1
|
Wang H, Tang R, Pan Q, Yin Q, Feng J, Deng L. Mitochondria dysfunction: A trigger for cardiovascular diseases in systemic lupus erythematosus. Int Immunopharmacol 2025; 144:113722. [PMID: 39622131 DOI: 10.1016/j.intimp.2024.113722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/09/2024] [Accepted: 11/25/2024] [Indexed: 12/15/2024]
Abstract
Cardiovascular disease (CVD), including pericarditis, myocarditis, sudden cardiac death, coronary heart disease, and stroke, are leading contributors to morbidity and mortality in systemic lupus erythematosus (SLE) patients. Emerging evidence highlights mitochondrial dysfunction as a key driver of cardiovascular pathology in SLE, with impaired oxidative phosphorylation, altered membrane potential, and disrupted metabolic processes promoting oxidative stress, inflammatory activation, and endothelial dysfunction. This review critically examines mitochondrial contributions to CVD in SLE, comparing these mechanisms with those in non-SLE CVD to highlight SLE-specific mitochondrial vulnerabilities. Furthermore, we discuss preclinical and clinical findings supporting mitochondrial pathways as potential therapeutic targets, aiming to bridge gaps in current understanding and outline future research directions. By synthesizing current knowledge of mitochondrial dysregulation, this review proposes therapeutic strategies to improve cardiovascular outcomes and advance patient care in SLE.
Collapse
Affiliation(s)
- Haitao Wang
- The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Rui Tang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Qinyu Pan
- The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Qiuyan Yin
- The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Jian Feng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Li Deng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Department of Rheumatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
2
|
Bolton C. Review of evidence linking exposure to environmental stressors and associated alterations in the dynamics of immunosenescence (ISC) with the global increase in multiple sclerosis (MS). Immun Ageing 2024; 21:73. [PMID: 39438909 PMCID: PMC11494837 DOI: 10.1186/s12979-024-00473-w] [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: 08/13/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
Historical survey confirms that, over the latter part of the 20th century, autoimmune-based diseases, including multiple sclerosis (MS), have shown a worldwide increase in incidence and prevalence. Analytical population studies have established that the exponential rise in MS is not solely due to improvements in diagnosis and healthcare but relates to an increase in autoimmune risk factors. Harmful environmental exposures, including non-communicable social determinants of health, anthropogens and indigenous or transmissible microbes, constitute a group of causal determinants that have been closely linked with the global rise in MS cases. Exposure to environmental stressors has profound effects on the adaptive arm of the immune system and, in particular, the associated intrinsic process of immune ageing or immunosenescence (ISC). Stressor-related disturbances to the dynamics of ISC include immune cell-linked untimely or premature (p) alterations and an accelerated replicative (ar) change. A recognised immune-associated feature of MS is pISC and current evidence supports the presence of an arISC during the disease. Moreover, collated data illustrates the immune-associated alterations that characterise pISC and arISC are inducible by environmental stressors strongly implicated in causing duplicate changes in adaptive immune cells during MS. The close relationship between exposure to environmental risk factors and the induction of pISC and arISC during MS offers a valid mechanism through which pro-immunosenescent stressors may act and contribute to the recorded increase in the global rate and number of new cases of the disease. Confirmation of alterations to the dynamics of ISC during MS provides a rational and valuable therapeutic target for the use of senolytic drugs to either prevent accumulation and enhance ablation of less efficient untimely senescent adaptive immune cells or decelerate the dysregulated process of replicative proliferation. A range of senotherapeutics are available including kinase and transcriptase inhibitors, rapalogs, flavanols and genetically-engineered T cells and the use of selective treatments to control emerging and unspecified aspects of pISC and arISC are discussed.
Collapse
|
3
|
Xiong F, Shen K, Long D, Zhou S, Ruan P, Xin Y, Xiao Y, Peng W, Yang M, Wu H, Lu Q. Quercetin ameliorates lupus symptoms by promoting the apoptosis of senescent Tfh cells via the Bcl-2 pathway. Immun Ageing 2024; 21:69. [PMID: 39407236 PMCID: PMC11476537 DOI: 10.1186/s12979-024-00474-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/08/2024] [Indexed: 10/20/2024]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder that commonly affects the skin, kidneys, joints, and various other systemic tissues, with its development intricately linked to the process of immunosenescence. Quercetin (QC), a phytochemical that occurs naturally, demonstrates many different biological capabilities, such as antibacterial, antioxidant, and anti-inflammatory activities. Our investigation found that QC effectively reduced kidney damage and relieved mesenteric lymph nodes (mLNs) swelling in MRL/lpr lupus mice. Moreover, QC has been found to decrease the number of senescent follicular helper T (Tfh) cells, a pivotal kind of T cells that contribute to the progression of SLE. In vitro, QC exhibited the capacity to modulate mRNA expression levels, with the downregulation of IL-6, IL21-AS1, IL-27, BCL6, and BCL2L12, and the upregulation of FOXP1 and BIM. This modulation resulted in the suppression of Tfh cells differentiation and the enhancement of apoptosis in senescent CD4+ T cells. In addition, the HuProtTM Human Proteome Microarray revealed that QC can directly bind to BCL-2 protein and therefore promote the apoptosis of senescent CD4+ T cell. As a result, our investigative elucidate the potent inhibitory action of QC on the ontogeny of Tfh cells, along with its capacity to abrogate the immunosenescent phenotype. This positions QC as a promising therapeutic strategy for treating SLE.
Collapse
Affiliation(s)
- Feng Xiong
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Kai Shen
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Di Long
- Department of Dermatology, The Second Affiliated Hospital, Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Suqing Zhou
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Pinglang Ruan
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Yue Xin
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Yuezheng Xiao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Weijun Peng
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ming Yang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China.
| | - Haijing Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China.
| | - Qianjin Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China.
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China.
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.
| |
Collapse
|
4
|
Han F, Riaz F, Pu J, Gao R, Yang L, Wang Y, Song J, Liang Y, Wu Z, Li C, Tang J, Xu X, Wang X. Connecting the Dots: Telomere Shortening and Rheumatic Diseases. Biomolecules 2024; 14:1261. [PMID: 39456194 PMCID: PMC11506250 DOI: 10.3390/biom14101261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/24/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
Telomeres, repetitive sequences located at the extremities of chromosomes, play a pivotal role in sustaining chromosomal stability. Telomerase is a complex enzyme that can elongate telomeres by appending telomeric repeats to chromosome ends and acts as a critical factor in telomere dynamics. The gradual shortening of telomeres over time is a hallmark of cellular senescence and cellular death. Notably, telomere shortening appears to result from the complex interplay of two primary mechanisms: telomere shelterin complexes and telomerase activity. The intricate interplay of genetic, environmental, and lifestyle influences can perturb telomere replication, incite oxidative stress damage, and modulate telomerase activity, collectively resulting in shifts in telomere length. This age-related process of telomere shortening plays a considerable role in various chronic inflammatory and oxidative stress conditions, including cancer, cardiovascular disease, and rheumatic disease. Existing evidence has shown that abnormal telomere shortening or telomerase activity abnormalities are present in the pathophysiological processes of most rheumatic diseases, including different disease stages and cell types. The impact of telomere shortening on rheumatic diseases is multifaceted. This review summarizes the current understanding of the link between telomere length and rheumatic diseases in clinical patients and examines probable telomere shortening in peripheral blood mononuclear cells and histiocytes. Therefore, understanding the intricate interaction between telomere shortening and various rheumatic diseases will help in designing personalized treatment and control measures for rheumatic disease.
Collapse
Affiliation(s)
- Fang Han
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| | - Farooq Riaz
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen 518000, China;
- Center for Cancer Immunology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Jincheng Pu
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| | - Ronglin Gao
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| | - Lufei Yang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| | - Yanqing Wang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| | - Jiamin Song
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| | - Yuanyuan Liang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| | - Zhenzhen Wu
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| | - Chunrui Li
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| | - Jianping Tang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| | - Xianghuai Xu
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China;
| | - Xuan Wang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai 200065, China; (F.H.); (J.P.); (R.G.); (L.Y.); (Y.W.); (J.S.); (Y.L.); (Z.W.); (C.L.); (J.T.)
| |
Collapse
|
5
|
Jaiswal S, Kumar S, Sarkar B, Sinha RK. Therapeutic potential of Nelumbo nucifera Linn. in systemic lupus erythematosus: Network pharmacology and molecular modeling insights. Lupus 2024; 33:1155-1167. [PMID: 39135520 DOI: 10.1177/09612033241273074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
BACKGROUND Systemic lupus erythematosus is a chronic autoimmune inflammatory disease characterized by multiple symptoms. The phenolic acids and other flavonoids in Nelumbo nucifera have anti-oxidants, anti-inflammatory, and immunomodulatory activities that are essential for managing SLE through natural sources. This study employs network pharmacology to unveil the multi-target and multi-pathway mechanisms of Nelumbo nucifera as a complementary therapy. The findings are validated through molecular modeling, which includes molecular docking followed by a molecular dynamics study. METHODS Active compounds and targets of SLE were obtained from IMPPAT, KNApAcKFamily and SwissTargetPrediction databases. SLE-related targets were retrieved from GeneCards and OMIM databases. A protein-protein interaction (PPI) network was built to screen out the core targets using Cytoscape software. ShinyGO was used for GO and KEGG pathway enrichment analyses. Interactions between potential targets and active compounds were assessed by molecular docking and molecular dynamics simulation study. RESULTS In total, 12 active compounds and 1190 targets of N. nucifera's were identified. A network analysis of the PPI network revealed 10 core targets. GO and KEGG pathway enrichment analyses indicated that the effects of N. nucifera are mediated mainly by AGE-RAGE and other associated signalling pathways. Molecular docking indicated favourable binding affinities, particularly leucocianidol exhibiting less than -4.5 kcal/mol for all 10 targets. Subsequent molecular dynamics simulations of the leucocianidol-ESR1 complex aimed to elucidate the optimal binding complex's stability and flexibility. CONCLUSIONS Our study unveiled the potential therapeutic mechanism of N. nucifera in managing SLE. These findings provide insights for subsequent experimental validation and open up new avenues for further research in this field.
Collapse
Affiliation(s)
- Sugandha Jaiswal
- Department of Bioengineering and Biotechnology, Birla Institute of Technology(BIT), Mesra, Ranch, Jharkhand, India
| | - Satish Kumar
- Group Polyphenol-BIT, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Biswatrish Sarkar
- Group Polyphenol-BIT, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Rakesh Kumar Sinha
- Department of Bioengineering and Biotechnology, Birla Institute of Technology(BIT), Mesra, Ranch, Jharkhand, India
| |
Collapse
|
6
|
Shi J, Zhang M, Zhang L, Yu X, Sun L, Liu J, Zhao Y, Zheng W. Shelterin dysfunction promotes CD4+ T cell senescence in Behçet's disease. Rheumatology (Oxford) 2024; 63:2819-2827. [PMID: 38145496 DOI: 10.1093/rheumatology/kead703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/26/2023] [Accepted: 12/16/2023] [Indexed: 12/27/2023] Open
Abstract
OBJECTIVES To investigate the potential role of shelterin dysfunction in naïve CD4+ T cells in the pathogenesis of Behçet's disease (BD). METHODS Naïve CD4+ T cells were isolated from 40 BD patients and 40 sex- and age-matched healthy controls (HC). Senescent profiles, shelterin subunits expression, telomere length, telomerase activity and critical DNA damage response (DDR) were evaluated. Telomere repeat factor-2 (TRF2) silencing was conducted for further validation. RESULTS Compared with HC, BD patients had significantly decreased naïve CD4+ T cells, increased cell apoptosis, senescence, and productions of TNF-α and IFN-γ upon activation. Notably, BD naïve CD4+ T cells had shortened telomere, impaired telomerase activity, and expressed lower levels of shelterin subunits TRF2, TRF1- and TRF2-Interacting Nuclear Protein 2 (TIN2) and Repressor/Activator Protein 1 (RAP1). Furthermore, BD naïve CD4+ T cells exhibited significantly increased DDR, evidenced by elevated phosphorylated ataxia telangiectasia (AT) mutated (pATM), phosphorylated p53 (pp53) and p21. Finally, TRF2 silencing markedly upregulated DDR, apoptosis and proinflammatory cytokines production in HC naïve CD4+ T cells. CONCLUSION Our study demonstrated that TRF2 deficiency in BD naïve CD4+ T cells promoted cell apoptosis and senescence, leading to proinflammatory cytokines overproduction. Therefore, restoring TRF2 might be a promising therapeutic strategy for BD.
Collapse
Affiliation(s)
- Jing Shi
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Beijing, China
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Menghao Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Beijing, China
| | - Lili Zhang
- Department of Rheumatology, Linyi People's Hospital, Linyi, Shandong, China
| | - Xin Yu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Beijing, China
| | - Luxi Sun
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Jinjing Liu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Beijing, China
| | - Yan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Beijing, China
| | - Wenjie Zheng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Beijing, China
| |
Collapse
|
7
|
Shen CY, Lu CH, Cheng CF, Li KJ, Kuo YM, Wu CH, Liu CH, Hsieh SC, Tsai CY, Yu CL. Advanced Glycation End-Products Acting as Immunomodulators for Chronic Inflammation, Inflammaging and Carcinogenesis in Patients with Diabetes and Immune-Related Diseases. Biomedicines 2024; 12:1699. [PMID: 39200164 PMCID: PMC11352041 DOI: 10.3390/biomedicines12081699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/22/2024] [Accepted: 07/27/2024] [Indexed: 09/02/2024] Open
Abstract
Increased production of advanced glycation end products (AGEs) among reducing sugars (glucose, fructose, galactose, or ribose) and amino acids/proteins via non-enzymatic Maillard reaction can be found in lifestyle-related disease (LSRD), metabolic syndrome (MetS), and obesity and immune-related diseases. Increased serum levels of AGEs may induce aging, diabetic complications, cardiovascular diseases (CVD), neurodegenerative diseases (NDD), cancer, and inflamm-aging (inflammation with immunosenescence). The Maillard reaction can also occur among reducing sugars and lipoproteins or DNAs to alter their structure and induce immunogenicity/genotoxicity for carcinogenesis. AGEs, as danger-associated molecular pattern molecules (DAMPs), operate via binding to receptor for AGE (RAGE) or other scavenger receptors on cell surface to activate PI3K-Akt-, P38-MAPK-, ERK1/2-JNK-, and MyD88-induced NF-κB signaling pathways to mediate various pathological effects. Recently, the concept of "inflamm-aging" became more defined, and we have unveiled some interesting findings in relation to it. The purpose of the present review is to dissect the potential molecular basis of inflamm-aging in patients with diabetes and immune-mediated diseases caused by different AGEs.
Collapse
Affiliation(s)
- Chieh-Yu Shen
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, # 7 Chung-Shan South Road, Taipei 10002, Taiwan; (C.-Y.S.); (C.-H.L.); (C.-F.C.); (K.-J.L.)
| | - Cheng-Hsun Lu
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, # 7 Chung-Shan South Road, Taipei 10002, Taiwan; (C.-Y.S.); (C.-H.L.); (C.-F.C.); (K.-J.L.)
- Institute of Clinical Medicine, National Taiwan University College of Medicine, # 7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Chiao-Feng Cheng
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, # 7 Chung-Shan South Road, Taipei 10002, Taiwan; (C.-Y.S.); (C.-H.L.); (C.-F.C.); (K.-J.L.)
- Institute of Clinical Medicine, National Taiwan University College of Medicine, # 7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Ko-Jen Li
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, # 7 Chung-Shan South Road, Taipei 10002, Taiwan; (C.-Y.S.); (C.-H.L.); (C.-F.C.); (K.-J.L.)
| | - Yu-Min Kuo
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, # 7 Chung-Shan South Road, Taipei 10002, Taiwan; (C.-Y.S.); (C.-H.L.); (C.-F.C.); (K.-J.L.)
| | - Cheng-Han Wu
- Department of Internal Medicine, National Taiwan University Hospital-Hsinchu Branch, # 2, Section 1, Shengyi Road, Hsinchu County 302058, Taiwan;
| | - Chin-Hsiu Liu
- Department of Internal Medicine, National Taiwan University Hospital-Yunlin Branch, # 579, Section 2, Yunlin Road, Yunlin County 640203, Taiwan;
| | - Song-Chou Hsieh
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, # 7 Chung-Shan South Road, Taipei 10002, Taiwan; (C.-Y.S.); (C.-H.L.); (C.-F.C.); (K.-J.L.)
| | - Chang-Youh Tsai
- Department of Internal Medicine, Fu-Jen Catholic University Hospital, College of Medicine, Fu-Jen Catholic University, # 69 Guizi Road, New Taipei City 24352, Taiwan
| | - Chia-Li Yu
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, # 7 Chung-Shan South Road, Taipei 10002, Taiwan; (C.-Y.S.); (C.-H.L.); (C.-F.C.); (K.-J.L.)
| |
Collapse
|
8
|
Goldner B, Staffier KL. Case series: raw, whole, plant-based nutrition protocol rapidly reverses symptoms in three women with systemic lupus erythematosus and Sjögren's syndrome. Front Nutr 2024; 11:1208074. [PMID: 38505266 PMCID: PMC10949923 DOI: 10.3389/fnut.2024.1208074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 01/05/2024] [Indexed: 03/21/2024] Open
Abstract
Systemic lupus erythematosus (SLE) and Sjögren's syndrome (SS) are chronic autoimmune diseases. Symptoms of SLE can vary widely but often include fatigue, pain, photosensitivity, and, in some cases, nephritis. SS is frequently characterized by extreme dry eye and mouth, resulting from damage to moisture-producing glands, and is often present in combination with SLE. While the health benefits of plant-based diets have been well-established with respect to weight and cardiometabolic outcomes, less research is available to support the role of diet in treatment and management of autoimmune disease. This case series presents three women with SLE and SS who adopted a nutrition protocol to reverse symptoms of autoimmune disease. The protocol emphasizes leafy greens, cruciferous vegetables, omega-3 polyunsaturated fatty acids, and water, and includes predominately raw foods. The three patients reported dramatic improvements in physical symptoms, with nearly all symptoms of SLE and SS resolving after 4 weeks or less of adhering to the protocol. All three patients have remained symptom-free, two of whom have remained symptom-free for 6+ years with no recent medication use. Patients and practitioners should be made aware of the promising possibility of food as medicine in the treatment of SLE and SS. Future research should explore whether dietary changes may be a potential treatment strategy for individuals suffering from severe symptoms and poor quality of life due to SLE and SS.
Collapse
|
9
|
Hu WS, Chen JY, Liao WY, Chang CH, Chen TS. Regulation of ROS/inflammasome Axis is Essential for Cardiac Regeneration in Aging Rats Receiving Transplantation of Mesenchymal Stem Cells. Curr Stem Cell Res Ther 2024; 19:1393-1401. [PMID: 38031779 DOI: 10.2174/011574888x276612231121065203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 10/02/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Aging is a biological and gradual deterioration of function in living organisms. Aging is one of the risk factors for heart disease. OBJECTIVE Although mesenchymal stem cell transplantation shows potential in heart disease treatment, the relationship between stem cell-based therapy and oxidative stress/inflammasome axis regulation remains unclear. This study hypothesized that intervention of stem cells showed protective effect on heart aging induced by D-galactose through regulation of oxidative stress/inflammasome axis. METHODS An aging animal model was designed to test the above hypothesis. Experimental animals were divided into three groups, including Sham, D-gal (aging rats induced by d-galactose), and D-gal+WJSC (aging rats receiving mesenchymal stem cells). RESULTS Compared to the Sham, the experimental results indicate that structural alteration (HE stain and Masson's Trichrome stain), oxidative stress elevation (increase of TBARS level, expression of gp-91 and suppression of Sirt-1 as well as SOD2), increase of aging marker p53, suppression of cardiogenesis marker Troponin T, and inflammasome related protein markers expression (NLRP3, caspase-1 and IL-1 beta) were significantly observed in D-gal. In contrast, all pathological pathways were significantly improved in D-gal+WJSC when compared to D-gal. In addition, migration of stem cells to aging heart tissues was observed in the D-gal+WJSC group. CONCLUSION These findings suggest that mesenchymal stem cell transplantation effectively ameliorates aging hearts through oxidative stress/inflammasome axis regulation. The results from this study provide clinical potential for stem cell-based therapy in the treatment of aging hearts.
Collapse
Affiliation(s)
- Wei-Syun Hu
- School of Medicine, College of Medicine, China Medical University, Taichung, 40042, Taiwan
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Jing-Yi Chen
- Department of Life Science, National Taiwan Normal University, Taipei, 11677, Taiwan
| | - Wei-Yu Liao
- Department of Life Science, National Taiwan Normal University, Taipei, 11677, Taiwan
- Department of Traditional Chinese Medicine, En Chu Kong Hospital, New Taipei City, 40237, Taiwan
- Department of Cosmetic Science, Chang Gung University of Science and Technology, Taoyuan, 33303, Taiwan
| | - Chin-Hsien Chang
- Department of Traditional Chinese Medicine, En Chu Kong Hospital, New Taipei City, 40237, Taiwan
- Department of Cosmetic Science, Chang Gung University of Science and Technology, Taoyuan, 33303, Taiwan
- College of Chinese Medicine, China Medical University, Taichung City, 40402, Taiwan
| | - Tung-Sheng Chen
- Graduate Program of Biotechnology and Pharmaceutical Industries, National Taiwan Normal University, Taipei, 11677, Taiwan
| |
Collapse
|
10
|
Wei D, Jiang Y, Cheng J, Wang H, Sha K, Zhao J. Assessing the association of leukocyte telomere length with ankylosing spondylitis and rheumatoid arthritis: A bidirectional Mendelian randomization study. Front Immunol 2023; 14:1023991. [PMID: 37033949 PMCID: PMC10080099 DOI: 10.3389/fimmu.2023.1023991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 03/08/2023] [Indexed: 04/11/2023] Open
Abstract
Background Telomere length shortening can cause senescence and apoptosis in various immune cells, resulting in immune destabilization and ageing of the organism. In this study, we aimed to systematically assess the causal relationship of leukocyte telomere length (LTL) with ankylosing spondylitis (AS) and rheumatoid arthritis (RA) using a Mendelian randomization study. Methods LTL (n=472174) was obtained from the UK Biobank genome-wide association study pooled data. AS (n=229640), RA (n=212472) were obtained from FinnGen database. MR-Egger, inverse variance weighting, and weighted median methods were used to estimate the effects of causes. Cochran's Q test, MR Egger intercept test, MR-PRESSO, leave-one-out analysis, and funnel plots were used to look at sensitivity, heterogeneity, and multiple effects. Forward MR analysis considered LTL as the exposure and AS, RA as the outcome. Reverse MR analysis considered AS, RA as the exposure and LTL as the outcome. Results In the forward MR analysis, inverse variance-weighted and weighted median analysis results indicated that longer LTL might be associated with increased risk of AS (IVW: OR = 1.55, 95% CI: 1.14-2.11, p = 0.006). MR Egger regression analysis showed no pleiotropy between instrumental variables (IVs) (Egger intercept= 0.008, p = 0.294). The leave-one-out analysis showed that each single nucleotide polymorphism (SNP) of AS was robust to each outcome. No significant causal effects were found between AS, RA and LTL in the reverse MR analysis. Conclusion Longer LTL may be related with an increased risk of developing AS, and these findings provide a foundation for future clinical research on the causal association between LTL and AS.
Collapse
Affiliation(s)
- Donglei Wei
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yage Jiang
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jianwen Cheng
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Hui Wang
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Ke Sha
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jinmin Zhao
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- *Correspondence: Jinmin Zhao,
| |
Collapse
|
11
|
Different Types of Chronic Inflammation Engender Distinctive Immunosenescent Profiles in Affected Patients. Int J Mol Sci 2022; 23:ijms232314688. [PMID: 36499016 PMCID: PMC9735546 DOI: 10.3390/ijms232314688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022] Open
Abstract
Immunosenescence encompasses a spectrum of lymphocyte phenotypic alterations. The aim of the study was to evaluate immunosenescent effect of two different forms of chronic inflammation, Systemic Lupus Erythematosous (SLE), a systemic autoimmune disease, and End-Stage Kidney Disease (ESKD), a chronic inflammatory disorder. Certain lymphocyte surface molecules, including CD31, CD45RA, CCR7, CD28, CD57, for T, and IgD, CD27 for B lymphocytes, were analyzed by flow cytometry in 30 SLE and 53 ESKD patients on hemodialysis (HD), and results were compared to 31 healthy controls (HC) of similar age, gender, and nationality. Significant Lymphopenia was evident in both SLE and ESKD-HD patients, compared to HC, affecting B cells 75.4 (14.4−520.8), 97 (32−341), and 214 (84−576) cells/μL, respectively, p < 0.0001, and CD4 cells 651.2 (71.1−1478.2), 713 (234−1509), and 986 (344−1591) cells/μL, respectively, p < 0.0001. The allocation of B cell subpopulations was remarkably different between SLE and ESKD-HD patients. SLE showed a clear shift to senescence (CD19IgD-CD27−) cells, compared to ESKD-HD and HC, 11.75 (10)% vs. 8 (6) vs. 8.1 (10), respectively. Regarding T lymphocytes, Central Memory CD8 cells predominated in both SLE and ESKD-HD patients compared to HC, 53 (50)%, 52 (63), and 24 (64)%, respectively, while ESKD-HD but not SLE patients also had increased expression of CD4CD28− and CD8CD28− cells. In conclusion, both diseases are followed by significant lymphopenia; however, the senescent phenomenon affects the B lymphocyte compartment in SLE patients and T lymphocytes in ESKD-HD patients.
Collapse
|
12
|
Yang Y, Chen Y, Li Y, Feng Y, Hu N, Xue L. Expression and Significance of Programmed Death-1 and Its Ligands in the Accelerated Formation of Atherosclerosis in an Induced Murine Lupus Model. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6255383. [PMID: 39050559 PMCID: PMC11268968 DOI: 10.1155/2022/6255383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 09/30/2022] [Accepted: 10/19/2022] [Indexed: 07/27/2024]
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease that occurs in artery walls, which seriously affects the survival and prognosis of patients with systemic lupus erythematosus (SLE). Immune and inflammatory responses have notable effects on all stages of AS. In this study, we modeled SLE combined with AS in vivo via intraperitoneal injection of pristane (2,6,10,14-tetramethylpentadecane) into apolipoprotein E-knockout (ApoE-/- ) mice that had accelerated atherosclerotic lesions compared with wild-type (WT) ApoE-/- mice. In pristane-induced ApoE-/- mice, expression of programmed death-1 (PD-1) and programmed death-ligand 1 (PD-L1) in peripheral blood and on the surfaces of atherosclerotic lesions significantly increased, and levels of proinflammatory cytokines, namely, interferon-gamma (IFN-γ) and tumor necrosis factor alpha (TNF-α) in peripheral blood were elevated. We did not detect expression of programmed death-ligand 2 (PD-L2) in the arterial plaques of either pristane-induced or WT ApoE-/- mice, nor did we observe any significant difference in PD-L2 expression in peripheral blood between the two groups. Taken together, these results suggested that PD-1/PD-L1 signaling pathway might play an important regulatory role in the progression of AS in an induced murine lupus model which implies a potential target for treatment of AS in SLE.
Collapse
Affiliation(s)
- Yue Yang
- Department of Rheumatology, Yueyang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yueying Chen
- Department of Rheumatology, Yueyang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yongming Li
- Department of Rheumatology, Yueyang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yiyi Feng
- Department of Rheumatology, Yueyang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Na Hu
- Science and Technology Experimental Center, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Luan Xue
- Department of Rheumatology, Yueyang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| |
Collapse
|
13
|
Zhang T, Liu X, Zhao Y, Xu X, Liu Y, Wu X. Excessive IL-15 promotes cytotoxic CD4 + CD28- T cell-mediated renal injury in lupus nephritis. Immun Ageing 2022; 19:50. [PMID: 36320075 PMCID: PMC9624042 DOI: 10.1186/s12979-022-00305-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 08/28/2022] [Accepted: 10/10/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Patients with systemic lupus erythematosus (SLE) are highly susceptible to infection and cardiovascular events, suggesting that chronic antigenic stimulation may accelerate premature aging in SLE patients. Premature aging in SLE is often accompanied with the expansion of cytotoxic CD4 + CD28-T cells. Damage caused by CD4 + CD28- T cells enhances the progressive aging of the tissue function and loss of organism's fitness. The high serum level of IL-15 has been implicated in the pathogenesis of SLE, but its role in CD4 + CD28-T cell-mediated cytotoxicity in nephritic SLE remains unclear. The aim of this study was to investigate the effect of IL-15 on functional properties and associated renal damage of cytotoxic CD4 + CD28- T cell in lupus nephritis (LN). RESULTS Flow cytometry showed that the number of circulating innate-like CD4 + CD28- T cells was increased in patients with nephritic SLE. Immunofluorescence showed CD4 + CD28- T cell infiltration in the kidney of LN patients, which was correlated with multiple clinicopathological features including estimated glomerular filtration rate (eGFR), proteinuria, the proportion of glomerulosclerosis and the degree of renal chronicity. In addition, a high level of IL-15 and IL15-expressing macrophage infiltration was detected in the periglomerular and intraglomerular tissues of LN patients, which enhanced the innate features, cytokine secretion and migratory capability of CD4 + CD28- T cells, and finally exerted direct TCR-independent cytotoxicity on glomerular endothelial cells in an IL-15-dependent manner in vitro. CONCLUSION Our study demonstrated that excessive IL-15 potentially promoted cytotoxic CD4 + CD28- T cell-mediated renal damage in LN. This finding may provide new insights into the potential association of premature aging and tissue damage in LN.
Collapse
Affiliation(s)
- Ti Zhang
- grid.41156.370000 0001 2314 964XJinling Hospital, National Clinical Research Center of Kidney Diseases, Nanjing University School of Medicine, Nanjing, China
| | - Xin Liu
- grid.73113.370000 0004 0369 1660Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai, China
| | - Yue Zhao
- grid.41156.370000 0001 2314 964XJinling Hospital, National Clinical Research Center of Kidney Diseases, Nanjing University School of Medicine, Nanjing, China
| | - Xiaodong Xu
- grid.41156.370000 0001 2314 964XJinling Hospital, National Clinical Research Center of Kidney Diseases, Nanjing University School of Medicine, Nanjing, China
| | - Yaoyang Liu
- grid.73113.370000 0004 0369 1660Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai, China
| | - Xin Wu
- grid.73113.370000 0004 0369 1660Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai, China
| |
Collapse
|
14
|
Exploring the Molecular Mechanism of Zhi Bai Di Huang Wan in the Treatment of Systemic Lupus Erythematosus Based on Network Pharmacology and Molecular Docking Techniques. Processes (Basel) 2022. [DOI: 10.3390/pr10101914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective: To investigate the molecular mechanism and simulated validation of Zhi Bai Di Huang Pill (ZBDHP) for the treatment of systemic lupus erythematosus (SLE) using network pharmacology and molecular docking techniques. Methods: The active ingredients of ZBDHP were obtained through the TCMSP database and the Canonical SMILES of the active ingredients were queried through Pubchem. The targets of the active ingredients were predicted in the SwissTarget database based on the SMILES. The SLE-related disease targets were obtained through the GeneCards, OMIM and DisGenets databases, and the intersection targets of ZBDHP and SLE were obtained using the Venny 2.1.0 online platform. Intersection targets build a visual protein interaction network (PPI) through the STRING database, and the core targets were identified by network topology analysis. GO analysis and KEGG pathway enrichment analysis of the intersecting targets were performed using the DAVID database. Finally, the molecular docking of the first four active ingredients and the first four core target genes were verified by Pubchem, the PDB database and CB-Dock online molecular docking technology. Results: ZBDHP screened 91 potential active ingredients and 816 potential targets. Among them, 141 genes were intersected by ZBDHP and SLE. The network topology analysis showed that the main active ingredients were Hydroxygenkwanin, Alisol B, asperglaucide, Cerevisterol, etc., and the key target genes were TNF, AKT1, EGFR, STAT3, etc. GO and KEGG enrichment analysis showed that common targets interfere with biological processes or molecular functions such as signal transduction protein phosphorylation, inflammatory response, transmembrane receptor protein tyrosine kinase activity, etc., through multiple signaling pathways, such as pathways in cancer, Kaposi sarcoma-associated herpesvirus infection, the PI3K-Akt signaling pathway, lipid and atherosclerosis, hepatitis B, etc. Molecular docking results showed that the active components of ZBDHP have good binding activity to the core targets of SLE. Conclusions: This study reveals that the ZBDHP treatment of SLE is a complex mechanistic process with multi-components, multi-targets and multi-pathways, and it may play a therapeutic role in SLE by inhibiting the production, proliferation and apoptosis of inflammatory factors. In conclusion, the present study provides a theoretical basis for further research on ZBDHP.
Collapse
|
15
|
Moschetti L, Piantoni S, Vizzardi E, Sciatti E, Riccardi M, Franceschini F, Cavazzana I. Endothelial Dysfunction in Systemic Lupus Erythematosus and Systemic Sclerosis: A Common Trigger for Different Microvascular Diseases. Front Med (Lausanne) 2022; 9:849086. [PMID: 35462989 PMCID: PMC9023861 DOI: 10.3389/fmed.2022.849086] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/04/2022] [Indexed: 11/13/2022] Open
Abstract
This review describes the complex interplay between inflammation, vasculopathy and fibrosis that involve the heart and peripheral small vessels, leading to endothelial stiffness, vascular damage, and early aging in patients with systemic lupus erythematosus and systemic sclerosis, which represents two different models of vascular dysfunction among systemic autoimmune diseases. In fact, despite the fact that diagnostic methods and therapies have been significantly improved in the last years, affected patients show an excess of cardiovascular mortality if compared with the general population. In addition, we provide a complete overview on the new techniques which are used for the evaluation of endothelial dysfunction in a preclinical phase, which could represent a new approach in the assessment of cardiovascular risk in these patients.
Collapse
Affiliation(s)
- Liala Moschetti
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Silvia Piantoni
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- *Correspondence: Silvia Piantoni,
| | - Enrico Vizzardi
- Cardiology Unit, ASST Spedali Civili of Brescia, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | | | - Mauro Riccardi
- Cardiology Unit, ASST Spedali Civili of Brescia, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Franco Franceschini
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Ilaria Cavazzana
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
16
|
Perdaens O, van Pesch V. Molecular Mechanisms of Immunosenescene and Inflammaging: Relevance to the Immunopathogenesis and Treatment of Multiple Sclerosis. Front Neurol 2022; 12:811518. [PMID: 35281989 PMCID: PMC8913495 DOI: 10.3389/fneur.2021.811518] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/27/2021] [Indexed: 12/18/2022] Open
Abstract
Aging is characterized, amongst other features, by a complex process of cellular senescence involving both innate and adaptive immunity, called immunosenescence and associated to inflammaging, a low-grade chronic inflammation. Both processes fuel each other and partially explain increasing incidence of cancers, infections, age-related autoimmunity, and vascular disease as well as a reduced response to vaccination. Multiple sclerosis (MS) is a lifelong disease, for which considerable progress in disease-modifying therapies (DMTs) and management has improved long-term survival. However, disability progression, increasing with age and disease duration, remains. Neurologists are now involved in caring for elderly MS patients, with increasing comorbidities. Aging of the immune system therefore has relevant implications for MS pathogenesis, response to DMTs and the risks mediated by these treatments. We propose to review current evidence regarding markers and molecular mechanisms of immunosenescence and their relevance to understanding MS pathogenesis. We will focus on age-related changes in the innate and adaptive immune system in MS and other auto-immune diseases, such as systemic lupus erythematosus and rheumatoid arthritis. The consequences of these immune changes on MS pathology, in interaction with the intrinsic aging process of central nervous system resident cells will be discussed. Finally, the impact of immunosenescence on disease evolution and on the safety and efficacy of current DMTs will be presented.
Collapse
Affiliation(s)
- Océane Perdaens
- Laboratory of Neurochemistry, Institute of Neuroscience, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Vincent van Pesch
- Laboratory of Neurochemistry, Institute of Neuroscience, Université catholique de Louvain (UCLouvain), Brussels, Belgium
- Department of Neurology, Cliniques universitaires Saint-Luc, Université catholique de Louvain (UCLouvain), Brussels, Belgium
- *Correspondence: Vincent van Pesch
| |
Collapse
|
17
|
Shim DW, Cho HJ, Hwang I, Jung TY, Kim HS, Ryu JH, Yu JW. Intracellular NAD+ Depletion Confers a Priming Signal for NLRP3 Inflammasome Activation. Front Immunol 2021; 12:765477. [PMID: 34987507 PMCID: PMC8722528 DOI: 10.3389/fimmu.2021.765477] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 12/03/2021] [Indexed: 11/13/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an important cofactor in many redox and non-redox NAD+-consuming enzyme reactions. Intracellular NAD+ level steadily declines with age, but its role in the innate immune potential of myeloid cells remains elusive. In this study, we explored whether NAD+ depletion by FK866, a highly specific inhibitor of the NAD salvage pathway, can affect pattern recognition receptor-mediated responses in macrophages. NAD+-depleted mouse bone marrow-derived macrophages (BMDMs) exhibited similar levels of proinflammatory cytokine production in response to LPS or poly (I:C) stimulation compared with untreated cells. Instead, FK866 facilitated robust caspase-1 activation in BMDMs in the presence of NLRP3-activating signals such as ATP and nigericin, a potassium ionophore. However, this FK866-mediated caspase-1 activation was completely abolished in Nlrp3-deficient macrophages. FK866 plus nigericin stimulation caused an NLRP3-dependent assembly of inflammasome complex. In contrast, restoration of NAD+ level by supplementation with nicotinamide mononucleotide abrogated the FK866-mediated caspase-1 cleavage. FK866 did not induce or increase the expression levels of NLRP3 and interleukin (IL)-1β but drove mitochondrial retrograde transport into the perinuclear region. FK866-nigericin-induced mitochondrial transport is critical for caspase-1 cleavage in macrophages. Consistent with the in vitro experiments, intradermal coinjection of FK866 and ATP resulted in robust IL-1β expression and caspase-1 activation in the skin of wild-type, but not Nlrp3-deficient mice. Collectively, our data suggest that NAD+ depletion provides a non-transcriptional priming signal for NLRP3 activation via mitochondrial perinuclear clustering, and aging-associated NAD+ decline can trigger NLRP3 inflammasome activation in ATP-rich environments.
Collapse
Affiliation(s)
- Do-Wan Shim
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyo-Joung Cho
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Inhwa Hwang
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Taek-Yeol Jung
- Department of Life Science, College of Natural Science, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Hyun-Seok Kim
- Department of Life Science, College of Natural Science, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Ju Hee Ryu
- Theragnosis Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Je-Wook Yu
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
- *Correspondence: Je-Wook Yu,
| |
Collapse
|
18
|
Eschborn M, Pawlitzki M, Wirth T, Nelke C, Pfeuffer S, Schulte-Mecklenbeck A, Lohmann L, Rolfes L, Pape K, Eveslage M, Bittner S, Gross CC, Ruck T, Wiendl H, Meuth SG, Klotz L. Evaluation of Age-Dependent Immune Signatures in Patients With Multiple Sclerosis. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/6/e1094. [PMID: 34667129 DOI: 10.1212/nxi.0000000000001094] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 08/30/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND OBJECTIVES In MS, an age-related decline in disease activity and a decreased efficacy of disease-modifying treatment have been linked to immunosenescence, a state of cellular dysfunction associated with chronic inflammation. METHODS To evaluate age-related immunologic alterations in MS, we compared immune signatures in peripheral blood (PB) and CSF by flow cytometry in patients with relapsing-remitting (RR) (PB n = 38; CSF n = 51) and primary progressive (PP) MS (PB n = 40; CSF n = 36) and respective controls (PB n = 40; CSF n = 85). RESULTS Analysis revealed significant age-related changes in blood immune cell composition, especially in the CD8 T-cell compartment of healthy donors (HDs) and patients with MS. However, HDs displayed a strong age-dependent decline in the expression of the immunoregulatory molecules KLRG1, LAG3, and CTLA-4 on memory CD8 T cells, whereas this age-dependent reduction was completely abrogated in patients with MS. An age-dependent increase in the expression of the costimulatory molecule CD226 on memory CD8 T cells was absent in patients with MS. CD226 expression correlated with disability in younger (≤50 years) patients with MS. CSF analysis revealed a significant age-dependent decline in various immune cell populations in PPMS but not RRMS, suggesting a differential effect of aging on the intrathecal compartment in PPMS. DISCUSSION Our data illustrate that aging in MS is associated with a dysbalance between costimulatory and immunoregulatory signals provided by CD8 T cells favoring a proinflammatory phenotype and, more importantly, a pattern of premature immune aging in the CD8 T-cell compartment of young patients with MS with potential implications for disease severity.
Collapse
Affiliation(s)
- Melanie Eschborn
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Marc Pawlitzki
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Timo Wirth
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Christopher Nelke
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Steffen Pfeuffer
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Andreas Schulte-Mecklenbeck
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Lisa Lohmann
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Leoni Rolfes
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Katrin Pape
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Maria Eveslage
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Stefan Bittner
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Catharina C Gross
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Tobias Ruck
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Heinz Wiendl
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Sven G Meuth
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia
| | - Luisa Klotz
- From the Department of Neurology with Institute of Translational Neurology (M. Eschborn, M.P., T.W., S.P., A.S.-M., L.L., C.C.G., H.W., L.K.), University Hospital Münster, Münster, Germany; Department of Neurology with Institute of Translational Neurology (C.N., L.R., T.R., S.G.M.), University Hospital Münster, Münster, Germany; present address: Department of Neurology, University Hospital Düsseldorf, Düsseldorf, Germany; Department of Neurology (K.P., S.B.), Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Biostatistics and Clinical Research (M. Eveslage), University of Münster, Münster, Germany; and Brain and Mind Centre (H.W.), Medical Faculty, University of Sydney, Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
19
|
Santos-Moreno P, Burgos-Angulo G, Martinez-Ceballos MA, Pizano A, Echeverri D, Bautista-Niño PK, Roks AJM, Rojas-Villarraga A. Inflammaging as a link between autoimmunity and cardiovascular disease: the case of rheumatoid arthritis. RMD Open 2021; 7:rmdopen-2020-001470. [PMID: 33468563 PMCID: PMC7817822 DOI: 10.1136/rmdopen-2020-001470] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/27/2020] [Accepted: 12/21/2020] [Indexed: 12/13/2022] Open
Abstract
Currently, traditional and non-traditional risk factors for cardiovascular disease have been established. The first group includes age, which constitutes one of the most important factors in the development of chronic diseases. The second group includes inflammation, the pathophysiology of which contributes to an accelerated process of vascular remodelling and atherogenesis in autoimmune diseases. Indeed, the term inflammaging has been used to refer to the inflammatory origin of ageing, explicitly due to the chronic inflammatory process associated with age (in healthy individuals). Taking this into account, it can be inferred that people with autoimmune diseases are likely to have an early acceleration of vascular ageing (vascular stiffness) as evidenced in the alteration of non-invasive cardiovascular tests such as pulse wave velocity. Thus, an association is created between autoimmunity and high morbidity and mortality rates caused by cardiovascular disease in this population group. The beneficial impact of the treatments for rheumatoid arthritis at the cardiovascular level has been reported, opening new opportunities for pharmacotherapy.
Collapse
Affiliation(s)
| | - Gabriel Burgos-Angulo
- Internal Medicine Department, Fundación Universitaria de Ciencias de la Salud (FUCS), Bogota, Cundinamarca, Colombia
| | | | - Alejandro Pizano
- Vascular Function Research Laboratory and Department of Interventional Cardiology, Fundación Cardioinfantil Instituto de Cardiología, Bogota, Cundinamarca, Colombia
| | - Dario Echeverri
- Vascular Function Research Laboratory and Department of Interventional Cardiology, Fundación Cardioinfantil Instituto de Cardiología, Bogota, Cundinamarca, Colombia
| | - Paula K Bautista-Niño
- Research Center, Fundación Cardiovascular de Colombia, Floridablanca, Santander, Colombia
| | - Anton J M Roks
- Department of Internal Medicine, Division of Pharmacology and Vascular Medicine, Erasmus Medical Center Department of Internal Medicine, Rotterdam, South Holland, The Netherlands
| | - Adriana Rojas-Villarraga
- Research Division, Fundación Universitaria de Ciencias de la Salud (FUCS), Bogotá, Cundinamarca, Colombia
| |
Collapse
|
20
|
Impaired Differentiation of Highly Proliferative ICOS +-Tregs Is Involved in the Transition from Low to High Disease Activity in Systemic Lupus Erythematosus (SLE) Patients. Int J Mol Sci 2021; 22:ijms22179501. [PMID: 34502409 PMCID: PMC8430608 DOI: 10.3390/ijms22179501] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/15/2021] [Accepted: 08/26/2021] [Indexed: 12/15/2022] Open
Abstract
Dysregulations in the differentiation of CD4+-regulatory-T-cells (Tregs) and CD4+-responder-T-cells (Tresps) are involved in the development of active systemic lupus erythematosus (SLE). Three differentiation pathways of highly proliferative inducible costimulatory molecule (ICOS)+- and less proliferative ICOS--CD45RA+CD31+-recent-thymic-emigrant (RTE)-Tregs/Tresps via CD45RA-CD31+-memory-Tregs/Tresps (CD31+-memory-Tregs/Tresps), their direct proliferation via CD45RA+CD31--mature naïve (MN)-Tregs/Tresps, and the production and differentiation of resting MN-Tregs/Tresp into CD45RA-CD31--memory-Tregs/Tresps (CD31--memory-Tregs/Tresps) were examined in 115 healthy controls, 96 SLE remission patients, and 20 active disease patients using six color flow cytometric analysis. In healthy controls an appropriate sequence of these pathways ensured regular age-dependent differentiation. In SLE patients, an age-independently exaggerated differentiation was observed for all Treg/Tresp subsets, where the increased conversion of resting MN-Tregs/Tresps particularly guaranteed the significantly increased ratios of ICOS+-Tregs/ICOS+-Tresps and ICOS--Tregs/ICOS--Tresps during remission. Changes in the differentiation of resting ICOS+-MN-Tresps and ICOS--MN-Tregs from conversion to proliferation caused a significant shift in the ratio of ICOS+-Tregs/ICOS+-Tresps in favor of ICOS+-Tresps and a further increase in the ratio of ICOS--Tregs/ICOS--Tresps with active disease. The differentiation of ICOS+-RTE-Tregs/Tresps seems to be crucial for keeping patients in remission, where their limited production of proliferating resting MN-Tregs may be responsible for the occurrence of active disease flares.
Collapse
|
21
|
Guimarães GR, Almeida PP, de Oliveira Santos L, Rodrigues LP, de Carvalho JL, Boroni M. Hallmarks of Aging in Macrophages: Consequences to Skin Inflammaging. Cells 2021; 10:cells10061323. [PMID: 34073434 PMCID: PMC8228751 DOI: 10.3390/cells10061323] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/22/2021] [Accepted: 05/22/2021] [Indexed: 12/12/2022] Open
Abstract
The skin is our largest organ and the outermost protective barrier. Its aging reflects both intrinsic and extrinsic processes resulting from the constant insults it is exposed to. Aging in the skin is accompanied by specific epigenetic modifications, accumulation of senescent cells, reduced cellular proliferation/tissue renewal, altered extracellular matrix, and a proinflammatory environment favoring undesirable conditions, including disease onset. Macrophages (Mφ) are the most abundant immune cell type in the skin and comprise a group of heterogeneous and plastic cells that are key for skin homeostasis and host defense. However, they have also been implicated in orchestrating chronic inflammation during aging. Since Mφ are related to innate and adaptive immunity, it is possible that age-modified skin Mφ promote adaptive immunity exacerbation and exhaustion, favoring the emergence of proinflammatory pathologies, such as skin cancer. In this review, we will highlight recent findings pertaining to the effects of aging hallmarks over Mφ, supporting the recognition of such cell types as a driving force in skin inflammaging and age-related diseases. We will also present recent research targeting Mφ as potential therapeutic interventions in inflammatory skin disorders and cancer.
Collapse
Affiliation(s)
- Gabriela Rapozo Guimarães
- Laboratory of Bioinformatics and Computational Biology, Division of Experimental and Translational Research, Brazilian National Cancer Institute (INCA), Rio de Janeiro 20231-050, Brazil; (G.R.G.); (P.P.A.); (L.d.O.S.)
| | - Palloma Porto Almeida
- Laboratory of Bioinformatics and Computational Biology, Division of Experimental and Translational Research, Brazilian National Cancer Institute (INCA), Rio de Janeiro 20231-050, Brazil; (G.R.G.); (P.P.A.); (L.d.O.S.)
| | - Leandro de Oliveira Santos
- Laboratory of Bioinformatics and Computational Biology, Division of Experimental and Translational Research, Brazilian National Cancer Institute (INCA), Rio de Janeiro 20231-050, Brazil; (G.R.G.); (P.P.A.); (L.d.O.S.)
| | - Leane Perim Rodrigues
- Genomic Sciences and Biotechnology Program, Catholic University of Brasilia, Brasilia 70790-160, Brazil; (L.P.R.); (J.L.d.C.)
| | - Juliana Lott de Carvalho
- Genomic Sciences and Biotechnology Program, Catholic University of Brasilia, Brasilia 70790-160, Brazil; (L.P.R.); (J.L.d.C.)
- Faculty of Medicine, University of Brasilia, Brasilia 70910-900, Brazil
| | - Mariana Boroni
- Laboratory of Bioinformatics and Computational Biology, Division of Experimental and Translational Research, Brazilian National Cancer Institute (INCA), Rio de Janeiro 20231-050, Brazil; (G.R.G.); (P.P.A.); (L.d.O.S.)
- Experimental Medicine Research Cluster (EMRC), University of Campinas (UNICAMP), Campinas 13083-970, Brazil
- Correspondence:
| |
Collapse
|
22
|
Bolton C. An evaluation of the recognised systemic inflammatory biomarkers of chronic sub-optimal inflammation provides evidence for inflammageing (IFA) during multiple sclerosis (MS). Immun Ageing 2021; 18:18. [PMID: 33853634 PMCID: PMC8045202 DOI: 10.1186/s12979-021-00225-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 03/12/2021] [Indexed: 01/11/2023]
Abstract
The pathogenesis of the human demyelinating disorder multiple sclerosis (MS) involves the loss of immune tolerance to self-neuroantigens. A deterioration in immune tolerance is linked to inherent immune ageing, or immunosenescence (ISC). Previous work by the author has confirmed the presence of ISC during MS. Moreover, evidence verified a prematurely aged immune system that may change the frequency and profile of MS through an altered decline in immune tolerance. Immune ageing is closely linked to a chronic systemic sub-optimal inflammation, termed inflammageing (IFA), which disrupts the efficiency of immune tolerance by varying the dynamics of ISC that includes accelerated changes to the immune system over time. Therefore, a shifting deterioration in immunological tolerance may evolve during MS through adversely-scheduled effects of IFA on ISC. However, there is, to date, no collective proof of ongoing IFA during MS. The Review addresses the constraint and provides a systematic critique of compelling evidence, through appraisal of IFA-related biomarker studies, to support the occurrence of a sub-optimal inflammation during MS. The findings justify further work to unequivocally demonstrate IFA in MS and provide additional insight into the complex pathology and developing epidemiology of the disease.
Collapse
|
23
|
Zhang B, Gems D. Gross ways to live long: Parasitic worms as an anti-inflammaging therapy? eLife 2021; 10:65180. [PMID: 33526169 PMCID: PMC7853715 DOI: 10.7554/elife.65180] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 01/05/2021] [Indexed: 12/11/2022] Open
Abstract
Evolutionary medicine argues that disease can arise because modern conditions do not match those in which we evolved. For example, a decline in exposure to commensal microbes and gastrointestinal helminths in developed countries has been linked to increased prevalence of allergic and autoimmune inflammatory disorders (the hygiene hypothesis). Accordingly, probiotic therapies that restore ‘old friend’ microbes and helminths have been explored as Darwinian treatments for these disorders. A further possibility is that loss of old friend commensals also increases the sterile, aging-associated inflammation known as inflammaging, which contributes to a range of age-related diseases, including cardiovascular disease, dementia, and cancer. Interestingly, Crowe et al., 2020 recently reported that treatment with a secreted glycoprotein from a parasitic nematode can protect against murine aging by induction of anti-inflammatory mechanisms. Here, we explore the hypothesis that restorative helminth therapy would have anti-inflammaging effects. Could worm infections provide broad-spectrum protection against age-related disease?
Collapse
Affiliation(s)
- Bruce Zhang
- Institute of Healthy Ageing, and Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| | - David Gems
- Institute of Healthy Ageing, and Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| |
Collapse
|
24
|
Katz-Agranov N, Zandman-Goddard G. The microbiome links between aging and lupus. Autoimmun Rev 2021; 20:102765. [PMID: 33476814 DOI: 10.1016/j.autrev.2021.102765] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 11/05/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS Many forms of immune dysregulation, which lead to inflammaging and senescence, have been demonstrated in patients with systemic lupus erythematosus (SLE; lupus) and in the aging population. The discovery of the microbiome and its association with human health and pathology has led it to be the center of investigation as a major contributor to the pathogenesis of immunosenescence in both populations. Similar alterations to the microbiome in the form of dysbiosis, that are demonstrated in both aging and in lupus patients, may help explain the significant overlap in clinical manifestations seen in these groups. METHODS We performed an extensive literature review, utilizing the Pubmed search engine and Google Scholar for studies evaluating the microbiome in two groups, elderly populations and lupus patients (both murine and human models), between the years 2000-2019. We searched for the terms: microbiome, dysbiosis, lupus, elderly, aging and inflammaging, which yielded hundreds of articles, of which 114 were used for preparation of this paper. We compared the similarities between the populations. RESULTS We found that the similar processes of immune dysregulation, in both aging populations and lupus patients, extend to the microbiome, in the form of dysbiosis. Some of these similarities include loss of microbiota biodiversity, increased representation of microbes that are associated with inflammation and disease (i.e Proteobacteria, Bacteroidetes), a relative decrease in protective microbes with "anti-inflammatory" properties (i.e Firmicutes) and a subsequent compromise to the intestinal barrier, leading to leakage of proinflammatory microbial components in both groups. CONCLUSIONS We conclude that there are several similar alterations in the composition and function of the microbiome of lupus patients and aging individuals, leading to immunosenescence, which may also be a contributing mechanism in lupus. It seems in fact that the microbiome of SLE may actually be analogous to immunosenescence. This knowledge may help the continuous efforts in finding a solution for both conditions.
Collapse
Affiliation(s)
- Nurit Katz-Agranov
- Department of Medicine, Saint Elizabeth's Medical Center, Boston, MA, USA; Tufts University School of Medicine, Boston, MA, USA
| | - Gisele Zandman-Goddard
- Department of Medicine C, Wolfson Medical Center, Holon, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
25
|
Tsai CY, Shen CY, Liu CW, Hsieh SC, Liao HT, Li KJ, Lu CS, Lee HT, Lin CS, Wu CH, Kuo YM, Yu CL. Aberrant Non-Coding RNA Expression in Patients with Systemic Lupus Erythematosus: Consequences for Immune Dysfunctions and Tissue Damage. Biomolecules 2020; 10:biom10121641. [PMID: 33291347 PMCID: PMC7762297 DOI: 10.3390/biom10121641] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 12/11/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex systemic autoimmune disease with heterogeneous clinical manifestations. A diverse innate and adaptive immune dysregulation is involved in the immunopathogenesis of SLE. The dysregulation of immune-related cells may derive from the intricate interactions among genetic, epigenetic, environmental, and immunological factors. Of these contributing factors, non-coding RNAs (ncRNAs), including microRNAs (miRNAs, miRs), and long non-coding RNAs (lncRNAs) play critical roles in the post-transcriptional mRNA expression of cytokines, chemokines, and growth factors, which are essential for immune modulation. In the present review, we emphasize the roles of ncRNA expression in the immune-related cells and cell-free plasma, urine, and tissues contributing to the immunopathogenesis and tissue damage in SLE. In addition, the circular RNAs (circRNA) and their post-translational regulation of protein synthesis in SLE are also briefly described. We wish these critical reviews would be useful in the search for biomarkers/biosignatures and novel therapeutic strategies for SLE patients in the future.
Collapse
MESH Headings
- Adaptive Immunity/genetics
- Autoimmunity/genetics
- Chemokines/genetics
- Chemokines/immunology
- Dendritic Cells/immunology
- Dendritic Cells/pathology
- Gene Expression Regulation
- Humans
- Immunity, Innate/genetics
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/immunology
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Lupus Erythematosus, Systemic/blood
- Lupus Erythematosus, Systemic/genetics
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/pathology
- MicroRNAs/genetics
- MicroRNAs/immunology
- Neutrophils/immunology
- Neutrophils/pathology
- RNA, Circular/genetics
- RNA, Circular/immunology
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/immunology
- RNA, Messenger/genetics
- RNA, Messenger/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
Collapse
Affiliation(s)
- Chang-Youh Tsai
- Division of Allergy, Immunology & Rheumatology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei 11217, Taiwan; (C.-W.L.); (H.-T.L.)
- Correspondence: (C.-Y.T.); (C.-L.Y.)
| | - Chieh-Yu Shen
- Division of Rheumatology, Immunology, & Allergy, Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (C.-Y.S.); (S.-C.H.); (K.-J.L.); (C.-S.L.); (C.-H.W.); (Y.-M.K.)
- Institute of Clinical Medicine, National Taiwan University School of Medicine, Taipei 10002, Taiwan
| | - Chih-Wei Liu
- Division of Allergy, Immunology & Rheumatology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei 11217, Taiwan; (C.-W.L.); (H.-T.L.)
| | - Song-Chou Hsieh
- Division of Rheumatology, Immunology, & Allergy, Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (C.-Y.S.); (S.-C.H.); (K.-J.L.); (C.-S.L.); (C.-H.W.); (Y.-M.K.)
| | - Hsien-Tzung Liao
- Division of Allergy, Immunology & Rheumatology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei 11217, Taiwan; (C.-W.L.); (H.-T.L.)
| | - Ko-Jen Li
- Division of Rheumatology, Immunology, & Allergy, Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (C.-Y.S.); (S.-C.H.); (K.-J.L.); (C.-S.L.); (C.-H.W.); (Y.-M.K.)
| | - Cheng-Shiun Lu
- Division of Rheumatology, Immunology, & Allergy, Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (C.-Y.S.); (S.-C.H.); (K.-J.L.); (C.-S.L.); (C.-H.W.); (Y.-M.K.)
| | - Hui-Ting Lee
- Mackay Memorial Hospital and Mackay College of Medicine, Taipei 10449, Taiwan;
| | - Cheng-Sung Lin
- Department of Thoracic Surgery, Ministry of Health and Welfare Taipei Hospital, New Taipei City 24213, Taiwan;
| | - Cheng-Han Wu
- Division of Rheumatology, Immunology, & Allergy, Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (C.-Y.S.); (S.-C.H.); (K.-J.L.); (C.-S.L.); (C.-H.W.); (Y.-M.K.)
| | - Yu-Min Kuo
- Division of Rheumatology, Immunology, & Allergy, Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (C.-Y.S.); (S.-C.H.); (K.-J.L.); (C.-S.L.); (C.-H.W.); (Y.-M.K.)
| | - Chia-Li Yu
- Division of Rheumatology, Immunology, & Allergy, Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (C.-Y.S.); (S.-C.H.); (K.-J.L.); (C.-S.L.); (C.-H.W.); (Y.-M.K.)
- Correspondence: (C.-Y.T.); (C.-L.Y.)
| |
Collapse
|
26
|
Ding X, Xiang W, He X. IFN-I Mediates Dysfunction of Endothelial Progenitor Cells in Atherosclerosis of Systemic Lupus Erythematosus. Front Immunol 2020; 11:581385. [PMID: 33262760 PMCID: PMC7686511 DOI: 10.3389/fimmu.2020.581385] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease including the cardiovascular system. Atherosclerosis is the most common cardiovascular complication of SLE and a significant risk factor for morbidity and mortality. Vascular damage/protection mechanism in SLE patients is out of balance, caused by the cascade reaction among oxidative stress, proinflammatory cytokines, Neutrophil Extracellular Traps, activation of B cells and autoantibodies and abnormal T cells. As a precursor cell repairing vascular endothelium, endothelial progenitor cells (EPCs) belong to the protective mechanism and show the reduced number and impaired function in SLE. However, the pathological mechanism of EPCs dysfunction in SLE remains ill-defined. This paper reviews the latest SLE epidemiology and pathogenesis, discusses the changes in the number and function of EPCs in SLE, expounds the role of EPCs in SLE atherosclerosis, and provides new guidance and theoretical basis for exploring novel targets for SLE treatment.
Collapse
Affiliation(s)
- Xuewei Ding
- Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wei Xiang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, NHC Key Laboratory of Control of Tropical diseases (Hainan Medical University), Haikou, China
| | - Xiaojie He
- Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
27
|
Behrouzi B, Araujo Campoverde MV, Liang K, Talbot HK, Bogoch II, McGeer A, Fröbert O, Loeb M, Vardeny O, Solomon SD, Udell JA. Influenza Vaccination to Reduce Cardiovascular Morbidity and Mortality in Patients With COVID-19: JACC State-of-the-Art Review. J Am Coll Cardiol 2020; 76:1777-1794. [PMID: 33032740 PMCID: PMC7535809 DOI: 10.1016/j.jacc.2020.08.028] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/24/2020] [Accepted: 08/07/2020] [Indexed: 02/06/2023]
Abstract
Viral respiratory infections are risk factors for cardiovascular disease (CVD). Underlying CVD is also associated with an increased risk of complications following viral respiratory infections, including increased morbidity, mortality, and health care utilization. Globally, these phenomena are observed with seasonal influenza and with the current coronavirus disease 2019 (COVID-19) pandemic. Persons with CVD represent an important target population for respiratory virus vaccines, with capacity developed within 3 large ongoing influenza vaccine cardiovascular outcomes trials to determine the potential cardioprotective effects of influenza vaccines. In the context of COVID-19, these international trial networks may be uniquely positioned to redeploy infrastructure to study therapies for primary and secondary prevention of COVID-19. Here, we describe mechanistic links between influenza and COVID-19 infection and the risk of acute cardiovascular events, summarize the data to date on the potential cardioprotective effects of influenza vaccines, and describe the ongoing influenza vaccine cardiovascular outcomes trials, highlighting important lessons learned that are applicable to COVID-19.
Collapse
Affiliation(s)
- Bahar Behrouzi
- Cardiovascular Division, Department of Medicine, Women's College Hospital, Toronto, Ontario, Canada; Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Maria Viviana Araujo Campoverde
- Cardiovascular Division, Department of Medicine, Women's College Hospital, Toronto, Ontario, Canada; Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Kyle Liang
- Women's College Hospital Institute for Health System Solutions and Virtual Care (WIHV), Women's College Hospital, Toronto, Ontario, Canada
| | - H Keipp Talbot
- Departments of Medicine and Health Policy, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Isaac I Bogoch
- Divisions of General Internal Medicine and Infectious Diseases, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Allison McGeer
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada; Division of Microbiology, Sinai Health System, Toronto, Ontario, Canada
| | - Ole Fröbert
- Department of Cardiology, Faculty of Health, Örebro University, Örebro, Sweden
| | - Mark Loeb
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Orly Vardeny
- Center for Care Delivery and Outcomes Research, Minneapolis Veteran Affairs Health Care System, Minneapolis, Minnesota
| | - Scott D Solomon
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard University, Boston, Massachusetts
| | - Jacob A Udell
- Cardiovascular Division, Department of Medicine, Women's College Hospital, Toronto, Ontario, Canada; Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada; Peter Munk Cardiac Centre, Toronto General Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
28
|
Li X, Zong L, Zhu Y, Li Y, Zhou Y, Zhou H. Penicillium janthinellum Pneumonia in an SLE Patient: A Case Study. Infect Drug Resist 2020; 13:2745-2749. [PMID: 32821135 PMCID: PMC7423344 DOI: 10.2147/idr.s255968] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/17/2020] [Indexed: 11/23/2022] Open
Abstract
The risk of opportunistic fungal infections is high in immunocompromised patients. The Penicillium genus is common and diverse in nature. However, it rarely causes infection in humans. Here, we reported a case of Penicillium janthinellum pneumonia in a systemic lupus erythematosus (SLE) patient, and the morphological characteristics of P. janthinellum were also described. The patient was a 64-year-old female. She had been diagnosed with SLE and membranous lupus nephritis 10 months previously. Her medications included methylprednisolone, cyclosporine, and hydroxychloroquine. She was admitted because of fever and diagnosed with pneumonia. P. janthinellum was isolated from sputum and bronchoalveolar lavage (BAL) samples. BAL fluid stained with multiple stains showed the presence of somewhat dichotomously branching septate fungal hyphae. P. janthinellum was identified, and its morphological features were described. Antibiotic susceptibility profiles showed that this strain had higher minimum inhibitory concentration (MIC) values in response to multiple antifungal drugs. The patient died 10 days after diagnosis. To the best of our knowledge, this report is the second to demonstrate that P. janthinellum causes infection and is the first to present an infection (pneumonia) caused by P. janthinellumi in an SLE patient. Clinical and laboratory personnel should be aware that the Penicillium genus also contains pathogenic bacteria that cannot simply be treated as contaminants, especially in immunosuppressed patients.
Collapse
Affiliation(s)
- Xi Li
- Centre of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, People's Republic of China
| | - Laibin Zong
- Centre of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, People's Republic of China
| | - Yongze Zhu
- Centre of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, People's Republic of China
| | - Yali Li
- Department of Dermatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, People's Republic of China
| | - Yonglie Zhou
- Centre of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, People's Republic of China
| | - Hua Zhou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, People's Republic of China
| |
Collapse
|
29
|
Fülöp T, Munawara U, Larbi A, Desroches M, Rodrigues S, Catanzaro M, Guidolin A, Khalil A, Bernier F, Barron AE, Hirokawa K, Beauregard PB, Dumoulin D, Bellenger JP, Witkowski JM, Frost E. Targeting Infectious Agents as a Therapeutic Strategy in Alzheimer's Disease. CNS Drugs 2020; 34:673-695. [PMID: 32458360 PMCID: PMC9020372 DOI: 10.1007/s40263-020-00737-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent dementia in the world. Its cause(s) are presently largely unknown. The most common explanation for AD, now, is the amyloid cascade hypothesis, which states that the cause of AD is senile plaque formation by the amyloid β peptide, and the formation of neurofibrillary tangles by hyperphosphorylated tau. A second, burgeoning theory by which to explain AD is based on the infection hypothesis. Much experimental and epidemiological data support the involvement of infections in the development of dementia. According to this mechanism, the infection either directly or via microbial virulence factors precedes the formation of amyloid β plaques. The amyloid β peptide, possessing antimicrobial properties, may be beneficial at an early stage of AD, but becomes detrimental with the progression of the disease, concomitantly with alterations to the innate immune system at both the peripheral and central levels. Infection results in neuroinflammation, leading to, and sustained by, systemic inflammation, causing eventual neurodegeneration, and the senescence of the immune cells. The sources of AD-involved microbes are various body microbiome communities from the gut, mouth, nose, and skin. The infection hypothesis of AD opens a vista to new therapeutic approaches, either by treating the infection itself or modulating the immune system, its senescence, or the body's metabolism, either separately, in parallel, or in a multi-step way.
Collapse
Affiliation(s)
- Tamàs Fülöp
- Geriatric Division, Department of Medicine, Faculty of Medicine and Health Sciences, Research Center on Aging, University of Sherbrooke, 3001, 12th Avenue North, Sherbrooke, QC, J1H 5N4, Canada.
| | - Usma Munawara
- Geriatric Division, Department of Medicine, Faculty of Medicine and Health Sciences, Research Center on Aging, University of Sherbrooke, 3001, 12th Avenue North, Sherbrooke, QC, J1H 5N4, Canada
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, Biopolis, Singapore, Singapore
- Department of Biology, Faculty of Science, University Tunis El Manar, Tunis, Tunisia
| | - Mathieu Desroches
- MathNeuro Team, Inria Sophia Antipolis Méditerranée, Valbonne, France
- Université Côte d'Azur, Nice, France
| | - Serafim Rodrigues
- Ikerbasque, The Basque Foundation for Science, Bilbao, Spain
- BCAM, The Basque Center for Applied Mathematics, Bilbao, Spain
| | - Michele Catanzaro
- Geriatric Division, Department of Medicine, Faculty of Medicine and Health Sciences, Research Center on Aging, University of Sherbrooke, 3001, 12th Avenue North, Sherbrooke, QC, J1H 5N4, Canada
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Andrea Guidolin
- BCAM, The Basque Center for Applied Mathematics, Bilbao, Spain
| | - Abdelouahed Khalil
- Geriatric Division, Department of Medicine, Faculty of Medicine and Health Sciences, Research Center on Aging, University of Sherbrooke, 3001, 12th Avenue North, Sherbrooke, QC, J1H 5N4, Canada
| | - François Bernier
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Japan
| | - Annelise E Barron
- Department of Bioengineering, Stanford School of Medicine, Stanford, CA, USA
| | - Katsuiku Hirokawa
- Department of Pathology, Institute of Health and Life Science, Tokyo and Nito-memory Nakanosogo Hospital, Tokyo Med. Dent. University, Tokyo, Japan
| | - Pascale B Beauregard
- Department of Biology, Faculty of Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | - David Dumoulin
- Department of Biology, Faculty of Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Jean-Philippe Bellenger
- Department of Chemistry, Faculty of Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Jacek M Witkowski
- Department of Pathophysiology, Medical University of Gdansk, Gdansk, Poland
| | - Eric Frost
- Department of Microbiology and Infectious diseases, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
30
|
Lu Y, Jiang H, Li B, Cao L, Shen Q, Yi W, Ju Z, Chen L, Han F, Appelgren D, Segelmark M, de Buhr N, von Köckritz-Blickwede M, Chen J. Telomere dysfunction promotes small vessel vasculitis via the LL37-NETs-dependent mechanism. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:357. [PMID: 32355801 PMCID: PMC7186648 DOI: 10.21037/atm.2020.02.130] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Small vessel vasculitis (SVV) is a group of systemic autoimmune diseases that are mediated by neutrophil extracellular traps (NETs) in response to cathelicidin LL37, an aging molecular marker, which could be induced by telomere dysfunction. Therefore, in this study, we evaluated the hypothesis that telomere dysfunction in neutrophils may promote SVV via an LL37-NETs-dependent mechanism. Methods We contrasted the release of neutrophil NETs from mice with telomere dysfunction, mice with DNA damage and wide-type mice. Neutrophil telomere length, the expression of LL37, and the formation of NETs were measured in SVV patients and healthy controls (HCs). The co-expression of γH2AX, LL37, and NETs were detected in SVV patients to evaluate the association of the immune aging of neutrophils and pro-inflammatory conditions. LL37 inhibitor was used to verify its key role in NETs release in SVV patients and DNA damage mice. Results We found that NETs were over-induced by telomere dysfunction and DNA damage in mice, which may be associated with a marked increase in LL37. For patients with SVV, telomeres in neutrophils were significantly shortened, which was also associated with higher levels of LL37 and NETs. Inhibition of LL37 reduced the NETs released from neutrophils. Conclusions Taken together, the results of these studies suggest that dysfunction of telomeres may promote SVV through the mechanism of LL37-dependent NETs. Thus, targeting the LL37-NETs may be a novel therapy for SVV.
Collapse
Affiliation(s)
- Yingying Lu
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.,Kidney Disease Immunology Laboratory, The Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou 310003, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou 310003, China.,Key Laboratory of Nephropathy, Hangzhou 310003, China
| | - Hong Jiang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.,Kidney Disease Immunology Laboratory, The Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou 310003, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou 310003, China.,Key Laboratory of Nephropathy, Hangzhou 310003, China
| | - Bingjue Li
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.,Kidney Disease Immunology Laboratory, The Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou 310003, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou 310003, China.,Key Laboratory of Nephropathy, Hangzhou 310003, China
| | - Luxi Cao
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.,Kidney Disease Immunology Laboratory, The Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou 310003, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou 310003, China.,Key Laboratory of Nephropathy, Hangzhou 310003, China
| | - Qixia Shen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.,Kidney Disease Immunology Laboratory, The Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou 310003, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou 310003, China.,Key Laboratory of Nephropathy, Hangzhou 310003, China
| | - Weiwei Yi
- Institute of Aging Research and Max-Planck-Research Group on Stem Cell Aging, Hangzhou Normal University, Hangzhou 311121, China
| | - Zhenyu Ju
- Institute of Aging Research and Max-Planck-Research Group on Stem Cell Aging, Hangzhou Normal University, Hangzhou 311121, China
| | - Liangliang Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.,Kidney Disease Immunology Laboratory, The Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou 310003, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou 310003, China.,Key Laboratory of Nephropathy, Hangzhou 310003, China
| | - Fei Han
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.,Kidney Disease Immunology Laboratory, The Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou 310003, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou 310003, China.,Key Laboratory of Nephropathy, Hangzhou 310003, China
| | - Daniel Appelgren
- Department of Medical and Health Sciences (IMH), Linkoping University, Linkoping, Sweden
| | - Mårten Segelmark
- Department of Medical and Health Sciences (IMH), Linkoping University, Linkoping, Sweden
| | - Nicole de Buhr
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Hannover, Germany.,Department of Physiological Chemistry and Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Maren von Köckritz-Blickwede
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Hannover, Germany.,Department of Physiological Chemistry and Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.,Kidney Disease Immunology Laboratory, The Third Grade Laboratory, State Administration of Traditional Chinese Medicine of PR China, Hangzhou 310003, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health, Hangzhou 310003, China.,Key Laboratory of Nephropathy, Hangzhou 310003, China
| |
Collapse
|
31
|
Tsai CY, Hsieh SC, Lu CS, Wu TH, Liao HT, Wu CH, Li KJ, Kuo YM, Lee HT, Shen CY, Yu CL. Cross-Talk between Mitochondrial Dysfunction-Provoked Oxidative Stress and Aberrant Noncoding RNA Expression in the Pathogenesis and Pathophysiology of SLE. Int J Mol Sci 2019; 20:ijms20205183. [PMID: 31635056 PMCID: PMC6829370 DOI: 10.3390/ijms20205183] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/10/2019] [Accepted: 10/14/2019] [Indexed: 12/16/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a prototype of systemic autoimmune disease involving almost every organ. Polygenic predisposition and complicated epigenetic regulations are the upstream factors to elicit its development. Mitochondrial dysfunction-provoked oxidative stress may also play a crucial role in it. Classical epigenetic regulations of gene expression may include DNA methylation/acetylation and histone modification. Recent investigations have revealed that intracellular and extracellular (exosomal) noncoding RNAs (ncRNAs), including microRNAs (miRs), and long noncoding RNAs (lncRNAs), are the key molecules for post-transcriptional regulation of messenger (m)RNA expression. Oxidative and nitrosative stresses originating from mitochondrial dysfunctions could become the pathological biosignatures for increased cell apoptosis/necrosis, nonhyperglycemic metabolic syndrome, multiple neoantigen formation, and immune dysregulation in patients with SLE. Recently, many authors noted that the cross-talk between oxidative stress and ncRNAs can trigger and perpetuate autoimmune reactions in patients with SLE. Intracellular interactions between miR and lncRNAs as well as extracellular exosomal ncRNA communication to and fro between remote cells/tissues via plasma or other body fluids also occur in the body. The urinary exosomal ncRNAs can now represent biosignatures for lupus nephritis. Herein, we’ll briefly review and discuss the cross-talk between excessive oxidative/nitrosative stress induced by mitochondrial dysfunction in tissues/cells and ncRNAs, as well as the prospect of antioxidant therapy in patients with SLE.
Collapse
Affiliation(s)
- Chang-Youh Tsai
- Division of Allergy, Immunology & Rheumatology, Taipei Veterans General Hospital & National Yang-Ming University, #201 Sec.2, Shih-Pai Road, Taipei 11217, Taiwan.
| | - Song-Chou Hsieh
- Department of Internal Medicine, National Taiwan University Hospital, #7 Chung-Shan South Road, Taipei 10002, Taiwan.
| | - Cheng-Shiun Lu
- Department of Internal Medicine, National Taiwan University Hospital, #7 Chung-Shan South Road, Taipei 10002, Taiwan.
- Institute of Clinical Medicine, National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan.
| | - Tsai-Hung Wu
- Division of Nephrology, Taipei Veterans General Hospital & National Yang-Ming University, #201 Sec. 2, Shih-Pai Road, Taipei 11217, Taiwan.
| | - Hsien-Tzung Liao
- Division of Allergy, Immunology & Rheumatology, Taipei Veterans General Hospital & National Yang-Ming University, #201 Sec.2, Shih-Pai Road, Taipei 11217, Taiwan.
| | - Cheng-Han Wu
- Department of Internal Medicine, National Taiwan University Hospital, #7 Chung-Shan South Road, Taipei 10002, Taiwan.
- Institute of Clinical Medicine, National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan.
| | - Ko-Jen Li
- Department of Internal Medicine, National Taiwan University Hospital, #7 Chung-Shan South Road, Taipei 10002, Taiwan.
| | - Yu-Min Kuo
- Department of Internal Medicine, National Taiwan University Hospital, #7 Chung-Shan South Road, Taipei 10002, Taiwan.
- Institute of Clinical Medicine, National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan.
| | - Hui-Ting Lee
- Section of Allergy, Immunology & Rheumatology, Mackay Memorial Hospital, #92 Sec. 2, Chung-Shan North Road, Taipei 10449, Taiwan.
| | - Chieh-Yu Shen
- Department of Internal Medicine, National Taiwan University Hospital, #7 Chung-Shan South Road, Taipei 10002, Taiwan.
- Institute of Clinical Medicine, National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan.
| | - Chia-Li Yu
- Department of Internal Medicine, National Taiwan University Hospital, #7 Chung-Shan South Road, Taipei 10002, Taiwan.
| |
Collapse
|