Palo Alto VA Health Care System, Division of Immunology &Rheumatology, 1000 Welch Road, Suite 203, Palo Alto, 94304, California, USA.
| |
Collapse
|
34
|
Miglioranza Scavuzzi B, Holoshitz J. Endoplasmic Reticulum Stress, Oxidative Stress, and Rheumatic Diseases. Antioxidants (Basel) 2022; 11:1306. [PMID: 35883795 PMCID: PMC9312221 DOI: 10.3390/antiox11071306] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 06/27/2022] [Accepted: 06/27/2022] [Indexed: 12/10/2022] Open
Abstract
BACKGROUND The endoplasmic reticulum (ER) is a multi-functional organelle responsible for cellular homeostasis, protein synthesis, folding and secretion. It has been increasingly recognized that the loss of ER homeostasis plays a central role in the development of autoimmune inflammatory disorders, such as rheumatic diseases. Purpose/Main contents: Here, we review current knowledge of the contribution of ER stress to the pathogenesis of rheumatic diseases, with a focus on rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). We also review the interplay between protein folding and formation of reactive oxygen species (ROS), where ER stress induces oxidative stress (OS), which further aggravates the accumulation of misfolded proteins and oxidation, in a vicious cycle. Intervention studies targeting ER stress and oxidative stress in the context of rheumatic diseases are also reviewed. CONCLUSIONS Loss of ER homeostasis is a significant factor in the pathogeneses of RA and SLE. Targeting ER stress, unfolded protein response (UPR) pathways and oxidative stress in these diseases both in vitro and in animal models have shown promising results and deserve further investigation.
Collapse
Affiliation(s)
| | - Joseph Holoshitz
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA;
| |
Collapse
|
35
|
Wang M, Ishikawa T, Lai Y, Nallapothula D, Singh RR. Diverse Roles of NETosis in the Pathogenesis of Lupus. Front Immunol 2022; 13:895216. [PMID: 35686129 PMCID: PMC9170953 DOI: 10.3389/fimmu.2022.895216] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
NETosis is a form of neutrophil cell death during which extracellular fibrillary structures composed of cytosolic and granule proteins assembled on scaffolds of decondensed chromatin, called neutrophil extracellular traps (NETs), are released. NETs normally contribute to host immune defense. Accumulating evidence implicates aberrant NET production and/or reduced NET clearance, along with alterations of molecules involved in NETosis pathway, in humans and animals with lupus. The extruded nuclear antigens released by NET are a source of autoantigens, which can contribute to the breakdown of self-tolerance in lupus. Excessive NET can also promote the production of pro-inflammatory cytokine interferon-α, elicit direct cytotoxic effect on various renal cells, and cause capillary necrosis and podocyte loss. Additionally, NET can induce endothelial-to-mesenchymal transdifferentiation, which can promote activated myofibroblasts leading to extracellular matrix production. Thus, aberrant NETosis can play diverse roles, including autoantibody production, inflammation, and tissue damage, at different stages of lupus pathogenesis. Evidence suggests that treatments currently used in lupus may reduce NETosis, suggesting a potential utility of targeting NETosis to treat lupus. In fact, several approaches are being experimented to therapeutically target pathways of NETosis. Future studies should precisely delineate distinct roles of NETosis at different stages of lupus pathogenesis in humans, which would offer a rational basis for NETosis-targeting treatments in the clinic.
Collapse
Affiliation(s)
- Meiying Wang
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA, United States.,Department of Rheumatology and Immunology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Tatsuya Ishikawa
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Yupeng Lai
- Department of Rheumatology and Immunology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Dhiraj Nallapothula
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Ram Raj Singh
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA, United States.,Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States.,Molecular Toxicology Interdepartmental Program, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States.,Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| |
Collapse
|
36
|
Kaseda S, Sannomiya Y, Horizono J, Kuwazuru J, Suico MA, Ogi S, Sasaki R, Sunamoto H, Fukiya H, Nishiyama H, Kamura M, Niinou S, Koyama Y, Nara F, Shuto T, Onuma K, Kai H. Novel Keap1-Nrf2 Protein-Protein Interaction Inhibitor UBE-1099 Ameliorates Progressive Phenotype in Alport Syndrome Mouse Model. KIDNEY360 2022; 3:687-699. [PMID: 35721612 PMCID: PMC9136903 DOI: 10.34067/kid.0004572021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/29/2021] [Indexed: 06/15/2023]
Abstract
BACKGROUND Bardoxolone methyl activates nuclear factor erythroid 2-related factor 2 (Nrf2) via covalent binding and irreversible inhibition of Kelch-like ECH-associated protein 1 (Keap1), the negative regulator of Nrf2. Ongoing clinical trials of bardoxolone methyl show promising effects for patients with CKD. However, the direct inhibition of Keap1-Nrf2 protein-protein interaction (PPI) as an approach to activate Nrf2 is less explored. METHODS We developed a noncovalent Nrf2 activator UBE-1099, which highly selectively inhibits Keap1-Nrf2 PPI, and evaluated its efficacy on the progressive phenotype in an Alport syndrome mouse model (Col4a5-G5X). RESULTS Similar to bardoxolone methyl, UBE-1099 transiently increased proteinuria and reduced plasma creatinine in Alport mice. Importantly, UBE-1099 improved the glomerulosclerosis, renal inflammation, and fibrosis, and prolonged the life span of Alport mice. UBE-1099 ameliorated the dysfunction of Nrf2 signaling in the renal tissue of Alport mice. Moreover, transcriptome analysis in the glomerulus showed that UBE-1099 induced the expression of genes associated with the cell cycle and cytoskeleton, which may explain its unique mechanism of improvement such as glomerular morphologic change. CONCLUSIONS UBE-1099 significantly ameliorates the progressive phenotype in Alport mice. Our results revealed the efficacy of Keap1-Nrf2 PPI inhibitor for glomerulosclerosis and present a potential therapeutic drug for CKD.
Collapse
Affiliation(s)
- Shota Kaseda
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
- Program for Leading Graduate School “HIGO (Health Life Science: Interdisciplinary and Glocal Oriented) Program,” Kumamoto University, Kumamoto, Japan
| | - Yuya Sannomiya
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Jun Horizono
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Jun Kuwazuru
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Mary Ann Suico
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
- Global Center for Natural Resources Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Sayaka Ogi
- Pharmaceuticals Research Laboratory, UBE Industries Ltd., Yamaguchi, Japan
| | - Ryoko Sasaki
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hidetoshi Sunamoto
- Pharmaceuticals Research Laboratory, UBE Industries Ltd., Yamaguchi, Japan
| | - Hirohiko Fukiya
- Pharmaceuticals Research Laboratory, UBE Industries Ltd., Yamaguchi, Japan
| | - Hayato Nishiyama
- Pharmaceuticals Research Laboratory, UBE Industries Ltd., Yamaguchi, Japan
| | - Misato Kamura
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
- Program for Leading Graduate School “HIGO (Health Life Science: Interdisciplinary and Glocal Oriented) Program,” Kumamoto University, Kumamoto, Japan
| | - Saki Niinou
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuimi Koyama
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Futoshi Nara
- Pharmaceuticals Research Laboratory, UBE Industries Ltd., Yamaguchi, Japan
| | - Tsuyoshi Shuto
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
- Global Center for Natural Resources Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazuhiro Onuma
- Pharmaceuticals Research Laboratory, UBE Industries Ltd., Yamaguchi, Japan
| | - Hirofumi Kai
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
- Program for Leading Graduate School “HIGO (Health Life Science: Interdisciplinary and Glocal Oriented) Program,” Kumamoto University, Kumamoto, Japan
- Global Center for Natural Resources Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
37
|
Extracellular vesicles derived from mesenchymal stromal cells as nanotherapeutics for liver ischaemia–reperfusion injury by transferring mitochondria to modulate the formation of neutrophil extracellular traps'. Biomaterials 2022; 284:121486. [DOI: 10.1016/j.biomaterials.2022.121486] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 02/08/2022] [Accepted: 03/23/2022] [Indexed: 12/23/2022]
|
38
|
Chen PM, Tsokos GC. Mitochondria in the Pathogenesis of Systemic Lupus Erythematosus. Curr Rheumatol Rep 2022; 24:88-95. [PMID: 35290598 DOI: 10.1007/s11926-022-01063-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2022] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by autoantibody production and inflammation in multiple organs. In this article, we present data on how various mitochondria pathologies are involved in the pathogenesis of the disease including the fact that they serve as a reservoir of autoantigens which contribute to the upending of lymphocyte tolerance. RECENT FINDINGS Mitochondrial DNA from various cell sources, including neutrophil extracellular traps, platelets, and red blood cells, elicits the production of type I interferon which contributes to breaking of peripheral tolerance. Mitochondrial DNA also serves as autoantigen targeted by autoantibodies. Mutations of mitochondrial DNA triggered by reactive oxygen species induce T cell cross-reactivity against self-antigens. Selective gene polymorphisms that regulate mitochondrial apoptosis in autoreactive B and T cells represent another key aspect in the induction of autoimmunity. Various mitochondrial abnormalities, including changes in mitochondrial function, oxidative stress, genetic polymorphism, mitochondrial DNA mutations, and apoptosis pathways, are each linked to different aspects of lupus pathogenesis. However, whether targeting these mitochondrial pathologies can be used to harness autoimmunity remains to be explored.
Collapse
Affiliation(s)
- Ping-Min Chen
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
39
|
Xin Y, Zhang B, Zhao J, Liu Q, Yin H, Lu Q. Animal models of systemic lupus erythematosus and their applications in drug discovery. Expert Opin Drug Discov 2022; 17:489-500. [PMID: 35287523 DOI: 10.1080/17460441.2022.2050691] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Systemic lupus erythematosus (SLE) is a multisystem autoimmune disease with substantial phenotypic heterogeneity. Currently, our understanding of the pathogenesis is still limited, and as a result, specific and efficacious therapies are lacking. Various mouse models have been established to serve as powerful tools that will promote a better understanding of the disease and the ability to test novel drugs before clinical application. AREAS COVERED The authors review the existing mouse models of SLE in terms of pathogenesis and manifestations, as well as their applications in drug discovery and development. The areas of focus include promising novel therapeutics that could benefit patients in the future and the contribution of mouse models used in preclinical studies. EXPERT OPINION Given the diversity of SLE mouse models with different characteristics, researchers must select a suitable model based on the mechanism involved. The use of multiple models is needed for drug testing studies to evaluate drug efficacy on different genetic backgrounds and other mechanisms to provide a reference for clinical trials.
Collapse
Affiliation(s)
- Yue Xin
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China.,Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiangsu, China
| | - Bo Zhang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China.,Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiangsu, China
| | - Junpeng Zhao
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China.,Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiangsu, China
| | - Qianmei Liu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China.,Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiangsu, China
| | - Haoyuan Yin
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China.,Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiangsu, China
| | - Qianjin Lu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China.,Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiangsu, China
| |
Collapse
|
40
|
Teng X, Brown J, Morel L. Redox Homeostasis Involvement in the Pharmacological Effects of Metformin in Systemic Lupus Erythematosus. Antioxid Redox Signal 2022; 36:462-479. [PMID: 34619975 PMCID: PMC8982129 DOI: 10.1089/ars.2021.0070] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 09/13/2021] [Accepted: 10/05/2021] [Indexed: 12/21/2022]
Abstract
Significance: Metformin has been proposed as a treatment for systemic lupus erythematosus (SLE). The primary target of metformin, the electron transport chain complex I in the mitochondria, is associated with redox homeostasis in immune cells, which plays a critical role in the pathogenesis of autoimmune diseases. This review addresses the evidence and knowledge gaps on whether a beneficial effect of metformin in lupus may be due to a restoration of a balanced redox state. Recent Advances: Clinical trials in SLE patients with mild-to-moderate disease activity and preclinical studies in mice have provided encouraging results for metformin. The mechanism by which this therapeutic effect was achieved is largely unknown. Metformin regulates redox homeostasis in a context-specific manner. Multiple cell types contribute to SLE, with evidence of increased mitochondrial oxidative stress in T cells and neutrophils. Critical Issues: The major knowledge gaps are whether the efficacy of metformin is linked to a restored redox homeostasis in the immune system, and if it does, in which cell types it occurs? We also need to know which patients may have a better response to metformin, and whether it corresponds to a specific mechanism? Finally, the identification of biomarkers to predict treatment outcomes would be of great value. Future Directions: Mechanistic studies must address the context-dependent pharmacological effects of metformin. Multiple cell types as well as a complex disease etiology should be considered. These studies must integrate the rapid advances made in understanding how metabolic programs direct the effector functions of immune cells. Antioxid. Redox Signal. 36, 462-479.
Collapse
Affiliation(s)
- Xiangyu Teng
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Josephine Brown
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Laurence Morel
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
41
|
Budd RC, Scharer CD, Barrantes-Reynolds R, Legunn S, Fortner KA. T Cell Homeostatic Proliferation Promotes a Redox State That Drives Metabolic and Epigenetic Upregulation of Inflammatory Pathways in Lupus. Antioxid Redox Signal 2022; 36:410-422. [PMID: 34328790 PMCID: PMC8982120 DOI: 10.1089/ars.2021.0078] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Significance: Numerous abnormalities in T cells have been described in patients with systemic lupus erythematosus (SLE), including lymphopenia, DNA demethylation, expression of endogenous retroviruses (ERVs), increased cell death, enlarged mitochondria, production of reactive oxygen species (ROS), and the appearance of unusual CD4-CD8- T cells. Our studies propose a model in which accelerated homeostatic proliferation of T cells promotes an epigenetic and metabolic program, leading to this cluster of abnormalities. Recent Advances: Growing knowledge of the innate immune disorders in SLE has included increased mitochondrial size and ROS production that induces oligomerization of the mitochondrial antiviral signaling (MAVS) protein and type I interferon production, as well as DNA demethylation, upregulation of inflammatory genes, and expression of certain ERVs in SLE peripheral blood mononuclear cells. All these events are part of the cellular program that occurs during homeostatic proliferation of T cells. Evidence from a murine model of SLE as well as in human SLE reveals that increased T cell homeostatic proliferation may be a driving factor in these processes. Critical Issues: Despite extensive knowledge of the myriad autoantibodies in SLE and other immune abnormalities, a cogent model has been lacking to link the numerous and seemingly disparate immune aberrations. This may partly explain the general lack of new drugs specifically for SLE in over 50 years. A more coherent model of SLE would not only unify the variety of immune abnormalities is SLE but would also suggest new therapies. Future Directions: The model of augmented homeostatic proliferation leading to increased mitochondrial mass, ROS, DNA demethylation, and upregulation of inflammatory genes suggests strategic new targets for SLE, including antioxidants and certain inhibitors of metabolism. Antioxid. Redox Signal. 36, 410-422.
Collapse
Affiliation(s)
- Ralph C Budd
- Department of Medicine, Vermont Center for Immunology and Infectious Diseases, Larner College of Medicine, The University of Vermont, Burlington, Vermont, USA
| | - Christopher D Scharer
- Department of Microbiology and Immunology, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Ramiro Barrantes-Reynolds
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, The University of Vermont, Burlington, Vermont, USA
| | - Scott Legunn
- Department of Medicine, Vermont Center for Immunology and Infectious Diseases, Larner College of Medicine, The University of Vermont, Burlington, Vermont, USA
| | - Karen A Fortner
- Department of Medicine, Vermont Center for Immunology and Infectious Diseases, Larner College of Medicine, The University of Vermont, Burlington, Vermont, USA
| |
Collapse
|
42
|
Badii M, Gaal O, Popp RA, Crisan TO, Joosten LAB. Trained immunity and inflammation in rheumatic diseases. Joint Bone Spine 2022; 89:105364. [DOI: 10.1016/j.jbspin.2022.105364] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/01/2022] [Accepted: 02/09/2022] [Indexed: 11/27/2022]
|
43
|
Sumikawa MH, Iwata S, Zhang M, Miyata H, Ueno M, Todoroki Y, Nagayasu A, Kanda R, Sonomoto K, Torimoto K, Lee S, Nakayamada S, Yamamoto K, Okada Y, Tanaka Y. An enhanced mitochondrial function through glutamine metabolism in plasmablast differentiation in systemic lupus erythematosus. Rheumatology (Oxford) 2021; 61:3049-3059. [PMID: 34730825 DOI: 10.1093/rheumatology/keab824] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/23/2021] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE To evaluate the dysfunction of B cell metabolism and its involvement in SLE pathology. METHODS We assessed the expression of metabolic markers of B cells in the peripheral blood of healthy controls (HCs) and SLE patients by using flow cytometry. In vitro, peripheral B cells were isolated from HCs and SLE patients to investigate the metabolic regulation mechanisms involved in their differentiation. RESULTS The expression level of DiOc6 (mitochondrial membrane hyperpolarization) was higher in B cells from SLE patients than in HCs, and correlated to the percentage of plasmablasts in CD19+ cells and with SLEDAI, a disease activity score. Stimulation of CD19+ cells with the Toll-like receptor 9 (TLR9) ligand CpG and IFN-α enhanced glycolysis, oxidative phosphorylation (OXPHOS), DiOc6 expression, and plasmablast differentiation in vitro. In the absence of glutamine, both glycolysis and OXPHOS were reduced, and plasmablast differentiation was suppressed, whereas there was no change in the absence of glucose. As glutamine is an important nutrient for protein synthesis, we further investigated the effect of the glutaminase inhibitor BPTES, which inhibits glutamine degradation, on metabolic regulation. BPTES reduced DiOc6 expression, OXPHOS, reactive oxygen species (ROS) production, ATP production, plasmablast differentiation without affecting glycolysis. Metformin inhibited CpG- and IFN-α-induced glutamine uptake, mitochondrial functions and suppressed plasmablast differentiation. CONCLUSIONS Mitochondrial dysfunction in B cells is associated with plasmablast differentiation and disease activity in SLE. Enhanced mitochondrial functions mediated by glutamine metabolism are important for plasmablast differentiation, which may be a potential therapeutic target for SLE.
Collapse
Affiliation(s)
- Maiko Hajime Sumikawa
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Shigeru Iwata
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Mingzeng Zhang
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Hiroko Miyata
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Masanobu Ueno
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Yasuyuki Todoroki
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Atsushi Nagayasu
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Ryuichiro Kanda
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Koshiro Sonomoto
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Keiichi Torimoto
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Seunghyun Lee
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Shingo Nakayamada
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Kazuo Yamamoto
- Biomedical Research Support Center, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Yosuke Okada
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| |
Collapse
|
44
|
Kalyanaraman B. Reactive oxygen species, proinflammatory and immunosuppressive mediators induced in COVID-19: overlapping biology with cancer. RSC Chem Biol 2021; 2:1402-1414. [PMID: 34704045 PMCID: PMC8496060 DOI: 10.1039/d1cb00042j] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022] Open
Abstract
This review analyzes the published literature linking the different mechanisms focused on oxidative stress and inflammation that contribute to COVID-19 disease severity. The objective is to bring together potential proinflammatory mechanisms of COVID-19 pathogenesis and address mitigation strategies using naturally occurring compounds and FDA-approved drugs. Outstanding questions addressed include the following: What is the mechanistic basis for linking enhanced vulnerability in COVID-19 to increased oxidative damage and proinflammatory mediators (e.g., cytokines), especially in high-risk people? Can we repurpose anti-inflammatory and immunomodulatory agents to mitigate inflammation in COVID-19 patients? How does 2-deoxy-d-glucose function as an anti-COVID drug? COVID-19, cancer biology, and immunotherapy share many mechanistic similarities. Repurposing drugs that already have been FDA-approved for mitigating inflammation and immunosuppression in cancer may be a way to counteract disease severity, progression, and chronic inflammation in COVID-19. What are the long-term effects of reactive oxygen species-inducing immune cells and sustained inflammation in so-called long-haulers (long COVID) after recovery from COVID-19? Can we use mitochondria-targeted agents prophylactically to prevent inflammation and boost immunity in long-haulers? Addressing the oxidative chemical biology of COVID-19 and the mechanistic commonalities with cancer may provide new insights potentially leading to appropriate clinical trials and new treatments.
Collapse
Affiliation(s)
- Balaraman Kalyanaraman
- Department of Biophysics, Cancer Center, Center for Disease Prevention Research, Medical College of Wisconsin 8701 Watertown Plank Road Milwaukee WI 53226 USA
| |
Collapse
|
45
|
Abnormal Mitochondrial Physiology in the Pathogenesis of Systemic Lupus Erythematosus. Rheum Dis Clin North Am 2021; 47:427-439. [PMID: 34215372 DOI: 10.1016/j.rdc.2021.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by abnormalities within the innate and adaptive immune systems. Activation and proliferation of a wide array of immune cells require significant up-regulation in cellular energy metabolism, with the mitochondria playing an essential role in the initiation and maintenance of this response. This article highlights how abnormal mitochondrial function may occur in SLE and focuses on how energy metabolism, oxidative stress, and impaired mitochondrial repair play a role in the pathogenesis of the disease. How this may represent an appealing novel therapeutic target for future drug therapy in SLE also is discussed.
Collapse
|
46
|
Abstract
Skewing of type I interferon (IFN) production and responses is a hallmark of systemic lupus erythematosus (SLE). Genetic and environmental contributions to IFN production lead to aberrant innate and adaptive immune activation even before clinical development of disease. Basic and translational research in this arena continues to identify contributions of IFNs to disease pathogenesis, and several promising therapeutic options for targeting of type I IFNs and their signaling pathways are in development for treatment of SLE patients.
Collapse
Affiliation(s)
- Sirisha Sirobhushanam
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, 5568 MSRB 2, 1150 West Medical Center Drive, Ann Arbor, MI 49109, USA
| | - Stephanie Lazar
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, 5568 MSRB 2, 1150 West Medical Center Drive, Ann Arbor, MI 49109, USA
| | - J Michelle Kahlenberg
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, 5570A MSRB 2, 1150 West Medical Center Drive, Ann Arbor, MI 49109, USA; Department of Dermatology, University of Michigan, 5570A MSRB 2, 1150 West Medical Center Drive, Ann Arbor, MI 49109, USA.
| |
Collapse
|
47
|
Scherlinger M, Tsokos GC. Reactive oxygen species: The Yin and Yang in (auto-)immunity. Autoimmun Rev 2021; 20:102869. [PMID: 34118461 DOI: 10.1016/j.autrev.2021.102869] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 04/21/2021] [Indexed: 12/24/2022]
Abstract
Reactive oxygen species (ROS) are produced by immune cells in response to antigens. They are produced mostly in the mitochondria and their levels are tightly controlled by a series of metabolic processes. ROS are necessary for the development of the immune response but the role of ROS in the development of autoimmune disease needs further clarification. Early clinical information points to the beneficial role of supplementation of antioxidant agents or the reduction of ROS production. We review recent information in the field in an effort to identify areas more studies are needed.
Collapse
Affiliation(s)
- Marc Scherlinger
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), France; Service de rhumatologie, Centre Hospitalier Universitaire de Strasbourg, 1 avenue Molière, 67098 Strasbourg, France.
| | - George C Tsokos
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
48
|
Liu Y, Kaplan MJ. Neutrophil Dysregulation in the Pathogenesis of Systemic Lupus Erythematosus. Rheum Dis Clin North Am 2021; 47:317-333. [PMID: 34215366 DOI: 10.1016/j.rdc.2021.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The recent identifications of a subset of proinflammatory neutrophils, low-density granulocytes, and their ability to readily form neutrophil extracellular traps led to a resurgence of interest in neutrophil dysregulation in the pathogenesis of systemic lupus erythematosus (SLE). This article presents an overview on how neutrophil dysregulation modulates the innate and adaptive immune responses in SLE and their putative roles in disease pathogenesis. The therapeutic potential of targeting this pathogenic process in the treatment of SLE is also discussed.
Collapse
Affiliation(s)
- Yudong Liu
- Department of Clinical Laboratory, Peking University People's Hospital, 11 Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, Intramural Research Program, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 10 Center Drive, 12N248C, Bethesda, MD 20892-1930, USA.
| |
Collapse
|
49
|
Yan LJ. NADH/NAD + Redox Imbalance and Diabetic Kidney Disease. Biomolecules 2021; 11:biom11050730. [PMID: 34068842 PMCID: PMC8153586 DOI: 10.3390/biom11050730] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 12/11/2022] Open
Abstract
Diabetic kidney disease (DKD) is a common and severe complication of diabetes mellitus. If left untreated, DKD can advance to end stage renal disease that requires either dialysis or kidney replacement. While numerous mechanisms underlie the pathogenesis of DKD, oxidative stress driven by NADH/NAD+ redox imbalance and mitochondrial dysfunction have been thought to be the major pathophysiological mechanism of DKD. In this review, the pathways that increase NADH generation and those that decrease NAD+ levels are overviewed. This is followed by discussion of the consequences of NADH/NAD+ redox imbalance including disruption of mitochondrial homeostasis and function. Approaches that can be applied to counteract DKD are then discussed, which include mitochondria-targeted antioxidants and mimetics of superoxide dismutase, caloric restriction, plant/herbal extracts or their isolated compounds. Finally, the review ends by pointing out that future studies are needed to dissect the role of each pathway involved in NADH-NAD+ metabolism so that novel strategies to restore NADH/NAD+ redox balance in the diabetic kidney could be designed to combat DKD.
Collapse
Affiliation(s)
- Liang-Jun Yan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
50
|
Spiroski AM, Niu Y, Nicholas LM, Austin-Williams S, Camm EJ, Sutherland MR, Ashmore TJ, Skeffington KL, Logan A, Ozanne SE, Murphy MP, Giussani DA. Mitochondria antioxidant protection against cardiovascular dysfunction programmed by early-onset gestational hypoxia. FASEB J 2021; 35:e21446. [PMID: 33788974 PMCID: PMC7612077 DOI: 10.1096/fj.202002705r] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/19/2021] [Accepted: 02/01/2021] [Indexed: 02/02/2023]
Abstract
Mitochondria-derived oxidative stress during fetal development increases cardiovascular risk in adult offspring of pregnancies complicated by chronic fetal hypoxia. We investigated the efficacy of the mitochondria-targeted antioxidant MitoQ in preventing cardiovascular dysfunction in adult rat offspring exposed to gestational hypoxia, integrating functional experiments in vivo, with those at the isolated organ and molecular levels. Rats were randomized to normoxic or hypoxic (13%-14% O2 ) pregnancy ± MitoQ (500 μM day-1 ) in the maternal drinking water. At 4 months of age, one cohort of male offspring was chronically instrumented with vascular catheters and flow probes to test in vivo cardiovascular function. In a second cohort, the heart was isolated and mounted onto a Langendorff preparation. To establish mechanisms linking gestational hypoxia with cardiovascular dysfunction and protection by MitoQ, we quantified the expression of antioxidant system, β-adrenergic signaling, and calcium handling genes in the fetus and adult, in frozen tissues from a third cohort. Maternal MitoQ in hypoxic pregnancy protected offspring against increased α1 -adrenergic reactivity of the cardiovascular system, enhanced reactive hyperemia in peripheral vascular beds, and sympathetic dominance, hypercontractility and diastolic dysfunction in the heart. Inhibition of Nfe2l2-mediated oxidative stress in the fetal heart and preservation of calcium regulatory responses in the hearts of fetal and adult offspring link molecular mechanisms to the protective actions of MitoQ treatment of hypoxic pregnancy. Therefore, these data show the efficacy of MitoQ in buffering mitochondrial stress through NADPH-induced oxidative damage and the prevention of programmed cardiovascular disease in adult offspring of hypoxic pregnancy.
Collapse
Affiliation(s)
- Ana-Mishel Spiroski
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.,Cambridge Cardiovascular Strategic Research Initiative, Cambridge, UK
| | - Youguo Niu
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.,Cambridge Cardiovascular Strategic Research Initiative, Cambridge, UK
| | - Lisa M Nicholas
- Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Shani Austin-Williams
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Emily J Camm
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Megan R Sutherland
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Thomas J Ashmore
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Katie L Skeffington
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Angela Logan
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Susan E Ozanne
- Cambridge Cardiovascular Strategic Research Initiative, Cambridge, UK.,Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.,Strategic Research Initiative in Reproduction, Cambridge, UK
| | - Michael P Murphy
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK.,Department of Medicine, University of Cambridge, Cambridge, UK
| | - Dino A Giussani
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.,Cambridge Cardiovascular Strategic Research Initiative, Cambridge, UK.,Strategic Research Initiative in Reproduction, Cambridge, UK
| |
Collapse
|