1
|
Duan L, Yao Y, Kong H, Zhou Y, Cui D. Chemokines and chemokine receptors: Potential therapeutic targets in systemic lupus erythematosus. Cytokine 2024; 184:156770. [PMID: 39326198 DOI: 10.1016/j.cyto.2024.156770] [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: 06/27/2024] [Revised: 09/07/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that affects connective tissue and can lead to multisystem organ damage. Chemokines are a class of small proteins that interact with receptors and participate in a variety of physiological functions, including cell growth, differentiation, apoptosis and distribution. They also play important roles in pathological processes, such as the inflammatory response, wound repair, tumor formation and metastasis. Previous studies have shown that the levels of chemokines and their receptors are elevated in the blood and inflamed tissues of SLE patients. In addition, chemokine ligand-receptor interactions control the recruitment of leukocytes into tissues, suggesting that chemokines and their receptors may be biomarkers and therapeutic targets for SLE. This review summarizes the causative role of chemokines and their receptors in SLE, as well as their clinical values and challenges as potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Lishuang Duan
- Department of Anesthesia, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.
| | - Yongxing Yao
- Department of Anesthesia, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.
| | - Haiying Kong
- Department of Anesthesia, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.
| | - Yanfeng Zhou
- Department of Anesthesia, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.
| | - Dawei Cui
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.
| |
Collapse
|
2
|
Atzeni F, Rodríguez-Pintó I, Cervera R. Cardiovascular disease risk in systemic lupus erythematous: Certainties and controversies. Autoimmun Rev 2024; 23:103646. [PMID: 39321952 DOI: 10.1016/j.autrev.2024.103646] [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: 06/09/2024] [Revised: 09/19/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024]
Abstract
Patients with systemic lupus erythematosus (SLE) experience greater cardiovascular morbidity and mortality compared to the general population. It is known that endothelial dysfunction, an early indicator of atherosclerosis development, can arise even without the presence of conventional cardiovascular risk factors. In fact, the risk factors contributing to cardiovascular disease can be classified into traditional risk factors and those uniquely associated with SLE such as disease activity, autoantibodies, etc.Furthermore, the pathogenesis of cardiovascular disease in SLE is linked to the activation of both the innate and adaptive immune systems. Given these findings, it is essential for clinicians to acknowledge the heightened CVD risk in SLE patients, perform comprehensive screenings for cardiovascular risk factors, and implement aggressive treatment strategies for those who exhibit signs of clinical CVD. The aim of this review is to summarize the findings on cardiovascular disease in SLE and to examine potential screening and therapeutic strategies for clinical practice.
Collapse
Affiliation(s)
- Fabiola Atzeni
- Rheumatology Unit, Department of Experimental and Internal Medicine, University of Messina, Messina, Italy
| | - Ignasi Rodríguez-Pintó
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases (UEC/CSUR) of the Catalan and Spanish Health Systems-Member of ERNReCONNET, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.
| | - Ricard Cervera
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases (UEC/CSUR) of the Catalan and Spanish Health Systems-Member of ERNReCONNET, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.
| |
Collapse
|
3
|
Zhang S, Xu R, Kang L. Biomarkers for systemic lupus erythematosus: A scoping review. Immun Inflamm Dis 2024; 12:e70022. [PMID: 39364719 PMCID: PMC11450456 DOI: 10.1002/iid3.70022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 08/31/2024] [Accepted: 09/06/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND In recent years, newly discovered potential biomarkers have great research potential in the diagnosis, disease activity prediction, and treatment of systemic lupus erythematosus (SLE). OBJECTIVE In this study, a scoping review of potential biomarkers for SLE over several years has identified the extent to which studies on biomarkers for SLE have been conducted, the specificity, sensitivity, and diagnostic value of potential biomarkers of SLE, the research potential of these biomarkers in disease diagnosis, and activity detection is discussed. METHODS In PubMed and Google Scholar databases, "SLE," "biomarkers," "predictor," "autoimmune diseases," "lupus nephritis," "neuropsychiatric SLE," "diagnosis," "monitoring," and "disease activity" were used as keywords to systematically search for SLE molecular biomarkers published from 2020 to 2024. Analyze and summarize the literature that can guide the article. CONCLUSIONS Recent findings suggest that some potential biomarkers may have clinical application prospects. However, to date, many of these biomarkers have not been subjected to repeated clinical validation. And no single biomarker has sufficient sensitivity and specificity for SLE. It is not scientific to choose only one or several biomarkers to judge the complex disease of SLE. It may be a good direction to carry out a meta-analysis of various biomarkers to find SLE biomarkers suitable for clinical use, or to evaluate SLE by combining multiple biomarkers through mathematical models. At the same time, advanced computational methods are needed to analyze large data sets and discover new biomarkers, and strive to find biomarkers that are sensitive and specific enough to SLE and can be used in clinical practice, rather than only staying in experimental research and data analysis.
Collapse
Affiliation(s)
- Su‐jie Zhang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, Key Laboratory of High Altitude Environment and Genes Related to Diseases of Tibet Autonomous RegionSchool of Medicine, Xizang Minzu UniversityXianyangShaanxiChina
| | - Rui‐yang Xu
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, Key Laboratory of High Altitude Environment and Genes Related to Diseases of Tibet Autonomous RegionSchool of Medicine, Xizang Minzu UniversityXianyangShaanxiChina
| | - Long‐li Kang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, Key Laboratory of High Altitude Environment and Genes Related to Diseases of Tibet Autonomous RegionSchool of Medicine, Xizang Minzu UniversityXianyangShaanxiChina
| |
Collapse
|
4
|
Sacre K, Vinet E, Pineau CA, Mendel A, Kalache F, Grenier LP, Huynh T, Bernatsky S. N-terminal pro-brain natriuretic peptide is a biomarker for cardiovascular damage in systemic lupus erythematous: a cross-sectional study. Rheumatology (Oxford) 2024; 63:1739-1745. [PMID: 37802912 DOI: 10.1093/rheumatology/kead522] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/05/2023] [Accepted: 09/11/2023] [Indexed: 10/08/2023] Open
Abstract
OBJECTIVES Prediction models based on traditional risk factors underestimate cardiovascular (CV) risk in systemic lupus erythematosus (SLE). In a large sample of unselected SLE patients, we investigated cross-sectional associations of NT-proBNP with cardiovascular damage (CVD). METHODS Serum NT-proBNP was measured in SLE patients enrolled in the MUHC Lupus Clinic registry. Serum was collected between March 2022 and April 2023 at annual research visits. The primary outcome was CVD identified on the SLICC Damage Index. Factors associated with CVD and NT-proBNP levels were determined. RESULTS Overall, 270 SLE patients [female 91%, median age 50.7 (first quartile to third quartile: 39.6-62.1) years] were analysed for the primary outcome. Among them, 33 (12%) had CVD. The ROC curve for NT-proBNP demonstrated strong associations with CVD (AUC 0.78, 95% CI 0.69-0.87) with a threshold of 133 pg/ml providing the best discrimination for those with/without CVD. Hypertension (OR 3.3, 95% CI 1.2-9.0), dyslipidaemia (OR 3.6, 95% CI 1.3-9.6) and NT-proBNP >133 pg/ml (OR 7.0, 95% CI, 2.6-19.1) were associated with CVD in the multivariable logistic regression model. Increased NT-proBNP levels were associated with age (OR 4.2, 95% CI 2.2-8.3), ever smoking (OR 1.9, 95% CI 1.0-3.5), reduced eGFR (4.1, 95% CI 1.3-13.1), prior pericarditis/pleuritis (OR 2.5, 95% CI 1.4-4.5) and aPL antibodies (OR 2.6, 95% CI 1.4-4.9). CONCLUSION NT-proBNP is a biomarker for CV damage in SLE. The novel associations of NT-proBNP levels with prior pericarditis/pleuritis and aPL antibodies suggest new avenues for research to better understand what drives CV risk in SLE.
Collapse
Affiliation(s)
- Karim Sacre
- Division of Clinical Epidemiology, McGill University Health Centre, Montreal, QC, Canada
- Departement de Médecine Interne, Université Paris-Cité, Assistance Publique Hopitaux de Paris, Hopital Bichat, Paris, France
| | - Evelyne Vinet
- Division of Clinical Epidemiology, McGill University Health Centre, Montreal, QC, Canada
- Division of Rheumatology, McGill University Health Centre, Montreal, QC, Canada
| | - Christian A Pineau
- Division of Rheumatology, McGill University Health Centre, Montreal, QC, Canada
| | - Arielle Mendel
- Division of Rheumatology, McGill University Health Centre, Montreal, QC, Canada
| | - Fares Kalache
- Division of Rheumatology, McGill University Health Centre, Montreal, QC, Canada
| | | | - Thao Huynh
- Division of Cardiology, McGill University Health Centre, Montreal, QC, Canada
| | - Sasha Bernatsky
- Division of Clinical Epidemiology, McGill University Health Centre, Montreal, QC, Canada
- Division of Rheumatology, McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
5
|
Ah Kioon MD, Laurent P, Chaudhary V, Du Y, Crow MK, Barrat FJ. Modulation of plasmacytoid dendritic cells response in inflammation and autoimmunity. Immunol Rev 2024; 323:241-256. [PMID: 38553621 DOI: 10.1111/imr.13331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
The discovery of toll-like receptors (TLRs) and the subsequent recognition that endogenous nucleic acids (NAs) could serve as TLR ligands have led to essential insights into mechanisms of healthy immune responses as well as pathogenic mechanisms relevant to systemic autoimmune and inflammatory diseases. In systemic lupus erythematosus, systemic sclerosis, and rheumatoid arthritis, NA-containing immune complexes serve as TLR ligands, with distinct implications depending on the additional immune stimuli available. Plasmacytoid dendritic cells (pDCs), the robust producers of type I interferon (IFN-I), are providing critical insights relevant to TLR-mediated healthy immune responses and tissue repair, as well as generation of inflammation, autoimmunity and fibrosis, processes central to the pathogenesis of many autoimmune diseases. In this review, we describe recent data characterizing the role of platelets and NA-binding chemokines in modulation of TLR signaling in pDCs, as well as implications for how the IFN-I products of pDCs contribute to the generation of inflammation and wound healing responses by monocyte/macrophages. Chemokine modulators of TLR-mediated B cell tolerance mechanisms and interactions between TLR signaling and metabolic pathways are also considered. The modulators of TLR signaling and their contribution to the pathogenesis of systemic autoimmune diseases suggest new opportunities for identification of novel therapeutic targets.
Collapse
Affiliation(s)
| | - Paôline Laurent
- HSS Research Institute, Hospital for Special Surgery, New York, New York, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, New York, USA
| | - Vidyanath Chaudhary
- HSS Research Institute, Hospital for Special Surgery, New York, New York, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, New York, USA
| | - Yong Du
- HSS Research Institute, Hospital for Special Surgery, New York, New York, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, New York, USA
| | - Mary K Crow
- HSS Research Institute, Hospital for Special Surgery, New York, New York, USA
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery, New York, New York, USA
- Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Franck J Barrat
- HSS Research Institute, Hospital for Special Surgery, New York, New York, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, New York, USA
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| |
Collapse
|
6
|
Accapezzato D, Caccavale R, Paroli MP, Gioia C, Nguyen BL, Spadea L, Paroli M. Advances in the Pathogenesis and Treatment of Systemic Lupus Erythematosus. Int J Mol Sci 2023; 24:6578. [PMID: 37047548 PMCID: PMC10095030 DOI: 10.3390/ijms24076578] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/05/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a genetically predisposed, female-predominant disease, characterized by multiple organ damage, that in its most severe forms can be life-threatening. The pathogenesis of SLE is complex and involves cells of both innate and adaptive immunity. The distinguishing feature of SLE is the production of autoantibodies, with the formation of immune complexes that precipitate at the vascular level, causing organ damage. Although progress in understanding the pathogenesis of SLE has been slower than in other rheumatic diseases, new knowledge has recently led to the development of effective targeted therapies, that hold out hope for personalized therapy. However, the new drugs available to date are still an adjunct to conventional therapy, which is known to be toxic in the short and long term. The purpose of this review is to summarize recent advances in understanding the pathogenesis of the disease and discuss the results obtained from the use of new targeted drugs, with a look at future therapies that may be used in the absence of the current standard of care or may even cure this serious systemic autoimmune disease.
Collapse
Affiliation(s)
- Daniele Accapezzato
- Division of Clinical Immunology, Department of Clinical, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Rosalba Caccavale
- Division of Clinical Immunology, Department of Clinical, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Maria Pia Paroli
- Eye Clinic, Department of Sense Organs, Sapienza University of Rome, 00185 Rome, Italy
| | - Chiara Gioia
- Division of Clinical Immunology, Department of Clinical, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Bich Lien Nguyen
- Division of Clinical Immunology, Department of Clinical, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Luca Spadea
- Post Graduate School of Public Health, University of Siena, 53100 Siena, Italy
| | - Marino Paroli
- Division of Clinical Immunology, Department of Clinical, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
7
|
Wang G, Sun Y, Jiang Y, Li S, Liu Y, Yuan Y, Nie H. CXCR3 deficiency decreases autoantibody production by inhibiting aberrant activated T follicular helper cells and B cells in lupus mice. Mol Immunol 2023; 156:39-47. [PMID: 36889185 DOI: 10.1016/j.molimm.2023.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/26/2023] [Accepted: 02/16/2023] [Indexed: 03/08/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by a high level of autoantibody production. T follicular helper (Tfh) cells and B cells participate in the development of SLE. Several studies have shown that CXCR3+ cells are increased in SLE patients. However, the mechanism through which CXCR3 influences lupus development remains unclear. In this study, we established lupus models to determine the role of CXCR3 in lupus pathogenesis. The concentration of autoantibodies was detected using the enzyme-linked immunosorbent assay (ELISA), and the percentages of Tfh cells and B cells were measured using flow cytometry. RNA sequencing (RNA-seq) was performed to detect the differentially expressed genes in CD4+ T cells from wild-type (WT) and CXCR3 knock-out (KO) lupus mice. Migration of CD4+ T cells in spleen section was assessed using immunofluorescence. CD4+ T cell function in helping B cells produce antibodies was determined using a co-culture experiment and supernatant IgG ELISA. Lupus mice were treated with a CXCR3 antagonist to confirm the therapeutic effects. We found that the expression of CXCR3 was increased in CD4+ T cells from lupus mice. CXCR3 deficiency reduced autoantibody production with decreased proportions of Tfh cells, germinal center (GC) B cells, and plasma cells. Expression of Tfh-related genes was downregulated in CD4+ T cells from CXCR3 KO lupus mice. Migration to B cell follicles and T-helper function of CD4+ T cells were reduced in CXCR3 KO lupus mice. CXCR3 antagonist AMG487 decreased the level of serum anti-dsDNA IgG in lupus mice. We clarify that CXCR3 may play an important role in autoantibody production by increasing the percentages of aberrant activated Tfh cells and B cells and promoting the migration and T-helper function of CD4+ T cells in lupus mice. Thus, CXCR3 may be a potential target for lupus therapy.
Collapse
Affiliation(s)
- Guojue Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Sun
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongshuai Jiang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengzhe Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunhui Liu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanyang Yuan
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Hong Nie
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
8
|
David C, Costedoat-Chalumeau N, Belhadi D, Laouénan C, Boutten A, Chezel J, Rouzaud D, Dehoux M, Guern VL, Mathian A, Chaves SDA, Duhaut P, Fain O, Galicier L, Ghillani-Dalbin P, Kahn JE, Morel N, Perard L, Pha M, Sarrot-Reynauld F, Aumaitre O, Chasset F, Limal N, Desmurs-Clavel H, Ackermann F, Amoura Z, Papo T, Sacre K. Soluble CD163 and incident cardiovascular events in patients with systemic lupus erythematosus: An observational cohort study. J Intern Med 2022; 292:536-539. [PMID: 35373869 PMCID: PMC9542031 DOI: 10.1111/joim.13490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Clémence David
- Département de Médecine Interne, Hôpital Bichat, Assistance Publique Hôpitaux de Paris (APHP), Institut national de la santé et de la recherche médicale (INSERM) U1149, Université de Paris, Paris, France
| | - Nathalie Costedoat-Chalumeau
- APHP, Hôpital Cochin, Département de Médecine Interne, Centre de Reference Maladies Auto-immunes et Systémiques Rares, Université de Paris, CRESS, INSERM, INRA, Paris, France
| | - Drifa Belhadi
- Departement d'Epidémiologie et de Recherche Clinique, Hôpital Bichat, APHP, Université de Paris, Paris, France
| | - Cedric Laouénan
- Departement d'Epidémiologie et de Recherche Clinique, Hôpital Bichat, APHP, Université de Paris, Paris, France
| | - Anne Boutten
- Département de Biochimie, Hôpital Bichat, APHP, Université de Paris, Paris, France
| | - Julie Chezel
- Département de Médecine Interne, Hôpital Bichat, Assistance Publique Hôpitaux de Paris (APHP), Institut national de la santé et de la recherche médicale (INSERM) U1149, Université de Paris, Paris, France
| | - Diane Rouzaud
- Département de Médecine Interne, Hôpital Bichat, Assistance Publique Hôpitaux de Paris (APHP), Institut national de la santé et de la recherche médicale (INSERM) U1149, Université de Paris, Paris, France
| | - Monique Dehoux
- Département de Biochimie, Hôpital Bichat, APHP, Université de Paris, Paris, France
| | - Véronique Le Guern
- APHP, Hôpital Cochin, Département de Médecine Interne, Centre de Reference Maladies Auto-immunes et Systémiques Rares, Université de Paris, CRESS, INSERM, INRA, Paris, France
| | - Alexis Mathian
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Groupement Hospitalier Pitié-Salpêtrière, French National Referral Center for Systemic Lupus Erythematosus, Antiphospholipid Antibody Syndrome and Other Autoimmune Disorders, Service de Médecine Interne 2, Paris, France
| | - Sébastien de Almeida Chaves
- Département de Médecine Interne, Hôpital Purpan, Centre Hospitalo-Universitaire (CHU) de Toulouse, Toulouse, France
| | - Pierre Duhaut
- Département de Médecine Interne, Hôpital Amiens Nord CHU d'Amiens, Amiens, France
| | - Olivier Fain
- Département de Médecine Interne, Hôpital Saint Antoine, APHP, Université Pierre et Marie Curie, Paris, France
| | - Lionel Galicier
- Département d'Immunologie Clinique, Hôpital Saint Louis, APHP, Université de Paris, Département de Médecine Interne, Hôpital Saint Joseph, Marseille, France
| | - Pascale Ghillani-Dalbin
- Département de Immunologie, Hôpital Pitié-Salpétrière, APHP, Université Pierre et Marie Curie, Paris, France
| | - Jean Emmanuel Kahn
- Département de Médecine Interne, Hôpital Ambroise Paré, APHP, Université de Versailles-Saint-Quentin en Yvelines, France
| | - Nathalie Morel
- APHP, Hôpital Cochin, Département de Médecine Interne, Centre de Reference Maladies Auto-immunes et Systémiques Rares, Université de Paris, CRESS, INSERM, INRA, Paris, France
| | - Laurent Perard
- Département de Médecine Interne, Hôpital St Joseph St Luc, Lyon, France
| | - Micheline Pha
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Groupement Hospitalier Pitié-Salpêtrière, French National Referral Center for Systemic Lupus Erythematosus, Antiphospholipid Antibody Syndrome and Other Autoimmune Disorders, Service de Médecine Interne 2, Paris, France
| | | | - Olivier Aumaitre
- Département de médecine interne, Hôpital Gabriel-Montpied, CHU de Clermont-Ferrand, France
| | - François Chasset
- Département de médecine interne, Hôpital Tenon, APHP Université Pierre et Marie Curie, Paris, France
| | - Nicolas Limal
- Département de médecine interne, Hôpital Henri Mondor, APHP Université Paris-Est Créteil, Paris, France
| | - Helene Desmurs-Clavel
- Département de médecine interne, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Felix Ackermann
- Département de médecine interne, Hôpital Foch, Suresnes, France
| | - Zahir Amoura
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Groupement Hospitalier Pitié-Salpêtrière, French National Referral Center for Systemic Lupus Erythematosus, Antiphospholipid Antibody Syndrome and Other Autoimmune Disorders, Service de Médecine Interne 2, Paris, France
| | - Thomas Papo
- Département de Médecine Interne, Hôpital Bichat, Assistance Publique Hôpitaux de Paris (APHP), Institut national de la santé et de la recherche médicale (INSERM) U1149, Université de Paris, Paris, France
| | - Karim Sacre
- Département de Médecine Interne, Hôpital Bichat, Assistance Publique Hôpitaux de Paris (APHP), Institut national de la santé et de la recherche médicale (INSERM) U1149, Université de Paris, Paris, France
| |
Collapse
|
9
|
Liu Y, Yu X, Zhang W, Zhang X, Wang M, Ji F. Mechanistic insight into premature atherosclerosis and cardiovascular complications in systemic lupus erythematosus. J Autoimmun 2022; 132:102863. [PMID: 35853760 DOI: 10.1016/j.jaut.2022.102863] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/02/2022] [Indexed: 12/12/2022]
Abstract
Systemic lupus erythematosus (SLE) is associated with a significant risk of cardiovascular disease (CVD), which substantially increases disease mortality and morbidity. The overall mechanisms associated with the development of premature atherosclerosis and CVD in SLE remain unclear, but has been considered as a result of an intricate interplay between the profound immune dysregulation and traditional CVD risk factors. Aberrant systemic inflammation in SLE may lead to an abnormal lipid profile and dysfunction, which can further fuel the pro-atherosclerotic environment. The existence of a strong imbalance between endothelial damage and vascular repair/angiogenesis promotes vascular injury, which is the early step in the progression of atherosclerotic CVD. Profound innate and adaptive immune dysregulation, characterized by excessive type I interferon burden, aberrant macrophage, platelet and complements activation, neutrophil dysregulation and neutrophil extracellular traps formation, uncontrolled T cell activation, and excessive autoantibody production and immune complex formation, have been proposed to promote accelerated CVD in SLE. While designing targeted therapies to correct the dysregulated immune activation may be beneficial in the treatment of SLE-related CVD, much additional work is needed to determine how to translate these findings into clinical practice. Additionally, a number of biomarkers display diagnostic potentials in improving CVD risk stratification in SLE, further prospective studies will help understand which biomarker(s) will be the most impactful one(s) in assessing SLE-linked CVD. Continued efforts to identify novel mechanisms and to establish criteria for assessing CVD risk as well as predicting CVD progression are in great need to improve CVD outcomes in SLE.
Collapse
Affiliation(s)
- Yudong Liu
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, PR China; The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, PR China
| | - Xue Yu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, PR China
| | - Wenduo Zhang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, PR China
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, PR China
| | - Min Wang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, PR China
| | - Fusui Ji
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, PR China.
| |
Collapse
|
10
|
Sierra-Galan LM, Bhatia M, Alberto-Delgado AL, Madrazo-Shiordia J, Salcido C, Santoyo B, Martinez E, Soto ME. Cardiac Magnetic Resonance in Rheumatology to Detect Cardiac Involvement Since Early and Pre-clinical Stages of the Autoimmune Diseases: A Narrative Review. Front Cardiovasc Med 2022; 9:870200. [PMID: 35911548 PMCID: PMC9326004 DOI: 10.3389/fcvm.2022.870200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Autoimmune diseases (ADs) encompass multisystem disorders, and cardiovascular involvement is a well-known feature of autoimmune and inflammatory rheumatic conditions. Unfortunately, subclinical and early cardiovascular involvement remains clinically silent and often undetected, despite its well-documented impact on patient management and prognostication with an even more significant effect on severe and future MACE events as the disease progresses. Cardiac magnetic resonance imaging (MRI), today, commands a unique position of supremacy versus its competition in cardiac assessment and is the gold standard for the non-invasive evaluation of cardiac function, structure, morphology, tissue characterization, and flow with the capability of evaluating biventricular function; myocardium for edema, ischemia, fibrosis, infarction; valves for thickening, large masses; pericardial inflammation, pericardial effusions, and tamponade; cardiac cavities for thrombosis; conduction related abnormalities and features of microvascular and large vessel involvement. As precise and early detection of cardiovascular involvement plays a critical role in improving the outcome of rheumatic and autoimmune conditions, our review aims to highlight the evolving role of CMR in systemic lupus erythematosus (SLE), antiphospholipid syndrome (APS), rheumatoid arthritis (RA), systemic sclerosis (SSc), limited sclerosis (LSc), adult-onset Still's disease (AOSD), polymyositis (PM), dermatomyositis (DM), eosinophilic granulomatosis with polyangiitis (EGPA) (formerly Churg-Strauss syndrome), and DRESS syndrome (DS). It draws attention to the need for concerted, systematic global interdisciplinary research to improve future outcomes in autoimmune-related rheumatic conditions with multiorgan, multisystem, and cardiovascular involvement.
Collapse
Affiliation(s)
- Lilia M. Sierra-Galan
- Cardiology Department of the Cardiovascular Division of the American British Cowdray Medical Center, Mexico City, Mexico
| | - Mona Bhatia
- Department of Imaging, Fortis Escorts Heart Institute, New Delhi, India
| | | | - Javier Madrazo-Shiordia
- Cardiology Department of the Cardiovascular Division of the American British Cowdray Medical Center, Mexico City, Mexico
| | - Carlos Salcido
- Cardiology Department of the Cardiovascular Division of the American British Cowdray Medical Center, Mexico City, Mexico
| | - Bernardo Santoyo
- Cardiology Department of the Cardiovascular Division of the American British Cowdray Medical Center, Mexico City, Mexico
| | - Eduardo Martinez
- Cardiology Department of the Cardiovascular Division of the American British Cowdray Medical Center, Mexico City, Mexico
| | - Maria Elena Soto
- Cardiology Department of the Cardiovascular Division of the American British Cowdray Medical Center, Mexico City, Mexico
- Immunology Department of the National Institute of Cardiology, “Ignacio Chavez”, Mexico City, Mexico
| |
Collapse
|
11
|
Dendritic cells in systemic lupus erythematosus: From pathogenesis to therapeutic applications. J Autoimmun 2022; 132:102856. [DOI: 10.1016/j.jaut.2022.102856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 06/15/2022] [Indexed: 11/18/2022]
|
12
|
Han Z, Ma K, Tao H, Liu H, Zhang J, Sai X, Li Y, Chi M, Nian Q, Song L, Liu C. A Deep Insight Into Regulatory T Cell Metabolism in Renal Disease: Facts and Perspectives. Front Immunol 2022; 13:826732. [PMID: 35251009 PMCID: PMC8892604 DOI: 10.3389/fimmu.2022.826732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/24/2022] [Indexed: 11/29/2022] Open
Abstract
Kidney disease encompasses a complex set of diseases that can aggravate or start systemic pathophysiological processes through their complex metabolic mechanisms and effects on body homoeostasis. The prevalence of kidney disease has increased dramatically over the last two decades. CD4+CD25+ regulatory T (Treg) cells that express the transcription factor forkhead box protein 3 (Foxp3) are critical for maintaining immune homeostasis and preventing autoimmune disease and tissue damage caused by excessive or unnecessary immune activation, including autoimmune kidney diseases. Recent studies have highlighted the critical role of metabolic reprogramming in controlling the plasticity, stability, and function of Treg cells. They are also likely to play a vital role in limiting kidney transplant rejection and potentially promoting transplant tolerance. Metabolic pathways, such as mitochondrial function, glycolysis, lipid synthesis, glutaminolysis, and mammalian target of rapamycin (mTOR) activation, are involved in the development of renal diseases by modulating the function and proliferation of Treg cells. Targeting metabolic pathways to alter Treg cells can offer a promising method for renal disease therapy. In this review, we provide a new perspective on the role of Treg cell metabolism in renal diseases by presenting the renal microenvironment、relevant metabolites of Treg cell metabolism, and the role of Treg cell metabolism in various kidney diseases.
Collapse
Affiliation(s)
- Zhongyu Han
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China.,Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kuai Ma
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hongxia Tao
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hongli Liu
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiong Zhang
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Xiyalatu Sai
- Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao, China
| | - Yunlong Li
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mingxuan Chi
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Qing Nian
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China.,Department of Blood Transfusion Sicuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Linjiang Song
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chi Liu
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| |
Collapse
|
13
|
Oliveira CB, Kaplan MJ. Cardiovascular disease risk and pathogenesis in systemic lupus erythematosus. Semin Immunopathol 2022; 44:309-324. [PMID: 35355124 PMCID: PMC9064999 DOI: 10.1007/s00281-022-00922-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/08/2022] [Indexed: 02/06/2023]
Abstract
Systemic lupus erythematosus (SLE) often features extensive cardiovascular (CV) comorbidity and patients with SLE are at significantly increased risk of CV event occurrence and CV-related mortality. While the specific mechanisms leading to this increased cardiovascular disease (CVD) risk remain to be fully characterized, this heightened risk cannot be fully explained by traditional CV risk factors and is likely driven by immunologic and inflammatory features of SLE. Widespread innate and adaptive immune dysregulation characterize SLE, and factors including excessive type I interferon burden, inappropriate formation and ineffective clearance of neutrophil extracellular traps, and autoantibody formation have been linked to clinical and metabolic features impacting CV risk in SLE and may represent pathogenic drivers of SLE-related CVD. Indeed, functional and phenotypic aberrations in almost every immune cell type are present in SLE and may impact CVD progression. As understanding of the contribution of SLE-specific factors to CVD in SLE improves, improved screening and monitoring of CV risk alongside development of therapeutic treatments aimed at prevention of CVD in SLE patients are required and remain the focus of several ongoing studies and lines of inquiry.
Collapse
Affiliation(s)
- Christopher B Oliveira
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 10 Center Drive, 12N248C, Bethesda, MD, 20892, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 10 Center Drive, 12N248C, Bethesda, MD, 20892, USA.
| |
Collapse
|
14
|
Oliveira CB, Kaplan MJ. Cardiovascular disease risk and pathogenesis in systemic lupus erythematosus. Semin Immunopathol 2022. [PMID: 35355124 DOI: 10.1007/s00281-02200922-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Systemic lupus erythematosus (SLE) often features extensive cardiovascular (CV) comorbidity and patients with SLE are at significantly increased risk of CV event occurrence and CV-related mortality. While the specific mechanisms leading to this increased cardiovascular disease (CVD) risk remain to be fully characterized, this heightened risk cannot be fully explained by traditional CV risk factors and is likely driven by immunologic and inflammatory features of SLE. Widespread innate and adaptive immune dysregulation characterize SLE, and factors including excessive type I interferon burden, inappropriate formation and ineffective clearance of neutrophil extracellular traps, and autoantibody formation have been linked to clinical and metabolic features impacting CV risk in SLE and may represent pathogenic drivers of SLE-related CVD. Indeed, functional and phenotypic aberrations in almost every immune cell type are present in SLE and may impact CVD progression. As understanding of the contribution of SLE-specific factors to CVD in SLE improves, improved screening and monitoring of CV risk alongside development of therapeutic treatments aimed at prevention of CVD in SLE patients are required and remain the focus of several ongoing studies and lines of inquiry.
Collapse
Affiliation(s)
- Christopher B Oliveira
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 10 Center Drive, 12N248C, Bethesda, MD, 20892, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 10 Center Drive, 12N248C, Bethesda, MD, 20892, USA.
| |
Collapse
|
15
|
David C, Duployez N, Eloy P, Belhadi D, Chezel J, Le Guern V, Laouénan C, Fenwarth L, Rouzaud D, Mathian A, de Almeida Chaves S, Duhaut P, Fain O, Galicier L, Ghillani-Dalbin P, Kahn JE, Morel N, Perard L, Pha M, Sarrot-Reynauld F, Aumaitre O, Chasset F, Limal N, Desmurs-Clavel H, Ackermann F, Amoura Z, Papo T, Preudhomme C, Costedoat-Chalumeau N, Sacre K. Clonal hematopoiesis of indeterminate potential and cardiovascular events in systemic lupus erythematosus (HEMATOPLUS study). Rheumatology (Oxford) 2022; 61:4355-4363. [PMID: 35176141 DOI: 10.1093/rheumatology/keac108] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE The detection of somatic mutations in genes of myeloid cells in asymptomatic patients - defining clonal hematopoiesis of indeterminate potential (CHIP) - predisposes to cardiovascular events (CVE) in the general population. We aimed to determine whether CHIP was associated with CVE in SLE patients. METHODS The study is an ancillary study of the randomized, double-blind, placebo-controlled, multicenter trial PLUS study conducted from June 2007 through August 2010 at 37 centers in France involving 573 SLE patients. The search for somatic mutations by high-throughput sequencing of 53 genes involved in clonal hematopoiesis was performed on genomic DNA collected at PLUS inclusion. The CHIP prevalence was assessed in SLE and in a retrospective cohort of 479 patients free of hematological malignancy. The primary outcome was the incident CVE in SLE. RESULTS Screening for CHIP was performed in 438 SLE patients (38 [29-47] years, 91·8% female). Overall, 63 somatic mutations were identified in 47 patients defining a CHIP prevalence of 10·7% in SLE. Most SLE patients (78·7%) carried a single mutation. Most variants (62·5%) were located in the DNMT3A gene. CHIP was associated with age, age at SLE diagnosis and a lower frequency of antiphospholipid antibodies. CHIP occurred more than 20-years earlier (p < 0·00001) in SLE than in controls. The detection of CHIP at inclusion was not associated with the occurrence of CVE during follow up (HR = 0·42 (0·06 - 3·21), p = 0·406). CONCLUSION The prevalence of CHIP is high in SLE with respect to age but was not associated with incident CVE. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov, https://clinicaltrials.gov, NCT05146414.
Collapse
Affiliation(s)
- Clémence David
- Département de Médecine Interne, Hôpital Bichat, Assistance Publique Hôpitaux de Paris (APHP), Institut national de la santé et de la recherche médicale (INSERM) U1149, Université de Paris, Paris, France
| | - Nicolas Duployez
- University of Lille, CNRS, Inserm, CHU Lille, Institut de Recherche contre le Cancer de Lille, UMR9020-UMR-S 1277-Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France
| | - Philippine Eloy
- Departement d'Epidémiologie et de Recherche Clinique, Hôpital Bichat, APHP, Université de Paris, Paris, France
| | - Drifa Belhadi
- Departement d'Epidémiologie et de Recherche Clinique, Hôpital Bichat, APHP, Université de Paris, Paris, France
| | - Julie Chezel
- Département de Médecine Interne, Hôpital Bichat, Assistance Publique Hôpitaux de Paris (APHP), Institut national de la santé et de la recherche médicale (INSERM) U1149, Université de Paris, Paris, France
| | - Véronique Le Guern
- APHP, Hôpital Cochin, Département de Médecine Interne, Centre de Reference Maladies Auto-immunes et Systémiques Rares, Paris, France; Université de Paris, CRESS, INSERM, INRA, Paris, France
| | - Cédric Laouénan
- Departement d'Epidémiologie et de Recherche Clinique, Hôpital Bichat, APHP, Université de Paris, Paris, France
| | - Laurène Fenwarth
- University of Lille, CNRS, Inserm, CHU Lille, Institut de Recherche contre le Cancer de Lille, UMR9020-UMR-S 1277-Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France
| | - Diane Rouzaud
- Département de Médecine Interne, Hôpital Bichat, Assistance Publique Hôpitaux de Paris (APHP), Institut national de la santé et de la recherche médicale (INSERM) U1149, Université de Paris, Paris, France
| | - Alexis Mathian
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Groupement Hospitalier Pitié-Salpêtrière, French National Referral Center for Systemic Lupus Erythematosus, Antiphospholipid Antibody Syndrome and Other Autoimmune Disorders, Service de Médecine Interne 2, Paris, France
| | - Sébastien de Almeida Chaves
- Département de Médecine Interne, Hôpital Purpan, Centre Hospitalo-Universitaire (CHU) de Toulouse, Toulouse, France
| | - Pierre Duhaut
- Département de Médecine Interne, Hôpital Amiens Nord, CHU d'Amiens, Amiens, France
| | - Olivier Fain
- Département de Médecine Interne, Hôpital Saint Antoine, APHP, Université Pierre et Marie Curie, Paris, France
| | - Lionel Galicier
- Département d'Immunologie Clinique, Hôpital Saint Louis, APHP, Université de Paris, Paris, France; Département de Médecine Interne, Hôpital Saint Joseph, Marseille, France
| | - Pascale Ghillani-Dalbin
- Département de Immunologie, Hôpital Pitié-Salpétrière, APHP, Université Pierre et Marie Curie, Paris, France
| | - Jean Emmanuel Kahn
- Département de Médecine Interne, Hôpital Ambroise Paré, APHP, Université de Versailles-Saint-Quentin en Yvelines, France
| | - Nathalie Morel
- APHP, Hôpital Cochin, Département de Médecine Interne, Centre de Reference Maladies Auto-immunes et Systémiques Rares, Paris, France; Université de Paris, CRESS, INSERM, INRA, Paris, France
| | - Laurent Perard
- Département de Médecine Interne, Hôpital St Joseph St Luc, Lyon, France
| | - Micheline Pha
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Groupement Hospitalier Pitié-Salpêtrière, French National Referral Center for Systemic Lupus Erythematosus, Antiphospholipid Antibody Syndrome and Other Autoimmune Disorders, Service de Médecine Interne 2, Paris, France
| | | | - Olivier Aumaitre
- Département de médecine interne, Hôpital Gabriel-Montpied, CHU de Clermont-Ferrand, France
| | - François Chasset
- Département de médecine interne, Hôpital Tenon, APHP Université Pierre et Marie Curie, Paris, France
| | - Nicolas Limal
- Département de médecine interne, Hôpital Henri Mondor, APHP Université Paris-Est Créteil, Paris, France
| | - Helene Desmurs-Clavel
- Département de médecine interne, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Felix Ackermann
- Département de médecine interne, Hôpital Foch, Suresnes, France
| | - Zahir Amoura
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Groupement Hospitalier Pitié-Salpêtrière, French National Referral Center for Systemic Lupus Erythematosus, Antiphospholipid Antibody Syndrome and Other Autoimmune Disorders, Service de Médecine Interne 2, Paris, France
| | - Thomas Papo
- Département de Médecine Interne, Hôpital Bichat, Assistance Publique Hôpitaux de Paris (APHP), Institut national de la santé et de la recherche médicale (INSERM) U1149, Université de Paris, Paris, France
| | - Claude Preudhomme
- University of Lille, CNRS, Inserm, CHU Lille, Institut de Recherche contre le Cancer de Lille, UMR9020-UMR-S 1277-Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France
| | - Nathalie Costedoat-Chalumeau
- APHP, Hôpital Cochin, Département de Médecine Interne, Centre de Reference Maladies Auto-immunes et Systémiques Rares, Paris, France; Université de Paris, CRESS, INSERM, INRA, Paris, France
| | - Karim Sacre
- Département de Médecine Interne, Hôpital Bichat, Assistance Publique Hôpitaux de Paris (APHP), Institut national de la santé et de la recherche médicale (INSERM) U1149, Université de Paris, Paris, France
| |
Collapse
|
16
|
Jha SB, Rivera AP, Flores Monar GV, Islam H, Puttagunta SM, Islam R, Kundu S, Sange I. Systemic Lupus Erythematosus and Cardiovascular Disease. Cureus 2022; 14:e22027. [PMID: 35282557 PMCID: PMC8910778 DOI: 10.7759/cureus.22027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2022] [Indexed: 12/14/2022] Open
|
17
|
Cardiovascular disease in systemic lupus erythematosus. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2021; 2:157-172. [PMID: 35880242 PMCID: PMC9242526 DOI: 10.2478/rir-2021-0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 09/08/2021] [Indexed: 11/21/2022]
Abstract
There is a well-known increased risk for cardiovascular disease that contributes to morbidity and mortality in systemic lupus erythematosus (SLE). Major adverse cardiovascular events and subclinical atherosclerosis are both increased in this patient population. While traditional cardiac risk factors do contribute to the increased risk that is seen, lupus disease-related factors, medications, and genetic factors also impact the overall risk. SLE-specific inflammation, including oxidized lipids, cytokines, and altered immune cell subtypes all are likely to play a role in the pathogenesis of atherosclerotic plaques. Research is ongoing to identify biomarkers that can help clinicians to predict which SLE patients are at the greatest risk for cardiovascular disease (CVD). While SLE-specific treatment regimens for the prevention of cardiovascular events have not been identified, current strategies include minimization of traditional cardiac risk factors and lowering of overall lupus disease activity.
Collapse
|
18
|
Pham PT, Fukuda D, Nishimoto S, Kim-Kaneyama JR, Lei XF, Takahashi Y, Sato T, Tanaka K, Suto K, Kawabata Y, Yamaguchi K, Yagi S, Kusunose K, Yamada H, Soeki T, Wakatsuki T, Shimada K, Kanematsu Y, Takagi Y, Shimabukuro M, Setou M, Barber GN, Sata M. STING, a cytosolic DNA sensor, plays a critical role in atherogenesis: a link between innate immunity and chronic inflammation caused by lifestyle-related diseases. Eur Heart J 2021; 42:4336-4348. [PMID: 34226923 DOI: 10.1093/eurheartj/ehab249] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 02/02/2021] [Accepted: 04/15/2021] [Indexed: 12/23/2022] Open
Abstract
AIMS Lifestyle-related diseases promote atherosclerosis, a chronic inflammatory disease; however, the molecular mechanism remains largely unknown. Endogenous DNA fragments released under over-nutrient condition provoke sterile inflammation through the recognition by DNA sensors. Here, we investigated the role of stimulator of interferon genes (STING), a cytosolic DNA sensor, in atherogenesis. METHODS AND RESULTS Apolipoprotein E-deficient (Apoe-/-) mice fed a western-type diet (WTD), a hypercholesterolaemic mouse model, showed higher STING expression and markers for DNA damage such as γH2AX, p53, and single-stranded DNA (ssDNA) accumulation in macrophages in the aorta compared with wild-type (WT) mice. The level of cGAMP, a STING agonist, in the aorta was higher in Apoe-/- mice. Genetic deletion of Sting in Apoe-/- mice reduced atherosclerotic lesions in the aortic arch, lipid, and macrophage accumulation in plaques, and inflammatory molecule expression in the aorta compared with the control. Pharmacological blockade of STING using a specific inhibitor, C-176, ameliorated atherogenesis in Apoe-/- mice. In contrast, bone marrow-specific STING expression in Apoe-/- mice stimulated atherogenesis. Expression or deletion of STING did not affect metabolic parameters and blood pressure. In vitro studies revealed that STING activation by cGAMP or mitochondrial DNA accelerated inflammatory molecule expression (e.g. TNF-α or IFN-β) in mouse and human macrophages. Activation of nuclear factor-κB and TANK binding kinase 1 was involved in STING-associated vascular inflammation and macrophage activation. Furthermore, human atherosclerotic lesions in the carotid arteries expressed STING and cGAMP. CONCLUSION Stimulator of interferon genes stimulates pro-inflammatory activation of macrophages, leading to the development of atherosclerosis. Stimulator of interferon genes signalling may serve as a potential therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Phuong Tran Pham
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Daiju Fukuda
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan.,Department of Cardio-Diabetes Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Sachiko Nishimoto
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan.,Faculty of Clinical Nutrition and Dietetics, Konan Women's University, 6-2-23, Morikita-machi, Higashinada-ku, Kobe 658-0001, Japan
| | - Joo-Ri Kim-Kaneyama
- Department of Biochemistry, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Xiao-Feng Lei
- Department of Biochemistry, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Yutaka Takahashi
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan.,Preppers, Co., Ltd, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Tokyo 140-001, Japan
| | - Tomohito Sato
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Kimie Tanaka
- Division for Health Service Promotion, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kumiko Suto
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Yutaka Kawabata
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Koji Yamaguchi
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Shusuke Yagi
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Kenya Kusunose
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Hirotsugu Yamada
- Department of Community Medicine for Cardiology, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima, 770-8503, Japan
| | - Takeshi Soeki
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Tetsuzo Wakatsuki
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Kenji Shimada
- Department of Neurosurgery, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Yasuhisa Kanematsu
- Department of Neurosurgery, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Yasushi Takagi
- Department of Neurosurgery, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Michio Shimabukuro
- Department of Cardio-Diabetes Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan.,Department of Diabetes, Endocrinology and Metabolism, School of Medicine, 1 Hikariga-oka, Fukushima 960-1295, Japan
| | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Glen N Barber
- Department of Cell Biology, University of Miami Miller School of Medicine, 1550 NW 10th Avenue, PAP 5th floor Miami, Florida 33136, USA
| | - Masataka Sata
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-Cho, Tokushima 770-8503, Japan
| |
Collapse
|
19
|
Type I Interferon as cardiovascular risk factor in systemic and cutaneous lupus erythematosus: A systematic review. Autoimmun Rev 2021; 20:102794. [PMID: 33722754 DOI: 10.1016/j.autrev.2021.102794] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 01/14/2021] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Patients with systemic lupus erythematosus (SLE) have a high burden of cardiovascular disease (CVD) of multifactorial origin. The aim of this systematic review is to analyze the role of the interferon I (IFN-I) signature and fibroblast growth factor-23 (FGF-23) in patients with SLE or cutaneous lupus erythematosus (CLE) herein. MATERIALS AND METHODS We conducted a systematic literature search in PubMed and Scopus using keywords for major adverse cardiovascular events (MACE) and intermediate outcomes (endothelial dysfunction, subclinical atherosclerosis, platelet activation) associated with IFN-I or FGF-23 in patients with SLE and CLE. RESULTS 4745 citations were screened, of which 12 studies were included. IFN-I was associated with MACE in two third of the studies and the association was strongest for cardiac events. An association of IFN-I was found in all studies investigating impaired vascular function, but only in 50% (respectively 40%) of reports examining the relation of IFN-I and platelet activation (respectively subclinical atherosclerosis). Altogether the reports were of variable bias and quality due to high variability of examined IFN-I biomarkers and inconsistent results for different outcome measures. No studies investigating the cardiovascular risk of circulating IFN-I in CLE, nor FGF-23 in SLE or CLE were found. CONCLUSION Clinical studies measuring the association between IFN-I and direct / intermediate measures of CVD are rare and ambiguous in SLE and nonexistent in CLE, hampering a definite conclusion.
Collapse
|
20
|
Saidoune F, Even G, Lamri Y, Chezel J, Gaston AT, Escoubet B, Papo T, Charles N, Nicoletti A, Sacre K. Effects of BAFF Neutralization on Atherosclerosis Associated With Systemic Lupus Erythematosus. Arthritis Rheumatol 2020; 73:255-264. [PMID: 32783382 DOI: 10.1002/art.41485] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 08/06/2020] [Indexed: 01/29/2023]
Abstract
OBJECTIVE Cardiovascular disease (CVD) is the leading cause of death in systemic lupus erythematosus (SLE). B cells play a key role in the pathogenesis of lupus, and anti-BAFF therapy has been approved for use in SLE. Since mature B cells also promote atherosclerosis, we undertook this study to evaluate, in a mouse model and in SLE patients, whether BAFF neutralization has an atheroprotective effect in SLE. METHODS The effect of BAFF on atherosclerosis associated with lupus was investigated in the atherosclerosis/lupus-prone apolipoprotein E-knockout D227K mouse model and in a cohort of SLE patients. Mice were treated with a blocking anti-BAFF monoclonal antibody (mAb), while fed a standard chow diet. Carotid plaque and carotid intima-media thickness were assessed by ultrasound at baseline and during follow-up in SLE patients who were asymptomatic for CVD. RESULTS Anti-BAFF mAb in ApoE-/- D227K mice induced B cell depletion, efficiently treated lupus, and improved atherosclerosis lesions (21% decrease; P = 0.007) in mice with low plasma cholesterol levels but worsened the lesions (17% increase; P = 0.06) in mice with high cholesterol levels. The atheroprotective effect of the BAFF-BAFF receptor signaling inhibition on B cells was counterbalanced by the proatherogenic effect of the BAFF-TACI signaling inhibition on macrophages. In SLE patients, blood BAFF levels were associated with subclinical atherosclerosis (r = 0.26, P = 0.03). Anti-BAFF mAb treatment had a differential effect on the intima-media thickness progression in SLE patients depending on body mass index. CONCLUSION Depending on the balance between lipid-induced and B cell-induced proatherogenic conditions, anti-BAFF could be detrimental or beneficial, respectively, to atherosclerosis development in SLE.
Collapse
Affiliation(s)
- Fanny Saidoune
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Laboratoire d'Excellence Inflamex, Université de Paris, Paris, France
| | - Guillaume Even
- Université de Paris, INSERM U1148, Laboratoire d'Excellence Inflamex, Paris, France
| | - Yasmine Lamri
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Laboratoire d'Excellence Inflamex, Université de Paris, Paris, France
| | - Julie Chezel
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Laboratoire d'Excellence Inflamex, Université de Paris, Hôpital Bichat-Claude-Bernard, AP-HP, Paris, France
| | - Anh-Thu Gaston
- Université de Paris, INSERM U1148, Laboratoire d'Excellence Inflamex, Paris, France
| | - Brigitte Escoubet
- Université de Paris, Hôpital Bichat-Claude-Bernard, AP-HP, Paris, France
| | - Thomas Papo
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Laboratoire d'Excellence Inflamex, Université de Paris, Hôpital Bichat-Claude-Bernard, AP-HP, Paris, France
| | - Nicolas Charles
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Laboratoire d'Excellence Inflamex, Université de Paris, Paris, France
| | - Antonino Nicoletti
- Université de Paris, INSERM U1148, Laboratoire d'Excellence Inflamex, Paris, France
| | - Karim Sacre
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Laboratoire d'Excellence Inflamex, Université de Paris, Hôpital Bichat-Claude-Bernard, AP-HP, Paris, France
| |
Collapse
|
21
|
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
|
22
|
Schwartz DM, Burma AM, Kitakule MM, Luo Y, Mehta NN. T Cells in Autoimmunity-Associated Cardiovascular Diseases. Front Immunol 2020; 11:588776. [PMID: 33117403 PMCID: PMC7576936 DOI: 10.3389/fimmu.2020.588776] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/21/2020] [Indexed: 12/24/2022] Open
Abstract
T cells are indisputably critical mediators of atherosclerotic cardiovascular disease (CVD), where they secrete pro-inflammatory cytokines that promote vascular pathology. Equally well-established is the fact that autoimmune diseases, which are mediated by autoreactive T cells, substantially increase the risk of developing CVD. Indeed, as immunomodulatory treatments have become more effective at treating end-organ pathology, CVD has become a leading cause of death in patients with autoimmune diseases. Despite this, investigators have only recently begun to probe the mechanisms by which autoreactive T cells promote CVD in the context of autoimmune diseases. T cells are best-studied in the pathogenesis of systemic vasculitides, where they react to self-antigen in the vessel wall. However, newer studies indicate that T cells also contribute to the increased CVD risk associated with lupus and rheumatoid arthritis. Given the central role of T-cell-derived cytokines in the pathogenesis of psoriasis, the role of these factors in psoriatic CVD is also under investigation. In the future, T cells are likely to represent major targets for the prevention and treatment of CVD in patients with autoimmune diseases.
Collapse
Affiliation(s)
- Daniella Muallem Schwartz
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Aarohan M. Burma
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Moses M. Kitakule
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Yiming Luo
- Rheumatology Fellowship Program, National Institute of Arthritis, Musculoskeletal, and Skin Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Nehal N. Mehta
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
23
|
Nishimoto S, Fukuda D, Sata M. Emerging roles of Toll-like receptor 9 in cardiometabolic disorders. Inflamm Regen 2020; 40:18. [PMID: 32714475 PMCID: PMC7374824 DOI: 10.1186/s41232-020-00118-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/21/2020] [Indexed: 02/08/2023] Open
Abstract
Growing evidence suggests that damage-associated molecule patterns (DAMPs) and their receptors, pattern recognition receptors (PRRs), are associated with the progression of cardiometabolic disorders, including obesity-related insulin resistance and atherosclerosis. Cardiometabolic disorders share sterile chronic inflammation as a major cause; however, the exact mechanisms are still obscure. Toll-like receptor 9 (TLR9), one of the nucleic acid-sensing TLRs, recognizes DNA fragments derived from pathogens and contributes to self-defense by activation of the innate immune system. In addition, previous studies demonstrated that TLR9 recognizes DNA fragments released from host cells, accelerating sterile inflammation, which is associated with inflammatory diseases such as autoimmune diseases. In obese adipose tissue and atherosclerotic vascular tissue, various stresses release DNA fragments and/or nuclear proteins as DAMPs from degenerated adipocytes and vascular cells. Recent studies indicated that the activation of TLR9 in immune cells including macrophages and dendritic cells by recognition of these DAMPs promotes inflammation in these tissues, which causes cardiometabolic disorders. This review discusses recent advances in understanding the role of sterile inflammation associated with TLR9 and its endogenous ligands in cardiometabolic disorders. New insights into innate immunity may provide better understanding of cardiometabolic disorders and new therapeutic options for these major health threats in recent decades.
Collapse
Affiliation(s)
- Sachiko Nishimoto
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-cho, Tokushima, 770-8503 Japan
| | - Daiju Fukuda
- Department of Cardio-Diabetes Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, 770-8503 Japan
| | - Masataka Sata
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15, Kuramoto-cho, Tokushima, 770-8503 Japan
| |
Collapse
|
24
|
Jamali A, Harris DL, Blanco T, Lopez MJ, Hamrah P. Resident plasmacytoid dendritic cells patrol vessels in the naïve limbus and conjunctiva. Ocul Surf 2020; 18:277-285. [PMID: 32109562 PMCID: PMC7397780 DOI: 10.1016/j.jtos.2020.02.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 02/15/2020] [Accepted: 02/22/2020] [Indexed: 12/21/2022]
Abstract
Plasmacytoid dendritic cells (pDCs) constitute a unique population of bone marrow-derived cells that play a pivotal role in linking innate and adaptive immune responses. While peripheral tissues are typically devoid of pDCs during steady state, few tissues do host resident pDCs. In the current study, we aim to assess presence and distribution of pDCs in naïve murine limbus and bulbar conjunctiva. Immunofluorescence staining followed by confocal microscopy revealed that the naïve bulbar conjunctiva of wild-type mice hosts CD45+ CD11clow PDCA-1+ pDCs. Flow cytometry confirmed the presence of resident pDCs in the bulbar conjunctiva through multiple additional markers, and showed that they express maturation markers, the T cell co-inhibitory molecules PD-L1 and B7-H3, and minor to negligible levels of T cell co-stimulatory molecules CD40, CD86, and ICAM-1. Epi-fluorescent microscopy of DPE-GFP×RAG1-/- transgenic mice with GFP-tagged pDCs indicated lower density of pDCs in the bulbar conjunctiva compared to the limbus. Further, intravital multiphoton microscopy revealed that resident pDCs accompany the limbal vessels and patrol the intravascular space. In vitro multiphoton microscopy showed that pDCs are attracted to human umbilical vein endothelial cells and interact with them during tube formation. In conclusion, our study shows that the limbus and bulbar conjunctiva are endowed with resident pDCs during steady state, which express maturation and classic T cell co-inhibitory molecules, engulf limbal vessels, and patrol intravascular spaces.
Collapse
Affiliation(s)
- Arsia Jamali
- Center for Translational Ocular Immunology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA; Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA; Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Deshea L Harris
- Center for Translational Ocular Immunology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA; Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA; Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Tomas Blanco
- Center for Translational Ocular Immunology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA; Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Maria J Lopez
- Center for Translational Ocular Immunology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA; Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA; Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Pedram Hamrah
- Center for Translational Ocular Immunology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA; Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA; Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Program in Immunology, School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA; Cornea Service, Tufts New England Eye Center, Boston, MA, USA.
| |
Collapse
|
25
|
Lazou A, Ikonomidis I, Bartekova M, Benedek T, Makavos G, Palioura D, Cabrera Fuentes H, Andreadou I. Chronic inflammatory diseases, myocardial function and cardioprotection. Br J Pharmacol 2020; 177:5357-5374. [PMID: 31943142 DOI: 10.1111/bph.14975] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/22/2019] [Accepted: 11/26/2019] [Indexed: 12/15/2022] Open
Abstract
The association between chronic inflammatory diseases (CIDs) and increased cardiovascular (CV) risk is well documented and can be a most threatening complication in these patients. However, the pathogenetic mechanisms underlying increased CV risk remain elusive, especially in their cellular and biochemical pathways. Using animal models to understand mechanisms underlying cardiac involvement are limited. Additionally, treatments may influence cardiovascular events through different outcomes. Some drugs used to treat CIDs can negatively affect cardiac function by a direct toxicity, whereas others may protect the myocardium. In the present article, we focus on the cardiac manifestations and risk factors, the pathogenetic mechanisms, and the effect of treatments on myocardial function and cardioprotection for five common worldwide CIDs (rheumatoid arthritis, systemic lupus erythematosus, systemic sclerosis, psoriasis and inflammatory bowel disease). We also give recommendations in order to evaluate common targets between CID and CV disease (CVD) and to design therapies to alleviate CID-related CVD. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.23/issuetoc.
Collapse
Affiliation(s)
- Antigone Lazou
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ignatios Ikonomidis
- Second Cardiology Department, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Monika Bartekova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Theodora Benedek
- Clinic of Cardiology, Cardiac Critical Care Unit, University of Medicine and Pharmacy, Târgu Mureş, Romania
| | - George Makavos
- Second Cardiology Department, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitra Palioura
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Hector Cabrera Fuentes
- SingHealth Duke-NUS Cardiovascular Sciences Academic Clinical Programme and Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore.,National Heart Research Institute Singapore, National Heart Centre, Singapore.,Institute of Physiology, Medical School, Justus-Liebig University, Giessen, Germany.,Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Monterrey, NL, Mexico.,Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russian Federation
| | - Ioanna Andreadou
- Laboratory of Pharmacology, School of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
26
|
Nailfold Capillaroscopy in Systemic Lupus Erythematosus (SLE): a Point-of-Care Tool That Parallels Disease Activity and Predicts Future Complications. CURRENT TREATMENT OPTIONS IN RHEUMATOLOGY 2019. [DOI: 10.1007/s40674-019-00133-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
27
|
Abstract
PURPOSE OF REVIEW The mechanisms leading to the development of premature atherosclerosis and vascular injury in systemic lupus erythematosus (SLE) remain to be fully elucidated. This is a comprehensive review of recent research developments related to the understanding of cardiovascular disease (CVD) in lupus. RECENT FINDINGS SLE patients with lupus nephritis display significantly increased risk of myocardial infarction and CVD mortality than SLE patients without lupus nephritis. SLE disease-related parameters could be taken into consideration when calculating CVD risks. The type I interferon pathway is detrimental to the vasculature and may contribute to the development of insulin resistance. The level of low-density granulocytes, a distinct subset of proinflammatory neutrophils present in SLE, was independently associated with coronary plaque burden and endothelial dysfunction. Invariant natural killer T cells may promote an atheroprotective effect in SLE patients with asymptomatic atherosclerotic plaques. Oxidized lupus high-density lipoprotein promotes proinflammatory responses in macrophages. SUMMARY Recent discoveries have further strengthened the critical role of SLE-related immune dysregulation and metabolic disturbances in promoting accelerated CVD. Understanding how these pathogenic factors promote vascular injury may provide better molecular candidates for therapeutic targeting, and ultimately to improve CVD outcomes.
Collapse
|
28
|
David C, Divard G, Abbas R, Escoubet B, Chezel J, Chauveheid MP, Rouzaud D, Boutten A, Papo T, Dehoux M, Sacre K. Soluble CD163 is a biomarker for accelerated atherosclerosis in systemic lupus erythematosus patients at apparent low risk for cardiovascular disease. Scand J Rheumatol 2019; 49:33-37. [PMID: 31161842 DOI: 10.1080/03009742.2019.1614213] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Objective: This study aimed to determine whether sCD163, a soluble macrophage marker up-regulated in numerous inflammatory disorders, is predictive of accelerated atherosclerosis associated with systemic lupus erythematosus (SLE).Methods: Carotid ultrasound was prospectively performed, at baseline and during follow-up, in 63 consecutive SLE patients asymptomatic for cardiovascular disease (CVD) and 18 volunteer health workers. Serum sCD163 level was determined at baseline using enzyme-linked immunosorbent assay. The primary outcome was the presence of a carotid plaque. Factors associated with carotid plaques were identified through multivariate analysis.Results: Despite a low risk for cardiovascular events according to Framingham score in both groups (2.1 ± 3.8% in SLE vs 2.1 ± 2.9% in controls; p = 0.416), ultrasound at baseline showed a carotid plaque in 23 SLE patients (36.5%) and two controls (11.1%) (p = 0.039). Multivariate analysis showed that SLE status increased the risk for carotid plaque by a factor of 9 (p = 0.017). In SLE patients, sCD163 level was high (483.7 ± 260.8 ng/mL vs 282.1 ± 97.5 ng/mL in controls; p < 0.001) and independently associated with carotid plaques, as assessed by stratification based on sCD163 quartile values (p = 0.009), receiver operating characteristics (p = 0.001), and multivariate analysis (p = 0.015). sCD163 at baseline was associated with the onset of carotid plaque during follow-up (3 ± 1.4 years) in SLE patients who had no carotid plaque at the first evaluation (p = 0.041).Conclusion: sCD163 is associated with progressing carotid plaque in SLE and may be a useful biomarker for accelerated atherosclerosis in SLE patients at apparent low risk for CVD.
Collapse
Affiliation(s)
- C David
- Departement of Internal Medicine, Bichat Hospital, University of Paris Diderot, PRES Sorbonne Paris Cité, APHP, Public Hospitals of Paris, Paris, France
| | - G Divard
- Departement of Internal Medicine, Bichat Hospital, University of Paris Diderot, PRES Sorbonne Paris Cité, APHP, Public Hospitals of Paris, Paris, France
| | - R Abbas
- Department of Epidemiology and Clinical Research, Bichat Hospital, University of Paris Diderot, PRES Sorbonne Paris Cité, APHP, Public Hospitals of Paris, Paris, France
| | - B Escoubet
- Departement of Physiology, Bichat Hospital, University of Paris Diderot, PRES Sorbonne Paris Cité, APHP, Public Hospitals of Paris, INSERM, Paris, France
| | - J Chezel
- Departement of Internal Medicine, Bichat Hospital, University of Paris Diderot, PRES Sorbonne Paris Cité, APHP, Public Hospitals of Paris, Paris, France
| | - M P Chauveheid
- Departement of Internal Medicine, Bichat Hospital, University of Paris Diderot, PRES Sorbonne Paris Cité, APHP, Public Hospitals of Paris, Paris, France
| | - D Rouzaud
- Departement of Internal Medicine, Bichat Hospital, University of Paris Diderot, PRES Sorbonne Paris Cité, APHP, Public Hospitals of Paris, Paris, France
| | - A Boutten
- Department of Metabolic and Cellular Biochemistry, Bichat Hospital, University of Paris Diderot, PRES Sorbonne Paris Cité, APHP, Public Hospitals of Paris, Paris, France
| | - T Papo
- Departement of Internal Medicine, Bichat Hospital, University of Paris Diderot, PRES Sorbonne Paris Cité, APHP, Public Hospitals of Paris, Paris, France.,INSERM U1149 (French Institute of Health and Medical Research), University of Paris Diderot, Paris, France.,Hospital-University Department of Fibrosis, Inflammation and Remodelling in Renal and Respiratory Diseases, FIRE, Paris, France
| | - M Dehoux
- Department of Metabolic and Cellular Biochemistry, Bichat Hospital, University of Paris Diderot, PRES Sorbonne Paris Cité, APHP, Public Hospitals of Paris, Paris, France
| | - K Sacre
- Departement of Internal Medicine, Bichat Hospital, University of Paris Diderot, PRES Sorbonne Paris Cité, APHP, Public Hospitals of Paris, Paris, France.,INSERM U1149 (French Institute of Health and Medical Research), University of Paris Diderot, Paris, France.,Hospital-University Department of Fibrosis, Inflammation and Remodelling in Renal and Respiratory Diseases, FIRE, Paris, France
| |
Collapse
|
29
|
Wei Y, Lao XM, Xiao X, Wang XY, Wu ZJ, Zeng QH, Wu CY, Wu RQ, Chen ZX, Zheng L, Li B, Kuang DM. Plasma Cell Polarization to the Immunoglobulin G Phenotype in Hepatocellular Carcinomas Involves Epigenetic Alterations and Promotes Hepatoma Progression in Mice. Gastroenterology 2019; 156:1890-1904.e16. [PMID: 30711627 DOI: 10.1053/j.gastro.2019.01.250] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 01/08/2019] [Accepted: 01/25/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND & AIMS Little is known about the composition and generation of plasma cell subsets in patients with hepatocellular carcinoma (HCC) and how these associate with outcomes. We investigated whether, or how, plasma cells differentiate and function in patients with HCC and mice with liver tumors. METHODS We analyzed subset composition and distribution of plasma cells in HCC samples from 342 patients who underwent curative resection at the Cancer Center of Sun Yat-sen University in China; samples of non-tumor liver tissue were used as controls. We associated plasma cell profiles with patient outcomes. Tissue-derived leukocytes were analyzed by flow cytometry and real-time polymerase chain reaction. The ability of macrophages to regulate plasma cell differentiation was determined in ex vivo cultures of cells from human HCC tissues. C57BL/6 and BALB/c mice were given injections of Hepa1-6 cells, which formed hepatomas, or H22 cells, which formed ascitic hepatomas. Gene expression patterns were analyzed in human HCC, mouse hepatoma, and non-tumor tissues by real-time polymerase chain reaction. Mice with hepatomas were given injections of GSK126 (an inhibitor of histone H3 lysine 27 methyltransferase [EZH2]) and 5-AZA-dC (an inhibitor of DNA methyltransferases); tumor tissues were analyzed by immunofluorescence and immunohistochemistry for the presence of immune cells and cytokines. RESULTS B cells isolated from HCCs had somatic hypermutations and class-switch recombinations to the IgG phenotype that were not observed in non-tumor tissues. Increased level of plasma cells correlated with poor outcomes of patients. Activated CD4+ T cells from HCCs stimulated C-X-C motif chemokine 10 (CXCL10) production by macrophages. CXCL10 bound CXC chemokine receptor 3 on B cells and signaled via extracellular signal-regulated kinase to cause them to become IgG-producing plasma cells. IgG activated Fc receptors on macrophages and induced them to produce interleukin 6, interleukin 10, and C-C motif chemokine ligand 20 (CCL20). In mice with hepatomas, depletion of B cells prevented generation of these macrophage, increased the anti-tumor T cell response, and reduced growth of hepatomas. However, these effects were lost after injection of CXC chemokine receptor 3-positive plasma cells. Human HCC and mouse hepatoma tissues had increased expression of DNA methyltransferase 1 and EZH2 compared with non-tumor tissues. Injection of mice with GSK126 and 5-AZA-dC induced expression of CXCL10 by tumor cells and caused plasma cell polarization, suppression of the anti-tumor T cell response, and hepatoma growth. CONCLUSIONS Human HCC tissues contain B cells with class-switch recombinations to the IgG phenotype. Activated CD4+ T cells from HCCs stimulate CXCL10 production by macrophages; CXCL10 binds CXC chemokine receptor 3 on B cells and causes them to become IgG-producing plasma cells. IgG activates Fc receptor in macrophages to produce cytokines that reduce the anti-tumor immune response. In mice with hepatomas, depletion of B cells prevented generation of these macrophages, increased the anti-tumor T cell response, and reduced growth of hepatomas. This pathway involves increased expression of DNA methyltransferase 1 and EZH2 by HCC and hepatoma cells.
Collapse
Affiliation(s)
- Yuan Wei
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiang-Ming Lao
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiao Xiao
- Cancer Immunoregulation and Immunotherapy Laboratory, Queensland Institute of Medical Research, Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Xu-Yan Wang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zong-Jian Wu
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Qiu-Hui Zeng
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Cai-Yuan Wu
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Rui-Qi Wu
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhen-Xin Chen
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Limin Zheng
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Bo Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Dong-Ming Kuang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, People's Republic of China; Cancer Immunoregulation and Immunotherapy Laboratory, Queensland Institute of Medical Research, Berghofer Medical Research Institute, Herston, Queensland, Australia; The Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
30
|
Tsiantoulas D, Sage AP, Göderle L, Ozsvar-Kozma M, Murphy D, Porsch F, Pasterkamp G, Menche J, Schneider P, Mallat Z, Binder CJ. B Cell-Activating Factor Neutralization Aggravates Atherosclerosis. Circulation 2018; 138:2263-2273. [PMID: 29858401 PMCID: PMC6181204 DOI: 10.1161/circulationaha.117.032790] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Atherosclerotic cardiovascular disease (heart attacks and strokes) is the major cause of death globally and is caused by the buildup of a plaque in the arterial wall. Genomic data showed that the B cell-activating factor (BAFF) receptor pathway, which is specifically essential for the survival of conventional B lymphocytes (B-2 cells), is a key driver of coronary heart disease. Deletion or antibody-mediated blockade of BAFF receptor ablates B-2 cells and decreases experimental atherosclerosis. Anti-BAFF immunotherapy is approved for treatment of autoimmune systemic lupus erythematosus, and can therefore be expected to limit their associated cardiovascular risk. However, direct effects of anti-BAFF immunotherapy on atherosclerosis remain unknown. METHODS To investigate the effect of BAFF neutralization in atherosclerosis, the authors treated Apoe-/- and Ldlr-/- mice with a well-characterized blocking anti-BAFF antibody. Moreover, to investigate the mechanism by which BAFF impacts atherosclerosis, the authors studied atherosclerosis-prone mice that lack the alternative receptor for BAFF: transmembrane activator and calcium modulator and cyclophilin ligand interactor. RESULTS The authors demonstrate here that anti-BAFF antibody treatment increased atherosclerosis in mice, despite efficient depletion of mature B-2 cells, suggesting a unique mechanism of action. Indeed, myeloid cell-specific deletion of transmembrane activator and calcium modulator and cyclophilin ligand interactor also results in increased atherosclerosis, while B cell-specific transmembrane activator and calcium modulator and cyclophilin ligand interactor deletion had no effect. Mechanistically, BAFF-transmembrane activator and calcium modulator and cyclophilin ligand interactor signaling represses macrophage IRF7-dependent (but not NF-κB-dependent) Toll-like receptor 9 responses including proatherogenic CXCL10 production. CONCLUSIONS These data identify a novel B cell-independent anti-inflammatory role for BAFF in atherosclerosis and may have important clinical implications.
Collapse
Affiliation(s)
- Dimitrios Tsiantoulas
- Department of Laboratory Medicine, Medical University of Vienna (D.T., L.G., M.O.-K., F.P., C.J.B.),CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences (D.T., L.G., M.O.-K., F.P., J.M., C.J.B.)
| | - Andrew P. Sage
- Division of Cardiovascular Medicine, University of Cambridge, UK (A.P.S., D.M., Z.M.)
| | - Laura Göderle
- Department of Laboratory Medicine, Medical University of Vienna (D.T., L.G., M.O.-K., F.P., C.J.B.),CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences (D.T., L.G., M.O.-K., F.P., J.M., C.J.B.)
| | - Maria Ozsvar-Kozma
- Department of Laboratory Medicine, Medical University of Vienna (D.T., L.G., M.O.-K., F.P., C.J.B.),CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences (D.T., L.G., M.O.-K., F.P., J.M., C.J.B.)
| | - Deirdre Murphy
- Division of Cardiovascular Medicine, University of Cambridge, UK (A.P.S., D.M., Z.M.)
| | - Florentina Porsch
- Department of Laboratory Medicine, Medical University of Vienna (D.T., L.G., M.O.-K., F.P., C.J.B.),CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences (D.T., L.G., M.O.-K., F.P., J.M., C.J.B.)
| | | | - Jörg Menche
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences (D.T., L.G., M.O.-K., F.P., J.M., C.J.B.)
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland (P.S.)
| | - Ziad Mallat
- Division of Cardiovascular Medicine, University of Cambridge, UK (A.P.S., D.M., Z.M.).,Institut National de la Santé et de la Recherche Médicale, Paris Cardiovascular Research Center (PARCC), Paris, France (Z.M.)
| | - Christoph J. Binder
- Department of Laboratory Medicine, Medical University of Vienna (D.T., L.G., M.O.-K., F.P., C.J.B.),CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences (D.T., L.G., M.O.-K., F.P., J.M., C.J.B.)
| |
Collapse
|
31
|
Qi S, Chen Q, Xu D, Xie N, Dai Y. Clinical application of protein biomarkers in lupus erythematosus and lupus nephritis. Lupus 2018; 27:1582-1590. [PMID: 29720035 DOI: 10.1177/0961203318773643] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Systemic lupus erythematosus (SLE) is a type of autoimmune disease that damages multiple organs, including the heart, joints, liver and kidneys. The main characteristics of SLE are the deposition of circulating autoantibodies; autoantigen complexes in the renal system; and abnormal expression of complements, cytokines and chemokines. Lupus nephritis (LN) is the most serious manifestation of SLE and is characterized by inflammation of the kidney. This review summarizes recent clinical applications of protein biomarkers including autoantibodies, complements, cytokines and chemokines and some new protein biomarkers in SLE and LN. The clinical differential diagnosis of protein biomarkers related to prognosis and diagnosis is discussed and highlighted. Protein biomarkers play crucial roles in the study of SLE and LN. Useful biomarkers will provide insights into effective treatments for these diseases.
Collapse
Affiliation(s)
- S Qi
- 1 National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, China
| | - Q Chen
- 1 National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, China
| | - D Xu
- 1 National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, China
| | - N Xie
- 1 National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, China
| | - Y Dai
- 2 Shenzhen People's Hospital, Shenzhen, China
| |
Collapse
|
32
|
Weigold F, Günther J, Pfeiffenberger M, Cabral-Marques O, Siegert E, Dragun D, Philippe A, Regensburger AK, Recke A, Yu X, Petersen F, Catar R, Biesen R, Hiepe F, Burmester GR, Heidecke H, Riemekasten G. Antibodies against chemokine receptors CXCR3 and CXCR4 predict progressive deterioration of lung function in patients with systemic sclerosis. Arthritis Res Ther 2018; 20:52. [PMID: 29566745 PMCID: PMC5863842 DOI: 10.1186/s13075-018-1545-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 02/18/2018] [Indexed: 01/16/2023] Open
Abstract
Background The chemokine receptors CXCR3 and CXCR4 are involved in the pathogenesis of fibrosis, a key feature of systemic sclerosis (SSc). It is hypothesized that immunoglobulin (Ig)G antibodies (abs) against these two receptors are present in patients with SSc and are associated with clinical findings. Methods Anti-CXCR3 and anti-CXCR4 ab levels were measured in 449 sera from 327 SSc patients and in 234 sera from healthy donors (HD) by enzyme-linked immunosorbent assay (ELISA). In SSc, ab levels were compared with clinical data in a cross-sectional and longitudinal setting. Protein expression of CXCR3 and CXCR4 on peripheral blood mononuclear cells (PBMCs) was analyzed in 17 SSc patients and 8 HD by flow cytometry. Results Anti-CXCR3 and anti-CXCR4 ab levels were different among SSc subgroups compared with HD and were highest in diffuse SSc patients. The ab levels strongly correlated with each other (r = 0.85). Patients with SSc-related interstitial lung disease (SSc-ILD) exhibited higher ab levels which negatively correlated with lung function parameters (e.g., r = −0.5 and r = −0.43 for predicted vital capacity, respectively). However, patients with deterioration of lung function showed lower anti-CXCR3/4 ab levels compared with those with stable disease. Frequencies and median fluorescence intensities (MFI) of CXCR3+ and CXCR4+ PBMCs were lower in SSc patients compared with HD and correlated with the severity of skin and lung fibrosis. They correlated with the severity of skin and lung fibrosis. Conclusions Anti-CXCR3/4 abs and their corresponding receptors are linked with the severity of SSc-ILD. Antibody levels discriminate patients with stable or decreasing lung function and could be used for risk stratification. Electronic supplementary material The online version of this article (10.1186/s13075-018-1545-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Florian Weigold
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Berlin, Germany
| | - Jeannine Günther
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Berlin, Germany.,Cell Autoimmunity Group, German Rheumatism Research Center (DRFZ), Berlin, Germany
| | | | | | - Elise Siegert
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Berlin, Germany
| | - Duska Dragun
- Department of Nephrology and Critical Care Medicine, Charité University Hospital, Campus Virchow, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Aurélie Philippe
- Department of Nephrology and Critical Care Medicine, Charité University Hospital, Campus Virchow, Berlin, Germany
| | | | - Andreas Recke
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Xinhua Yu
- Research Center Borstel, Airway Research Center North (ARCN), Members of the German Center for Lung Research (DZL), Borstel, Germany
| | - Frank Petersen
- Research Center Borstel, Airway Research Center North (ARCN), Members of the German Center for Lung Research (DZL), Borstel, Germany
| | - Rusan Catar
- Department of Nephrology and Critical Care Medicine, Charité University Hospital, Campus Virchow, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Robert Biesen
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Berlin, Germany
| | - Falk Hiepe
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Berlin, Germany
| | - Gerd R Burmester
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Berlin, Germany
| | | | - Gabriela Riemekasten
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Berlin, Germany. .,Cell Autoimmunity Group, German Rheumatism Research Center (DRFZ), Berlin, Germany. .,Department of Rheumatology, University of Lübeck, Lübeck, Germany. .,Research Center Borstel, Airway Research Center North (ARCN), Members of the German Center for Lung Research (DZL), Borstel, Germany.
| |
Collapse
|
33
|
Maffia P, Döring Y, Biessen EAL, Mallat Z. Commentary: Indoleamine 2,3-Dioxygenase-Expressing Aortic Plasmacytoid Dendritic Cells Protect against Atherosclerosis by Induction of Regulatory T Cells. Front Immunol 2017; 8:140. [PMID: 28243244 PMCID: PMC5304905 DOI: 10.3389/fimmu.2017.00140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 01/27/2017] [Indexed: 01/31/2023] Open
Affiliation(s)
- Pasquale Maffia
- Centre for Immunobiology, College of Medical, Veterinary and Life Sciences, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK; BHF Centre of Excellence in Vascular Science and Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK; Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Yvonne Döring
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Erik A L Biessen
- Experimental Vascular Pathology Group, Department of Pathology, CARIM, Maastricht University Medical Center, Maastricht, Netherlands; Institute of Molecular Cardiovascular Research, RWTH Klinikum Aachen, Aachen, Germany
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK; Unit 970, Paris Cardiovascular Research Center, Institut National de la Santé et de la Recherche Médicale, Paris, France
| |
Collapse
|
34
|
Mina-Osorio P. Stem Cell Therapy in the Treatment of Rheumatic Diseases and Application in the Treatment of Systemic Lupus Erythematosus. NEXT-GENERATION THERAPIES AND TECHNOLOGIES FOR IMMUNE-MEDIATED INFLAMMATORY DISEASES 2017. [PMCID: PMC7123283 DOI: 10.1007/978-3-319-42252-7_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Current systemic therapies help to improve the symptoms and quality of life for patients with severe life-threatening rheumatic diseases but provide no curative treatment. For the past two decades, preclinical and clinical studies of stem cell transplantation (SCT) have demonstrated tremendous therapeutic potential for patients with autoimmune rheumatic diseases. Herein, the current advances on stem cell therapies, both in animal models and clinical studies, are discussed, with particular attention on systemic lupus erythematosus (SLE). Despite extensive research and promising data, our knowledge on mechanisms of action for SCT, its administration route and timing, the optimal dose of cells, the cells’ fate and distribution in vivo, and the safety and efficacy of the treatments remains limited. Further research on stem cell biology is required to ensure that therapeutic safety and efficacy, as observed in animal models, can be successfully translated in clinical trials. Current understanding, limitations, and future directions for SCT with respect to rheumatic diseases are also discussed.
Collapse
|
35
|
Doria A, Gershwin ME, Selmi C. From old concerns to new advances and personalized medicine in lupus: The end of the tunnel is approaching. J Autoimmun 2016; 74:1-5. [DOI: 10.1016/j.jaut.2016.08.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 08/23/2016] [Indexed: 12/11/2022]
|
36
|
|
37
|
Breakdown of Immune Tolerance in Systemic Lupus Erythematosus by Dendritic Cells. J Immunol Res 2016; 2016:6269157. [PMID: 27034965 PMCID: PMC4789470 DOI: 10.1155/2016/6269157] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 01/15/2016] [Accepted: 02/07/2016] [Indexed: 02/06/2023] Open
Abstract
Dendritic cells (DC) play an important role in the pathogenesis of systemic lupus erythematosus (SLE), an autoimmune disease with multiple tissue manifestations. In this review, we summarize recent studies on the roles of conventional DC and plasmacytoid DC in the development of both murine lupus and human SLE. In the past decade, studies using selective DC depletions have demonstrated critical roles of DC in lupus progression. Comprehensive in vitro and in vivo studies suggest activation of DC by self-antigens in lupus pathogenesis, followed by breakdown of immune tolerance to self. Potential treatment strategies targeting DC have been developed. However, many questions remain regarding the mechanisms by which DC modulate lupus pathogenesis that require further investigations.
Collapse
|
38
|
Choi J, Selmi C, Leung PSC, Kenny TP, Roskams T, Gershwin ME. Chemokine and chemokine receptors in autoimmunity: the case of primary biliary cholangitis. Expert Rev Clin Immunol 2016; 12:661-72. [PMID: 26821815 DOI: 10.1586/1744666x.2016.1147956] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Chemokines represent a major mediator of innate immunity and play a key role in the selective recruitment of cells during localized inflammatory responses. Beyond critical extracellular mediators of leukocyte trafficking, chemokines and their cognate receptors are expressed by a variety of resident and infiltrating cells (monocytes, lymphocytes, NK cells, mast cells, and NKT cells). Chemokines represent ideal candidates for mechanistic studies (particularly in murine models) to better understand the pathogenesis of chronic inflammation and possibly become biomarkers of disease. Nonetheless, therapeutic approaches targeting chemokines have led to unsatisfactory results in rheumatoid arthritis, while biologics against pro-inflammatory cytokines are being used worldwide with success. In this comprehensive review we will discuss the evidence supporting the involvement of chemokines and their specific receptors in mediating the effector cell response, utilizing the autoimmune/primary biliary cholangitis setting as a paradigm.
Collapse
Affiliation(s)
- Jinjung Choi
- a Division of Rheumatology, Allergy and Clinical Immunology , University of California Davis , Davis , CA , USA.,b Division of Rheumatology , CHA University Medical Center , Bundang , Korea
| | - Carlo Selmi
- c Rheumatology and Clinical Immunology , Humanitas Research Hospital , Rozzano , Italy.,d BIOMETRA Department , University of Milan , Milano , Italy
| | - Patrick S C Leung
- a Division of Rheumatology, Allergy and Clinical Immunology , University of California Davis , Davis , CA , USA
| | - Thomas P Kenny
- a Division of Rheumatology, Allergy and Clinical Immunology , University of California Davis , Davis , CA , USA
| | - Tania Roskams
- e Translational Cell and Tissue Research , University of Leuven , Leuven , Belgium
| | - M Eric Gershwin
- a Division of Rheumatology, Allergy and Clinical Immunology , University of California Davis , Davis , CA , USA
| |
Collapse
|