1
|
Jury EC, Peng J, Van Vijfeijken A, Martin Gutierrez L, Woodridge L, Wincup C, Pineda-Torra I, Ciurtin C, Robinson GA. Systemic lupus erythematosus patients have unique changes in serum metabolic profiles across age associated with cardiometabolic risk. Rheumatology (Oxford) 2024; 63:2741-2753. [PMID: 38048621 PMCID: PMC11443078 DOI: 10.1093/rheumatology/kead646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/02/2023] [Accepted: 11/05/2023] [Indexed: 12/06/2023] Open
Abstract
OBJECTIVES Cardiovascular disease through accelerated atherosclerosis is a leading cause of mortality for patients with systemic lupus erythematosus (SLE), likely due to increased chronic inflammation and cardiometabolic defects over age. We investigated age-associated changes in metabolomic profiles of SLE patients and healthy controls (HCs). METHODS Serum NMR metabolomic profiles from female SLE patients (n = 164, age = 14-76) and HCs (n = 123, age = 13-72) were assessed across age by linear regression and by age group between patients/HCs (Group 1, age ≤ 25, n = 62/46; Group 2, age = 26-49, n = 50/46; Group 3, age ≥ 50, n = 52/31) using multiple t tests. The impact of inflammation, disease activity and treatments were assessed, and UK Biobank disease-wide association analysis of metabolites was performed. RESULTS Age-specific metabolomic profiles were identified in SLE patients vs HCs, including reduced amino acids (Group 1), increased very-low-density lipoproteins (Group 2), and increased low-density lipoproteins (Group 3). Twenty-five metabolites were significantly altered in all SLE age groups, dominated by decreased atheroprotective high-density lipoprotein (HDL) subsets, HDL-bound apolipoprotein (Apo)A1 and increased glycoprotein acetyls (GlycA). Furthermore, ApoA1 and GlycA were differentially associated with disease activity and serological measures, as well as atherosclerosis incidence and myocardial infarction mortality risk through disease-wide association. Separately, glycolysis pathway metabolites (acetone/citrate/creatinine/glycerol/lactate/pyruvate) uniquely increased with age in SLE, significantly influenced by prednisolone (increased pyruvate/lactate) and hydroxychloroquine (decreased citrate/creatinine) treatment and associated with type 1 and type 2 diabetes by disease-wide association. CONCLUSIONS Increasing HDL (ApoA1) levels through therapeutic/nutritional intervention, whilst maintaining low disease activity, in SLE patients from a young age could improve cardiometabolic disease outcomes. Biomarkers from the glycolytic pathway could indicate adverse metabolic effects of current therapies.
Collapse
Affiliation(s)
- Elizabeth C Jury
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
| | - Junjie Peng
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| | | | - Lucia Martin Gutierrez
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| | - Laurel Woodridge
- Centre for Experimental & Translational Medicine, Division of Medicine, University College London, London, UK
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
| | - Chris Wincup
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
| | - Ines Pineda-Torra
- Centre for Experimental & Translational Medicine, Division of Medicine, University College London, London, UK
| | - Coziana Ciurtin
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| | - George A Robinson
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| |
Collapse
|
2
|
Oliveira C, Temesgen-Oyelakin Y, Naqi M, Davis M, Naz F, Dell'Orso S, Brooks S, Kuhn S, Hill T, Li X, Patel N, Parel P, Gadina M, Gupta S, Mehta N, Hasni SA, Kaplan MJ. A Multiomic Analysis to Identify Drivers of Subclinical Vascular Disease in Systemic Lupus Erythematosus. Arthritis Rheumatol 2024; 76:1501-1511. [PMID: 38923259 DOI: 10.1002/art.42925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/05/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) increases cardiovascular disease (CVD) risk, and this is not explained by traditional risk factors. Characterization of blood immunologic signatures that associate with subclinical CVD and predict its progression has been challenging and may help identify subgroups at risk. METHODS Patients with SLE (n = 77) and healthy controls (HCs) (n = 27) underwent assessments of arterial stiffness, vascular wall inflammation, and coronary atherosclerosis burden with cardio-ankle vascular index (CAVI); fluorodeoxyglucose-positron emission tomography/computed tomography (CT) (target-to-background ratio [TBR]); and coronary CT angiography. Whole blood bulk RNA sequencing was performed in a subset of study participants (HC n = 10, SLE n = 20). In a partially overlapping subset (HC n = 24, SLE n = 64), serum inflammatory protein biomarkers were quantified with an Olink platform. RESULTS CAVI, TBR, and noncalcified coronary plaque burden (NCB) were increased in patients with SLE compared to HCs. When comparing patients with SLE with high CAVI scores to those with low CAVI scores or to HCs, there was a down-regulation of genes in pathways involved in the cell cycle and differentially regulated pathways related to metabolism. Distinct serum proteins associated with increased CAVI (CCL23, colony-stimulating factor 1, latency-activating peptide transforming growth factor β1, interleukin 33 [IL-33], CD8A, and IL-12B), NCB (monocyte chemotactic protein 4 and FMS-like tyrosine kinase 3 ligand [Flt3L]), and TBR (CD5, IL-1α, AXIN1, cystatin D [CST5], and tumor necrosis factor receptor superfamily 9; P < 0.05). CONCLUSION Blood gene expression patterns and serum proteins that associate with worse vascular phenotypes suggest dysregulated immune and metabolic pathways linked to premature CVD. Cytokines and chemokines identified in associations with arterial stiffness, inflammation, and NCB in SLE may allow for characterization of new CVD biomarkers in lupus.
Collapse
Affiliation(s)
- Christopher Oliveira
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland
| | | | - Mohammad Naqi
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland
| | - Michael Davis
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland
| | - Faiza Naz
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland
| | - Stefania Dell'Orso
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland
| | - Stephen Brooks
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland
| | - Skyler Kuhn
- National Institute of Allergy and Infectious Diseases Collaborative Bioinformatics Resource, NIH, Bethesda, Maryland
| | - Tom Hill
- National Institute of Allergy and Infectious Diseases Collaborative Bioinformatics Resource, NIH, Bethesda, Maryland
| | | | - Nidhi Patel
- National Heart, Lung and Blood Institute, NIH, Bethesda, Maryland
| | - Philip Parel
- National Heart, Lung and Blood Institute, NIH, Bethesda, Maryland
| | - Massimo Gadina
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland
| | - Sarthak Gupta
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland
| | - Nehal Mehta
- National Heart, Lung and Blood Institute, NIH, Bethesda, Maryland
| | - Sarfaraz A Hasni
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland
| | - Mariana J Kaplan
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland
| |
Collapse
|
3
|
Joyce DP, Berger JS, Guttmann A, Hasan G, Buyon JP, Belmont HM, Salmon J, Askanase A, Bathon J, Geraldino-Pardilla L, Ali Y, Ginzler EM, Putterman C, Gordon C, Helmick CG, Barbour KE, Gold HT, Parton H, Izmirly PM. Prevalence of cardiovascular events in a population-based registry of patients with systemic lupus erythematosus. Arthritis Res Ther 2024; 26:160. [PMID: 39272198 PMCID: PMC11401284 DOI: 10.1186/s13075-024-03395-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND The Manhattan Lupus Surveillance Program (MLSP), a population-based retrospective registry of patients with systemic lupus erythematosus (SLE), was used to investigate the prevalence of cardiovascular disease events (CVE) and compare rates among sex, age and race/ethnicity to population-based controls. METHODS Patients with prevalent SLE in 2007 aged ≥ 20 years in the MLSP were included. CVE required documentation of a myocardial infarction or cerebrovascular accident. We calculated crude risk ratios and adjusted risk ratios (ARR) controlling for sex, age group, race and ethnicity, and years since diagnosis. Data from the 2009-2010 National Health and Nutrition Examination Survey (NHANES) and the 2013-2014 NYC Health and Nutrition Examination Survey (NYC HANES) were used to calculate expected CVE prevalence by multiplying NHANES and NYC HANES estimates by strata-specific counts of patients with SLE. Crude prevalence ratios (PRs) using national and NYC estimates and age standardized prevalence ratios (ASPRs) using national estimates were calculated. RESULTS CVE occurred in 13.9% of 1,285 MLSP patients with SLE, and risk was increased among men (ARR:1.7, 95%CI:1.2-2.5) and older adults (age > 60 ARR:2.5, 95%CI:1.7-3.8). Compared with non-Hispanic Asian patients, CVE risk was elevated among Hispanic/Latino (ARR:3.1, 95%CI:1.4-7.0) and non-Hispanic Black (ARR:3.5, 95%CI1.6-7.9) patients as well as those identified as non-Hispanic and in another or multiple racial groups (ARR:4.2, 95%CI:1.1-15.8). Overall, CVE prevalence was higher among patients with SLE than nationally (ASPR:3.1, 95%CI:3.0-3.1) but did not differ by sex. Compared with national race and ethnicity-stratified estimates, CVE among patients with SLE was highest among Hispanics/Latinos (ASPR:4.3, 95%CI:4.2-4.4). CVE was also elevated among SLE registry patients compared with all NYC residents. Comparisons with age-stratified national estimates revealed PRs of 6.4 (95%CI:6.2-6.5) among patients aged 20-49 years and 2.2 (95%CI:2.1-2.2) among those ≥ 50 years. Male (11.3, 95%CI:10.5-12.1), Hispanic/Latino (10.9, 95%CI:10.5-11.4) and non-Hispanic Black (6.2, 95%CI:6.0-6.4) SLE patients aged 20-49 had the highest CVE prevalence ratios. CONCLUSIONS These population-based estimates of CVE in a diverse registry of patients with SLE revealed increased rates among younger male, Hispanic/Latino and non-Hispanic Black patients. These findings reinforce the need to appropriately screen for CVD among all SLE patients but particularly among these high-risk patients.
Collapse
Affiliation(s)
- Daniel P Joyce
- New York University Grossman School of Medicine, New York, NY, USA
| | - Jeffrey S Berger
- New York University Grossman School of Medicine, New York, NY, USA
| | - Allison Guttmann
- Institute for Rheumatic & Autoimmune Diseases, Atlantic Medical Group Rheumatology, Overlook Medical Center, Atlantic Health System, Summit, Morristown, NJ, NJ, USA
| | | | - Jill P Buyon
- New York University Grossman School of Medicine, New York, NY, USA
| | | | - Jane Salmon
- Hospital for Special Surgery, New York, NY, USA
| | - Anca Askanase
- Columbia University Medical Center, New York, NY, USA
| | - Joan Bathon
- Columbia University Medical Center, New York, NY, USA
| | | | - Yousaf Ali
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ellen M Ginzler
- SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | | | - Caroline Gordon
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | | | - Kamil E Barbour
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Heather T Gold
- New York University Grossman School of Medicine, New York, NY, USA
| | - Hilary Parton
- New York City Department of Health and Mental Hygiene, New York, NY, USA
| | - Peter M Izmirly
- New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
4
|
Bergkamp SC, Bergkamp ND, Wahadat MJ, Gruppen MP, Nassar-Sheikh Rashid A, Tas SW, Smit MJ, Versnel MA, van den Berg JM, Kamphuis S, Schonenberg-Meinema D. Learning from serum markers reflecting endothelial activation: longitudinal data in childhood-onset systemic lupus erythematosus. Lupus Sci Med 2024; 11:e001190. [PMID: 39242108 PMCID: PMC11381702 DOI: 10.1136/lupus-2024-001190] [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: 03/22/2024] [Accepted: 08/04/2024] [Indexed: 09/09/2024]
Abstract
OBJECTIVES In childhood-onset SLE (cSLE), patients have an increased risk of premature atherosclerosis. The pathophysiological mechanisms for this premature atherosclerosis are not yet completely understood, but besides traditional risk factors, the endothelium plays a major role. The first aim of this study was to measure levels of SLE-associated markers involved in endothelial cell (EC) function and lipids in a cSLE cohort longitudinally in comparison with healthy controls (HC). Next aim was to correlate these levels with Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) and nailfold capillaroscopic patterns. METHODS Blood serum samples, videocapillaroscopy images and patient characteristics were collected in a multicentre longitudinal cSLE cohort and from age and sex comparable HC. Disease activity was evaluated by SLEDAI. A total of 15 EC markers and six lipids were measured in two longitudinal cSLE samples (minimum interval of 6 months) and in HC. Nailfold videocapillaroscopy images were scored according to the guidelines from the EULAR Study Group on Microcirculation in Rheumatic Diseases. RESULTS In total, 47 patients with cSLE and 42 HCs were analysed. Median age at diagnosis was 15 years (IQR 12-16 years). Median time between t=1 and t=2 was 14.5 months (IQR 9-24 months). Median SLEDAI was 12 (IQR 6-18) at t=1 and 2 (IQR 1-4) at t=2. Serum levels of angiopoietin-2, CCL2, CXCL10, GAS6, pentraxin-3, thrombomodulin, VCAM-1 and vWF-A2 were elevated in cSLE compared with HC at t=1. While many elevated EC markers at t=1 normalised over time after treatment, several markers remained significantly increased compared with HC (angiopoietin-2, CCL2, CXCL10, GAS6, thrombomodulin and VCAM-1). CONCLUSION In serum from patients with cSLE different markers of endothelial activation were dysregulated. While most markers normalised during treatment, others remained elevated in a subset of patients, even during low disease activity. These results suggest a role for the dysregulated endothelium in early and later phases of cSLE, possibly also during lower disease activity. TRIAL REGISTRATION NUMBER NL60885.018.17.
Collapse
Affiliation(s)
- Sandy C Bergkamp
- Department of Paediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Centres (AUMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Nick D Bergkamp
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Mohamed Javad Wahadat
- Department of Paediatric Rheumatology, Sophia Children's Hospital, Erasmus University Medical Centre, Rotterdam, The Netherlands
- Department of Immunology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Mariken P Gruppen
- Department of Paediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Centres (AUMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Amara Nassar-Sheikh Rashid
- Department of Paediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Centres (AUMC), University of Amsterdam, Amsterdam, The Netherlands
- Department of Paediatrics, Zaans Medisch Centrum, Zaandam, The Netherlands
| | - Sander W Tas
- Amsterdam Rheumatology and Immunology Centre, Department of Rheumatology and Clinical Immunology, and Laboratory for Experimental Immunology, Amsterdam University Medical Centres (AUMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Martine J Smit
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Marjan A Versnel
- Department of Immunology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - J Merlijn van den Berg
- Department of Paediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Centres (AUMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Sylvia Kamphuis
- Department of Paediatric Rheumatology, Sophia Children's Hospital, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Dieneke Schonenberg-Meinema
- Department of Paediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Centres (AUMC), University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Brandt J, Ramly E, Lim SS, Bao G, Messina ML, Piper ME, Bartels CM. Implementing a Staff-Led Smoking Cessation Intervention in a Diverse Safety-Net Rheumatology Clinic: A Pre-Post Scalability Study in a Low-Resource Setting. Arthritis Care Res (Hoboken) 2024; 76:1342-1350. [PMID: 38622089 PMCID: PMC11349476 DOI: 10.1002/acr.25349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 03/21/2024] [Accepted: 04/11/2024] [Indexed: 04/17/2024]
Abstract
OBJECTIVE Quit Connect (QC), our specialty clinic smoking cessation intervention, supports clinic staff to check, advise, and connect willing patients to a state quit line or class. QC improved tobacco screening and quit line referrals 26-fold in a predominantly White academic health care system population. Implementing QC includes education, electronic health record (EHR) reminders, and periodic audit feedback. This study tested QC's feasibility and impact in a safety-net rheumatology clinic with a predominantly Black population. METHODS In this pre-post study, adult rheumatology visits were analyzed 12 months before through 18 months after QC intervention (November 2019 through November 2021, omitting COVID-19 peak April through November 2020). EHR data compared process and clinical outcomes, including offers, referrals to resources, completed referrals, and documented cessation. Clinic staff engaged in pre-post focus groups and questionnaires regarding intervention feasibility and acceptability. Cost-effectiveness was also assessed. RESULTS Visit-level patients who smoked were 89.8% Black and 69.5% women (n = 550). Before intervention, clinic staff rarely asked patients about readiness to cut back smoking (<10% assessment). After QC intervention, staff assessed quit readiness in 31.8% of visits with patients who smoked (vs 8.1% before); 58.9% of these patients endorsed readiness to cut back or quit. Of 102 accepting cessation services, 37% (n = 17) of those reached set a quit date. Staff found the intervention feasible and acceptable. Each quit attempt cost approximately $4 to $10. CONCLUSION In a safety-net rheumatology clinic with a predominantly Black population, QC improved tobacco screening, readiness-to-quit assessment, and referrals and was also feasible and cost-effective.
Collapse
Affiliation(s)
- Jennifer Brandt
- Emory University School of Medicine and Grady Health System, Atlanta, Georgia
| | - Edmond Ramly
- University of Wisconsin School of Medicine and Public Health and University of Wisconsin College of Engineering, Madison
- Indiana University School of Public Health, Bloomington, Indiana
| | - S Sam Lim
- Emory University School of Medicine and Grady Health System, Atlanta, Georgia
| | - Gaobin Bao
- Emory University School of Medicine, Atlanta, Georgia
| | | | - Megan E Piper
- UW Center for Tobacco Research and Intervention, Madison, Wisconsin
| | | |
Collapse
|
6
|
Nguyen AT, Curtis KM, Tepper NK, Kortsmit K, Brittain AW, Snyder EM, Cohen MA, Zapata LB, Whiteman MK. U.S. Medical Eligibility Criteria for Contraceptive Use, 2024. MMWR Recomm Rep 2024; 73:1-126. [PMID: 39106314 PMCID: PMC11315372 DOI: 10.15585/mmwr.rr7304a1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2024] Open
Abstract
The 2024 U.S. Medical Eligibility Criteria for Contraceptive Use (U.S. MEC) comprises recommendations for the use of specific contraceptive methods by persons who have certain characteristics or medical conditions. These recommendations for health care providers were updated by CDC after review of the scientific evidence and a meeting with national experts in Atlanta, Georgia, during January 25-27, 2023. The information in this report replaces the 2016 U.S. MEC (CDC. U.S. Medical Eligibility Criteria for Contraceptive Use, 2016. MMWR 2016:65[No. RR-3]:1-103). Notable updates include 1) the addition of recommendations for persons with chronic kidney disease; 2) revisions to the recommendations for persons with certain characteristics or medical conditions (i.e., breastfeeding, postpartum, postabortion, obesity, surgery, deep venous thrombosis or pulmonary embolism with or without anticoagulant therapy, thrombophilia, superficial venous thrombosis, valvular heart disease, peripartum cardiomyopathy, systemic lupus erythematosus, high risk for HIV infection, cirrhosis, liver tumor, sickle cell disease, solid organ transplantation, and drug interactions with antiretrovirals used for prevention or treatment of HIV infection); and 3) inclusion of new contraceptive methods, including new doses or formulations of combined oral contraceptives, contraceptive patches, vaginal rings, progestin-only pills, levonorgestrel intrauterine devices, and vaginal pH modulator. The recommendations in this report are intended to serve as a source of evidence-based clinical practice guidance for health care providers. The goals of these recommendations are to remove unnecessary medical barriers to accessing and using contraception and to support the provision of person-centered contraceptive counseling and services in a noncoercive manner. Health care providers should always consider the individual clinical circumstances of each person seeking contraceptive services. This report is not intended to be a substitute for professional medical advice for individual patients; when needed, patients should seek advice from their health care providers about contraceptive use.
Collapse
Affiliation(s)
- Antoinette T. Nguyen
- Division of Reproductive Health, National Center for
Chronic Disease Prevention and Health Promotion, CDC, Atlanta, Georgia
| | - Kathryn M. Curtis
- Division of Reproductive Health, National Center for
Chronic Disease Prevention and Health Promotion, CDC, Atlanta, Georgia
| | - Naomi K. Tepper
- Division of Reproductive Health, National Center for
Chronic Disease Prevention and Health Promotion, CDC, Atlanta, Georgia
| | - Katherine Kortsmit
- Division of Reproductive Health, National Center for
Chronic Disease Prevention and Health Promotion, CDC, Atlanta, Georgia
| | - Anna W. Brittain
- Division of Reproductive Health, National Center for
Chronic Disease Prevention and Health Promotion, CDC, Atlanta, Georgia
| | - Emily M. Snyder
- Division of Reproductive Health, National Center for
Chronic Disease Prevention and Health Promotion, CDC, Atlanta, Georgia
| | - Megan A. Cohen
- Division of Reproductive Health, National Center for
Chronic Disease Prevention and Health Promotion, CDC, Atlanta, Georgia
| | - Lauren B. Zapata
- Division of Reproductive Health, National Center for
Chronic Disease Prevention and Health Promotion, CDC, Atlanta, Georgia
| | - Maura K. Whiteman
- Division of Reproductive Health, National Center for
Chronic Disease Prevention and Health Promotion, CDC, Atlanta, Georgia
| |
Collapse
|
7
|
Choi MY, Guan H, Yoshida K, Paudel M, Kargere BA, Li D, Ellrodt J, Stevens E, Cai T, Weber BN, Everett BM, Costenbader KH. Personalizing cardiovascular risk prediction for patients with systemic lupus erythematosus. Semin Arthritis Rheum 2024; 67:152468. [PMID: 38788567 PMCID: PMC11214838 DOI: 10.1016/j.semarthrit.2024.152468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 04/12/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024]
Abstract
OBJECTIVE Cardiovascular disease (CVD) risk is increased in SLE and underestimated by general population prediction algorithms. We aimed to develop a novel SLE-specific prediction tool, SLECRISK, to provide a more accurate estimate of CVD risk in SLE. METHODS We studied patients in the Brigham and Women's Hospital SLE cohort. We collected one-year baseline data including the presence of traditional CVD factors and SLE-related features at cohort enrollment. Ten-year follow-up for the first major adverse cardiovascular event (MACE; myocardial infarction (MI), stroke, or cardiac death) began at day +1 following the baseline period (index date). ICD-9/10 codes identified MACE were adjudicated by board-certified cardiologists. Least absolute shrinkage and selection operator regression selected SLE-related variables to add to the American College of Cardiology/American Heart Association (ACC/AHA) Pooled Cohort Risk Equations 10-year risk Cox regression model. Model fit statistics and performance (sensitivity, specificity, positive/negative predictive value, c-statistic) for predicting moderate/high 10-year risk (≥7.5 %) of MACE were assessed and compared to ACC/AHA, Framingham risk score (FRS), and modified FRS (mFRS). Optimism adjustment internal validation was performed using bootstrapping. RESULTS We included 1,243 patients with 90 MACEs (46 MIs, 36 strokes, 19 cardiac deaths) over 8946.5 person-years of follow-up. SLE variables selected for the new prediction algorithm (SLECRISK) were SLE activity (remission/mild vs. moderate/severe), disease duration (years), creatinine (mg/dL), anti-dsDNA, anti-RNP, lupus anticoagulant, anti-Ro positivity, and low C4. The sensitivity for detecting moderate/high-risk (≥7.5 %) of MACE using SLECRISK was 0.74 (95 %CI: 0.65, 0.83), which was better than the sensitivity of the ACC/AHA model (0.38 (95 %CI: 0.28, 0.48)). It also identified 3.4-fold more moderate/high-risk patients than the ACC/AHA. Patients who were moderate/high-risk according to SLECRISK but not ACC/AHA, were more likely to be young women with severe SLE and few other traditional CVD risk factors. Model performance between SLECRISK, FRS, and mFRS were similar. CONCLUSION The novel SLECRISK tool is more sensitive than the ACC/AHA for predicting moderate/high 10-year risk for MACE and may be particularly useful in predicting risk for young females with severe SLE. Future external validation studies utilizing cohorts with more severe SLE are needed.
Collapse
Affiliation(s)
- May Y Choi
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Division of Rheumatology, University of Calgary, Calgary, Alberta, Canada.
| | - Hongshu Guan
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Kazuki Yoshida
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Misti Paudel
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Daniel Li
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jack Ellrodt
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Emma Stevens
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Tianrun Cai
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Brittany N Weber
- Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Brendan M Everett
- Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Karen H Costenbader
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
8
|
Al-Ewaidat OA, Naffaa MM. Deciphering Mechanisms, Prevention Strategies, Management Plans, Medications, and Research Techniques for Strokes in Systemic Lupus Erythematosus. MEDICINES (BASEL, SWITZERLAND) 2024; 11:15. [PMID: 39189161 PMCID: PMC11348055 DOI: 10.3390/medicines11070015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/30/2024] [Accepted: 07/26/2024] [Indexed: 08/28/2024]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune rheumatic condition characterized by an unpredictable course and a wide spectrum of manifestations varying in severity. Individuals with SLE are at an increased risk of cerebrovascular events, particularly strokes. These strokes manifest with a diverse range of symptoms that cannot be solely attributed to conventional risk factors, underscoring their significance among the atypical risk factors in the context of SLE. This complexity complicates the identification of optimal management plans and the selection of medication combinations for individual patients. This susceptibility is further complicated by the nuances of neuropsychiatric SLE, which reveals a diverse array of neurological symptoms, particularly those associated with ischemic and hemorrhagic strokes. Given the broad range of clinical presentations and associated risks linking strokes to SLE, ongoing research and comprehensive care strategies are essential. These efforts are critical for improving patient outcomes by optimizing management strategies and discovering new medications. This review aims to elucidate the pathological connection between SLE and strokes by examining neurological manifestations, risk factors, mechanisms, prediction and prevention strategies, management plans, and available research tools and animal models. It seeks to explore this medical correlation and discover new medication options that can be tailored to individual SLE patients at risk of stroke.
Collapse
Affiliation(s)
- Ola A. Al-Ewaidat
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA;
| | - Moawiah M. Naffaa
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
| |
Collapse
|
9
|
García-Villegas EA, Vargas-de-León C, Villa-Romero AR, Ibarra Valdovinos I, Márquez-González H. [Study of the metabolic syndrome severity index as a predictive factor of a major cardiovascular event in premenopausal women with systemic lupus erythematosus]. Med Clin (Barc) 2024; 163:62-69. [PMID: 38697892 DOI: 10.1016/j.medcli.2024.01.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND Patients with systemic lupus erythematosus (SLE) have an increased risk of metabolic syndrome (MS) and cardiovascular (CV) disease. MS is evaluated binary, limiting the understanding of each component's severity individually. Therefore, severity scores for MS that evaluate them separately have been developed. This study aims to determine the prognosis between MS severity and the occurrence of major adverse cardiovascular events (MACE) in SLE patients. METHODS Ten-year follow-up cohort study. Premenopausal>18-year-old women with a previous diagnosis of SLE were included. Patients with recent CV events, pregnancy, thyroid disease, and liposuction were excluded. The variables of interest were CV events; the confounding variables, and the MS severity indexes were examined. Hazard ratios and Kaplan-Meier survival curves were estimated through Cox regression. RESULTS A total of 238 women were analyzed: 22 presented MACE, and 216 did not. MS prevalence, measured according to consensus and ATP-III criteria, was higher in MACE patients (50 and 40,95%, respectively). The MetSx-IMC severity index was higher within the MACE group. Cox analysis showed an increase in the MetSx-IMC associated with the risk of suffering MACE in a 1.107 ratio. CONCLUSIONS The MetSx-IMC severity index, contrary to the binary approaches, is recommended to evaluate MS as a predictor of MACE in SLE patients. Offering improved and more accurate prognosis in patients at risk of developing MCE.
Collapse
Affiliation(s)
- Elsy Aidé García-Villegas
- Departamento de Vigilancia Epidemiológica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, México
| | - Cruz Vargas-de-León
- División de Investigación, Hospital Juárez de México, Ciudad de México, México; Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México, México
| | | | - Itzelly Ibarra Valdovinos
- Departamento de Vigilancia Epidemiológica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, México
| | - Horacio Márquez-González
- Investigación Clínica, Hospital Infantil de México Federico Gómez, Ciudad de México, México; Departamento de Cardiopatías Congénitas, Hospital de Cardiología Centro Médico Nacional Siglo XXI, Ciudad de México, México.
| |
Collapse
|
10
|
McCallinhart PE, Chade AR, Bender SB, Trask AJ. Expanding landscape of coronary microvascular disease in co-morbid conditions: Metabolic disease and beyond. J Mol Cell Cardiol 2024; 192:26-35. [PMID: 38734061 PMCID: PMC11340124 DOI: 10.1016/j.yjmcc.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/26/2024] [Accepted: 05/08/2024] [Indexed: 05/13/2024]
Abstract
Coronary microvascular disease (CMD) and impaired coronary blood flow control are defects that occur early in the pathogenesis of heart failure in cardiometabolic conditions, prior to the onset of atherosclerosis. In fact, recent studies have shown that CMD is an independent predictor of cardiac morbidity and mortality in patients with obesity and metabolic disease. CMD is comprised of functional, structural, and mechanical impairments that synergize and ultimately reduce coronary blood flow in metabolic disease and in other co-morbid conditions, including transplant, autoimmune disorders, chemotherapy-induced cardiotoxicity, and remote injury-induced CMD. This review summarizes the contemporary state-of-the-field related to CMD in metabolic and these other co-morbid conditions based on mechanistic data derived mostly from preclinical small- and large-animal models in light of available clinical evidence and given the limitations of studying these mechanisms in humans. In addition, we also discuss gaps in current understanding, emerging areas of interest, and opportunities for future investigations in this field.
Collapse
Affiliation(s)
- Patricia E McCallinhart
- Center for Cardiovascular Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States of America
| | - Alejandro R Chade
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, United States of America; Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States of America
| | - Shawn B Bender
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States of America; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States of America; Research Service, Harry S Truman Memorial Veterans Hospital, Columbia, MO, United States of America.
| | - Aaron J Trask
- Center for Cardiovascular Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States of America; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, United States of America.
| |
Collapse
|
11
|
Akiash N, Abbaspour S, Mowla K, Moradi A, Madjidi S, Sharifi P, Pazoki M. Three-dimensional speckle tracking echocardiography for evaluation of ventricular function in patients with systemic lupus erythematosus: relationship between duration of lupus erythematosus and left ventricular dysfunction by using global longitudinal strain. Egypt Heart J 2024; 76:79. [PMID: 38914877 PMCID: PMC11196547 DOI: 10.1186/s43044-024-00511-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 06/19/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND Cardiovascular diseases are leading causes of morbidity and mortality in patients with systemic lupus erythematosus (SLE). Cardiac involvement in SLE can often go undetected. Three-dimensional (3D) speckle tracking echocardiography (STE) is a noninvasive imaging technique that can assess the function of the heart's ventricles in an accurate and reproducible way. This makes it an attractive option for detecting early signs of heart disease in SLE patients. By identifying these subclinical cardiac abnormalities, 3D-STE may help reduce the negative impact of cardiovascular diseases in SLE population. Therefore, this study aimed to compare the left ventricular (LV) function between patients with SLE compared to age- and gender-matched controls using two-dimensional (2D) and 3D-STE. RESULTS The current study found no significant differences in left ventricle ejection fraction, left ventricle end-diastolic volume, left ventricle end-systolic volume, left ventricle end-diastolic mass, and left ventricle end-systolic mass between the two groups. However, the SLE group exhibited a significantly lower LV global longitudinal strain (GLS) compared to the control group according to all types of echocardiographic assessments, including 3D and 2D long-axis strain, apical 2-chamber, and apical 4-chamber assessments (all P values < 0.05). Furthermore, a good inter-rater reliability and intra-rater reliability were observed regarding the LVGLS measurement with 3D-STE. Additionally, the study identified a significant correlation between LVGLS and SLE duration (r (50) = 0.46, P < 0.001). The use of prednisolone and nephrology disorders was also found to impact LVGLS measurements. CONCLUSIONS Despite a normal LVEF in patients with SLE, LVGLS measurements indicated that LV systolic dysfunction was observed more frequently in SLE patients compared to their healthy counterparts. Therefore, advanced 3D-STE techniques may be useful in identifying subtle abnormalities in LV function in SLE patients.
Collapse
Affiliation(s)
- Nehzat Akiash
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Golestan Blvd., Ahvaz, Iran
| | - Somayeh Abbaspour
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Golestan Blvd., Ahvaz, Iran
| | - Karim Mowla
- Department of Rheumatology, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Amir Moradi
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Golestan Blvd., Ahvaz, Iran.
| | | | - Parisa Sharifi
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Golestan Blvd., Ahvaz, Iran
| | - Mahboubeh Pazoki
- Department of Cardiology, School of Medicine, Hazarat-e Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Ang SP, Chia JE, Misra K, Krittanawong C, Iglesias J, Gewirtz D, Mukherjee D. Autoimmune Rheumatic Diseases and Outcomes Following Percutaneous Coronary Intervention: A Systematic Review and Meta-Analysis. Angiology 2024:33197241255167. [PMID: 38771845 DOI: 10.1177/00033197241255167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
Autoimmune Rheumatic Diseases (AIRDs) are associated with increased cardiovascular mortality. However, the post-percutaneous coronary intervention (PCI) outcomes in this population present a research gap, given the limited and discordant findings in existing studies. We conducted a systematic review and meta-analysis to assess the relationship between AIRDs and clinical outcomes after PCI; 9 studies with 7,027,270 patients (126,914 with AIRD, 6,900,356 without AIRD) were included. The AIRD cohort was characterized by an older age, a predominantly female demographic, and a greater prevalence of hypertension and diabetes mellitus. Over a mean follow-up period of 4.6 ± 3.5 years, AIRD patients demonstrated significantly higher odds of all-cause mortality (odds ratio (OR) 1.45, 95% CI: 1.25-1.78, P < .001) and major adverse cardiovascular events (MACE) (OR 1.63, 95% CI: 1.01-2.62, P = .04) compared with non-AIRD patients. Sensitivity analysis using adjusted estimates, confirmed the higher all-cause mortality (hazard ratio 1.32, 95% CI: 1.05-1.64, P = .01). Patients with rheumatoid arthritis had a significantly elevated odds of all-cause mortality (OR 1.50, 95% CI: 1.27-1.77) and MACE (OR 1.18, 95% CI: 1.14-1.21). Our study demonstrated an association between AIRDs and suboptimal long-term outcomes post-PCI. Prospective studies are warranted to explore the risk factors of unfavorable prognoses in patients with AIRDs.
Collapse
Affiliation(s)
- Song Peng Ang
- Department of Internal Medicine, Rutgers Health/Community Medical Center, Toms River, NJ, USA
| | - Jia Ee Chia
- Department of Internal Medicine, Texas Tech University Health Science Center, El Paso, TX, USA
| | - Kanchan Misra
- Department of Radiology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | | | - Jose Iglesias
- Department of Internal Medicine, Rutgers Health/Community Medical Center, Toms River, NJ, USA
- Department of Internal Medicine, Hackensack Meridian School of Medicine, Nutley, NJ, USA
| | - Daniel Gewirtz
- Department of Internal Medicine, Rutgers Health/Community Medical Center, Toms River, NJ, USA
| | - Debabrata Mukherjee
- Department of Internal Medicine, Texas Tech University Health Science Center, El Paso, TX, USA
- Department of Cardiovascular Medicine, Texas Tech University Health Science Center, El Paso, TX, USA
| |
Collapse
|
13
|
Liang R, Xu H, Yao R, Pei W, Wang Z, Liang R, Han X, Zhou Y, An Y, Su Y. A predictive model for premature atherosclerosis in systemic lupus erythematosus based on clinical characteristics. Clin Rheumatol 2024; 43:1541-1550. [PMID: 38565803 DOI: 10.1007/s10067-024-06934-3] [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: 12/29/2023] [Revised: 02/18/2024] [Accepted: 03/10/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is associated with a significant risk of atherosclerotic cardiovascular disease, especially in the development of premature atherosclerosis. Specific prediction models for premature atherosclerosis in SLE patients are still limited. The objective of this study was to establish a predictive model for premature atherosclerosis in SLE. METHOD The study collected clinical and laboratory data from 148 SLE patients under the age of 55, between January 2021 and June 2023. The least absolute shrinkage and selection operator logistic regression model was utilized to identify potentially relevant features. Subsequently, a nomogram was developed using multivariable logistic analysis. The performance of the nomogram was evaluated through a receiver-operating characteristic curve, calibration curve, and decision curve analysis (DCA). RESULTS A total of 148 SLE patients who fulfilled the inclusion criteria were enrolled in the study, of whom 53 patients (35.81%) met the definition of premature atherosclerosis. Hypertension, antiphospholipid syndrome, azathioprine use, duration of glucocorticoid, and age of patients were included in the multivariable regression. The nomogram, based on the non-overfitting multivariable model, was internally validated and demonstrated sufficient clinical utility for assessing the risk of premature atherosclerosis (area under curve: 0.867). CONCLUSIONS The comprehensive nomogram constructed in this study serves as a useful and convenient tool for evaluating the risk of premature atherosclerosis in SLE patients. It is helpful for clinicians to early identify SLE patients with premature atherosclerosis and facilitates the implementation of more effective preventive measures. Key Points • SLE patients are at a significantly higher risk of developing premature atherosclerosis compared to the general population, and this risk persists even in cases with low disease activity. Traditional models used to evaluate and predict premature atherosclerosis in SLE patients often underestimate the risk. • This study establishes a comprehensive and visually orientated predictive model of premature atherosclerosis in SLE patients, based on clinical characteristics. • The scoring system allows for convenient and effective prediction of individual incidence of premature atherosclerosis, and could provide valuable information for identification and making further intervention decision.
Collapse
Affiliation(s)
- Ruyu Liang
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China
| | - Haojie Xu
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China
| | - Ranran Yao
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China
| | - Wenwen Pei
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China
| | - Ziye Wang
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China
| | - Renge Liang
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China
| | - Xiao Han
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China
| | - Yunshan Zhou
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China
| | - Yuan An
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China.
| | - Yin Su
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China.
- Peking University People's Hospital, Qingdao, China.
| |
Collapse
|
14
|
Koletsos N, Lazaridis A, Triantafyllou A, Anyfanti P, Lamprou S, Stoimeni A, Papadopoulos NG, Koravou EE, Gkaliagkousi E. Accumulation of Microvascular Target Organ Damage in Systemic Lupus Erythematosus Patients Is Associated with Increased Cardiovascular Risk. J Clin Med 2024; 13:2140. [PMID: 38610905 PMCID: PMC11012611 DOI: 10.3390/jcm13072140] [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: 03/12/2024] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Background: Systemic lupus erythematosus (SLE) is a prototype autoimmune disease associated with increased cardiovascular (CV) burden. Besides increased arterial stiffness and subclinical atherosclerosis, microvascular dysfunction is considered an important component in the pathophysiology of CV disease. However, there is a lack of data regarding the effect of multiple target organ damage (TOD) on CV health. Objectives: This study aimed to evaluate (i) the presence of microvascular changes in SLE in various vascular beds, (ii) the possible associations between the accumulation of microvascular TOD and CV risk and (iii) whether Galectin-3 represents a predictor of combined microvascular TOD. Methods: Participants underwent (i) evaluation of skin microvascular perfusion (laser speckle contrast analysis), (ii) fundoscopy (non-mydriatic fundus camera), (iii) indirect assessment of myocardial perfusion (subendocardial viability ratio) and (iv) determination of urine albumin-to-creatinine ratio (UACR). CV risk was calculated using the QResearch Risk Estimator version 3 (QRISK3). Serum Galectin-3 levels were determined. Results: Forty-seven SLE patients and fifty controls were studied. SLE patients demonstrated impaired skin microvascular reactivity (160.2 ± 41.0 vs. 203.6 ± 40.1%), retinal arteriolar narrowing (88.1 ± 11.1 vs. 94.6 ± 13.5 μm) and higher UACR levels compared to controls. Furthermore, SLE individuals had significantly higher Galectin-3 levels [21.5(6.1) vs. 6.6(6.6) ng/dL], QRISK3 scores [7.0(8.6) vs. 1.3(3.6)%] and a greater chance for microvascular dysfunction. In the SLE group, patients with multiple TOD exhibited higher QRISK3. In the multivariate analysis, the accumulation of TOD correlated with disease activity and Galectin-3 (p < 0.05). Conclusions: Our study showed for the first time that SLE patients exhibit a greater number of cases of TOD. The accumulation of TOD was associated with increased CV risk. Clinicians dealing with SLE should be aware and seek microvascular alterations.
Collapse
Affiliation(s)
- Nikolaos Koletsos
- 3rd Department of Internal Medicine, Papageorgiou General Hospital, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece; (A.L.); (A.T.); (S.L.); (A.S.); (E.G.)
| | - Antonios Lazaridis
- 3rd Department of Internal Medicine, Papageorgiou General Hospital, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece; (A.L.); (A.T.); (S.L.); (A.S.); (E.G.)
| | - Areti Triantafyllou
- 3rd Department of Internal Medicine, Papageorgiou General Hospital, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece; (A.L.); (A.T.); (S.L.); (A.S.); (E.G.)
| | - Panagiota Anyfanti
- Second Medical Department, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| | - Stamatina Lamprou
- 3rd Department of Internal Medicine, Papageorgiou General Hospital, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece; (A.L.); (A.T.); (S.L.); (A.S.); (E.G.)
| | - Anastasia Stoimeni
- 3rd Department of Internal Medicine, Papageorgiou General Hospital, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece; (A.L.); (A.T.); (S.L.); (A.S.); (E.G.)
| | | | | | - Eugenia Gkaliagkousi
- 3rd Department of Internal Medicine, Papageorgiou General Hospital, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece; (A.L.); (A.T.); (S.L.); (A.S.); (E.G.)
| |
Collapse
|
15
|
Ambler WG, Kaplan MJ. Vascular damage in systemic lupus erythematosus. Nat Rev Nephrol 2024; 20:251-265. [PMID: 38172627 PMCID: PMC11391830 DOI: 10.1038/s41581-023-00797-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2023] [Indexed: 01/05/2024]
Abstract
Vascular disease is a major cause of morbidity and mortality in patients with systemic autoimmune diseases, particularly systemic lupus erythematosus (SLE). Although comorbid cardiovascular risk factors are frequently present in patients with SLE, they do not explain the high burden of premature vascular disease. Profound innate and adaptive immune dysregulation seems to be the primary driver of accelerated vascular damage in SLE. In particular, evidence suggests that dysregulation of type 1 interferon (IFN-I) and aberrant neutrophils have key roles in the pathogenesis of vascular damage. IFN-I promotes endothelial dysfunction directly via effects on endothelial cells and indirectly via priming of immune cells that contribute to vascular damage. SLE neutrophils are vasculopathic in part because of their increased ability to form immunostimulatory neutrophil extracellular traps. Despite improvements in clinical care, cardiovascular disease remains the leading cause of mortality among patients with SLE, and treatments that improve vascular outcomes are urgently needed. Improved understanding of the mechanisms of vascular injury in inflammatory conditions such as SLE could also have implications for common cardiovascular diseases, such as atherosclerosis and hypertension, and may ultimately lead to personalized therapeutic approaches to the prevention and treatment of this potentially fatal complication.
Collapse
Affiliation(s)
- William G Ambler
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
16
|
Corona-Meraz FI, Vázquez-Del Mercado M, Sandoval-García F, Robles-De Anda JA, Tovar-Cuevas AJ, Rosales-Gómez RC, Guzmán-Ornelas MO, González-Inostroz D, Peña-Nava M, Martín-Márquez BT. Biomarkers in Systemic Lupus Erythematosus along with Metabolic Syndrome. J Clin Med 2024; 13:1988. [PMID: 38610754 PMCID: PMC11012563 DOI: 10.3390/jcm13071988] [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: 02/29/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Metabolic syndrome (MetS) is a group of physiological abnormalities characterized by obesity, insulin resistance (IR), and hypertriglyceridemia, which carry the risk of developing cardiovascular disease (CVD) and type 2 diabetes (T2D). Immune and metabolic alterations have been observed in MetS and are associated with autoimmune development. Systemic lupus erythematosus (SLE) is an autoimmune disease caused by a complex interaction of environmental, hormonal, and genetic factors and hyperactivation of immune cells. Patients with SLE have a high prevalence of MetS, in which elevated CVD is observed. Among the efforts of multidisciplinary healthcare teams to make an early diagnosis, a wide variety of factors have been considered and associated with the generation of biomarkers. This review aimed to elucidate some primary biomarkers and propose a set of assessments to improve the projection of the diagnosis and evolution of patients. These biomarkers include metabolic profiles, cytokines, cardiovascular tests, and microRNAs (miRs), which have been observed to be dysregulated in these patients and associated with outcomes.
Collapse
Affiliation(s)
- Fernanda Isadora Corona-Meraz
- Multidisciplinary Health Research Center, Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara 45425, Jalisco, Mexico; (A.-J.T.-C.); (R.-C.R.-G.); (M.-O.G.-O.)
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
| | - Mónica Vázquez-Del Mercado
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
- Rheumatology Service, Internal Medicine Division, Civil Hospital of Guadalajara “Dr. Juan I. Menchaca”, Guadalajara 44340, Jalisco, Mexico
- Academic Group UDG-CA-703, “Immunology and Rheumatology”, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Flavio Sandoval-García
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
- Academic Group UDG-CA-703, “Immunology and Rheumatology”, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Jesus-Aureliano Robles-De Anda
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
| | - Alvaro-Jovanny Tovar-Cuevas
- Multidisciplinary Health Research Center, Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara 45425, Jalisco, Mexico; (A.-J.T.-C.); (R.-C.R.-G.); (M.-O.G.-O.)
| | - Roberto-Carlos Rosales-Gómez
- Multidisciplinary Health Research Center, Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara 45425, Jalisco, Mexico; (A.-J.T.-C.); (R.-C.R.-G.); (M.-O.G.-O.)
| | - Milton-Omar Guzmán-Ornelas
- Multidisciplinary Health Research Center, Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara 45425, Jalisco, Mexico; (A.-J.T.-C.); (R.-C.R.-G.); (M.-O.G.-O.)
| | - Daniel González-Inostroz
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
| | - Miguel Peña-Nava
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
| | - Beatriz-Teresita Martín-Márquez
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
- Academic Group UDG-CA-703, “Immunology and Rheumatology”, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
17
|
Noroozi Gilandehi S, Wong KH, Francis T, Wright MA, Lord J, Stehlik J, Kemeyou L, Smith T, Clardy SL. Cardiac Involvement in Neurosarcoidosis: A Single-Center Investigation. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200196. [PMID: 38181319 DOI: 10.1212/nxi.0000000000200196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 11/06/2023] [Indexed: 01/07/2024]
Abstract
BACKGROUND AND OBJECTIVES Sarcoidosis is a multisystem inflammatory granulomatous disease. Among systemic sarcoidosis manifestations, cardiac or nervous system involvement can result in significant morbidity and mortality. We describe the overlapping incidence of cardiac sarcoidosis (CS) within a neurosarcoidosis (NS) cohort and determine the frequency of other nonsarcoid cardiac diseases in these patients. METHODS We performed a retrospective chart review of patients evaluated at the University of Utah from 2010 to 2022. Patients were included if they had (1) at least one instance of a diagnostic code for sarcoidosis in their medical record-International Classification of Diseases (ICD) 9 code 135 or ICD 10 code D86; (2) at least one outpatient visit in the Neurology Department within the University of Utah electronic health record with a diagnosis of definite, probable, or possible NS based on 2018 consensus criteria; (3) at least one outpatient visit in the Cardiology Department within the University of Utah electronic health record; and (4) ECG available in their medical record for review. Of 64 definite, probable, or possible patients with NS in the University of Utah cohort, 52 met our inclusion criteria and were included in this study. RESULTS Of 52 patients with NS who met our inclusion criteria, 65.38% were female, with an average age of 60.9 years (range 38-84). More than half (58%) were obese (BMI ≥ 30). CS was diagnosed in 6 patients with NS (12%). Symptoms suggestive of possible cardiac dysfunction included lower extremity edema (50%), palpitations (46%), chest pain (44%), and shortness of breath (27%). ECG abnormalities included nonspecific T-wave change (40%) and right bundle branch block (17%). Three patients experienced ventricular tachycardia: sustained in one patient and nonsustained in 2 patients. Cardiac MRI was performed in 17 patients (32.7%) and in 3 patients (17.6%), which revealed diffuse myocardial enhancement suggesting CS. DISCUSSION In this cohort, 12% of patients with NS also had confirmed CS. In addition, these patients had a high burden of cardiovascular disease not directly attributed to sarcoidosis. Our data suggest that patients with NS require comprehensive cardiac evaluation. Future studies are needed to clarify the extent of the direct contribution of granulomatous inflammation on the cardiovascular system from the indirect contribution of treatments such as glucocorticoids that lead to increased risk of cardiovascular disease in sarcoidosis.
Collapse
Affiliation(s)
- Sama Noroozi Gilandehi
- From the Department of Neurology (S.N.G., K.-H.W., T.F., M.A.W., J.L., T.S., S.L.C.); Division of Cardiovascular Medicine (J.S., L.K.), Department of Internal Medicine, University of Utah; Department of Neurology (J.S., L.K., T.S., S.L.C.); Geriatric Research Education and Clinical Center (T.S.), George E. Whalen Department of Veterans Affairs Medical Center; and Department of Pathology (T.S.), University of Utah, Salt Lake City
| | - Ka-Ho Wong
- From the Department of Neurology (S.N.G., K.-H.W., T.F., M.A.W., J.L., T.S., S.L.C.); Division of Cardiovascular Medicine (J.S., L.K.), Department of Internal Medicine, University of Utah; Department of Neurology (J.S., L.K., T.S., S.L.C.); Geriatric Research Education and Clinical Center (T.S.), George E. Whalen Department of Veterans Affairs Medical Center; and Department of Pathology (T.S.), University of Utah, Salt Lake City
| | - Trieste Francis
- From the Department of Neurology (S.N.G., K.-H.W., T.F., M.A.W., J.L., T.S., S.L.C.); Division of Cardiovascular Medicine (J.S., L.K.), Department of Internal Medicine, University of Utah; Department of Neurology (J.S., L.K., T.S., S.L.C.); Geriatric Research Education and Clinical Center (T.S.), George E. Whalen Department of Veterans Affairs Medical Center; and Department of Pathology (T.S.), University of Utah, Salt Lake City
| | - Melissa A Wright
- From the Department of Neurology (S.N.G., K.-H.W., T.F., M.A.W., J.L., T.S., S.L.C.); Division of Cardiovascular Medicine (J.S., L.K.), Department of Internal Medicine, University of Utah; Department of Neurology (J.S., L.K., T.S., S.L.C.); Geriatric Research Education and Clinical Center (T.S.), George E. Whalen Department of Veterans Affairs Medical Center; and Department of Pathology (T.S.), University of Utah, Salt Lake City
| | - Jennifer Lord
- From the Department of Neurology (S.N.G., K.-H.W., T.F., M.A.W., J.L., T.S., S.L.C.); Division of Cardiovascular Medicine (J.S., L.K.), Department of Internal Medicine, University of Utah; Department of Neurology (J.S., L.K., T.S., S.L.C.); Geriatric Research Education and Clinical Center (T.S.), George E. Whalen Department of Veterans Affairs Medical Center; and Department of Pathology (T.S.), University of Utah, Salt Lake City
| | - Josef Stehlik
- From the Department of Neurology (S.N.G., K.-H.W., T.F., M.A.W., J.L., T.S., S.L.C.); Division of Cardiovascular Medicine (J.S., L.K.), Department of Internal Medicine, University of Utah; Department of Neurology (J.S., L.K., T.S., S.L.C.); Geriatric Research Education and Clinical Center (T.S.), George E. Whalen Department of Veterans Affairs Medical Center; and Department of Pathology (T.S.), University of Utah, Salt Lake City
| | - Line Kemeyou
- From the Department of Neurology (S.N.G., K.-H.W., T.F., M.A.W., J.L., T.S., S.L.C.); Division of Cardiovascular Medicine (J.S., L.K.), Department of Internal Medicine, University of Utah; Department of Neurology (J.S., L.K., T.S., S.L.C.); Geriatric Research Education and Clinical Center (T.S.), George E. Whalen Department of Veterans Affairs Medical Center; and Department of Pathology (T.S.), University of Utah, Salt Lake City
| | - Tammy Smith
- From the Department of Neurology (S.N.G., K.-H.W., T.F., M.A.W., J.L., T.S., S.L.C.); Division of Cardiovascular Medicine (J.S., L.K.), Department of Internal Medicine, University of Utah; Department of Neurology (J.S., L.K., T.S., S.L.C.); Geriatric Research Education and Clinical Center (T.S.), George E. Whalen Department of Veterans Affairs Medical Center; and Department of Pathology (T.S.), University of Utah, Salt Lake City
| | - Stacey L Clardy
- From the Department of Neurology (S.N.G., K.-H.W., T.F., M.A.W., J.L., T.S., S.L.C.); Division of Cardiovascular Medicine (J.S., L.K.), Department of Internal Medicine, University of Utah; Department of Neurology (J.S., L.K., T.S., S.L.C.); Geriatric Research Education and Clinical Center (T.S.), George E. Whalen Department of Veterans Affairs Medical Center; and Department of Pathology (T.S.), University of Utah, Salt Lake City
| |
Collapse
|
18
|
Yennemadi AS, Jordan N, Diong S, Keane J, Leisching G. The Link Between Dysregulated Immunometabolism and Vascular Damage: Implications for the Development of Atherosclerosis in Systemic Lupus Erythematosus and Other Rheumatic Diseases. J Rheumatol 2024; 51:234-241. [PMID: 38224981 DOI: 10.3899/jrheum.2023-0833] [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] [Accepted: 11/30/2023] [Indexed: 01/17/2024]
Abstract
A bimodal pattern of mortality in systemic lupus erythematosus (SLE) exists. Early-stage deaths are predominantly caused by infection, whereas later-stage deaths are mainly caused by atherosclerotic disease. Further, although SLE-related mortality has reduced considerably in recent years, cardiovascular (CV) events remain one of the leading causes of death in people with SLE. Accelerated atherosclerosis in SLE is attributed to both an increase in traditional CV risk factors and the inflammatory effects of SLE itself. Many of these changes occur within the microenvironment of the vascular-immune interface, the site of atherosclerotic plaque development. Here, an intimate interaction between endothelial cells, vascular smooth muscle cells, and immune cells dictates physiological vs pathological responses to a chronic type 1 interferon environment. Low-density neutrophils (LDNs) have also been implicated in eliciting vasculature-damaging effects at such lesion sites. These changes are thought to be governed by dysfunctional metabolism of immune cells in this niche due at least in part to the chronic induction of type 1 interferons. Understanding these novel pathophysiological mechanisms and metabolic pathways may unveil potential innovative pharmacological targets and therapeutic opportunities for atherosclerosis, as well as shed light on the development of premature atherosclerosis in patients with SLE who develop CV events.
Collapse
Affiliation(s)
- Anjali S Yennemadi
- A.S. Yennemadi, MSc, J. Keane, MD, G. Leisching, PhD, TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, School of Medicine, Trinity College Dublin, University of Dublin
| | - Natasha Jordan
- N. Jordan, PhD, Department of Rheumatology, St. James's Hospital
| | - Sophie Diong
- S. Diong, MD, Department of Dermatology, St. James's Hospital, Dublin, Ireland
| | - Joseph Keane
- A.S. Yennemadi, MSc, J. Keane, MD, G. Leisching, PhD, TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, School of Medicine, Trinity College Dublin, University of Dublin
| | - Gina Leisching
- A.S. Yennemadi, MSc, J. Keane, MD, G. Leisching, PhD, TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, School of Medicine, Trinity College Dublin, University of Dublin;
| |
Collapse
|
19
|
Huang YC, Lai ECC, Liao TC, Weng MY. Evaluating the risk of ischemic stroke at a young age in patients with autoimmune inflammatory rheumatic diseases: a population-based cohort study in Taiwan. Front Immunol 2024; 15:1272557. [PMID: 38404587 PMCID: PMC10884215 DOI: 10.3389/fimmu.2024.1272557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/22/2024] [Indexed: 02/27/2024] Open
Abstract
Background Recent studies have demonstrated an increased incidence of ischemic stroke among patients with certain autoimmune inflammatory rheumatic diseases (AIIRDs). However, the associations between young stroke and AIIRDs have not been fully investigated. This study aimed to evaluate the risk of ischemic stroke among young patients with AIIRDs. Methods The National Health Insurance Research Database in Taiwan was utilized to establish cohorts of patients with AIIRDs diagnosed between 2004 and 2015, who were compared with 1,000,000 control participants. Cox proportional hazards regression models were used to calculate the hazard ratio of ischemic stroke and young ischemic stroke for individual AIIRDs after adjustment for relative risk factors. Results During the study period, a total of 64,120 patients with AIIRDss and 1,000,000 control patients were identified. The overall mean follow-up time was 5.33 years. There were 223 (0.8%) and 1,923 (0.3%) young ischemic stroke-related hospitalizations among patients with AIIRDs and controls, respectively. The incidence rate of young ischemic stroke was 0.08 in patients with rheumatoid arthritis, 0.08 in patients with Sjögren's syndrome, 0.26 in patients with systemic lupus erythematosus, 0.17 in patients with idiopathic inflammatory myositis, 0.24 in patients with systemic sclerosis, 0.05 in patients with Behçet's disease, and 0.44 in patients with systemic vasculitis, versus 0.05 per 100 person-years in the general population. The adjusted hazard ratios for young ischemic stroke were 1.07 (95% CI 0.70-1.43) for rheumatoid arthritis, 1.39 (95% CI 0.94-2.06) for Sjögren's syndrome, 5.79 (95% CI 4.68-7.17) for systemic lupus erythematosus, 2.07 for idiopathic inflammatory myositis (95% CI 0.98-4.38), 2.79 for systemic sclerosis (95% CI 1.38-5.63), 0.82 for Behçet's disease (95% CI 0.26-2.55), and 4.15 (95% CI 1.96-8.82) for systemic vasculitis. Conclusions Patients younger than 50 years with systemic lupus erythematosus, systemic sclerosis, or systemic vasculitis have a significantly elevated risk of developing ischemic stroke. Further research is needed to elucidate the pathogenesis of accelerated atherosclerosis in these AIIRDs.
Collapse
Affiliation(s)
- Ya-Chun Huang
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Edward Chia-Cheng Lai
- School of Pharmacy, Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Chi Liao
- School of Pharmacy, Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Meng-Yu Weng
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
20
|
Rafael-Vidal C, Martínez-Ramos S, Malvar-Fernández B, Altabás-González I, Mouriño C, Veale DJ, Floudas A, Fearon U, Reigosa JMP, García S. Type I Interferons induce endothelial destabilization in Systemic Lupus Erythematosus in a Tie2-dependent manner. Front Immunol 2023; 14:1277267. [PMID: 38162654 PMCID: PMC10756137 DOI: 10.3389/fimmu.2023.1277267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/29/2023] [Indexed: 01/03/2024] Open
Abstract
Endothelial cell (EC) dysfunction is a hallmark of Systemic Lupus Erythematosus (SLE) and Tie2 is a receptor essential for vascular stability. Inflammatory processes promote inhibition of Tie2 homeostatic activation, driving vascular dysfunction. In this work we determined whether type I Interferons (IFN) induce Tie2 signalling-mediated endothelial dysfunction in patients with SLE. Serum levels of Angiopoietin (Ang)-1, Ang-2 and soluble (s)Tie1 in patients with SLE and healthy controls were measured by ELISA. Monocytes from patients with SLE and Human Umbilical Vein EC (HUVEC) were stimulated with IFN-α, IFN-β (1000 I.U.) or SLE serum (20%). mRNA and protein expression, phosphorylation and translocation were determined by quantitative PCR, ELISA, Western Blot, flow cytometry and confocal microscopy. Viability and angiogenic capacity were determined by calcein and tube formation assays. We found that sTie1 and Ang-2 serum levels were increased and Ang-1 decreased in patients with SLE and were associated with clinical characteristics. Type I IFN significantly decreased Ang-1 and increased Ang-2 in monocytes from patients with SLE. Type I IFN increased sTie1 and Ang-2 secretion and reduced Tie2 activation in HUVEC. Functionally, type I IFN significantly reduced EC viability and impaired angiogenesis in a Tie2 signalling-dependent manner. Finally, SLE serum increased Ang-2 and sTie1 secretion and significantly decreased tube formation. Importantly, Tie1 and IFNAR1 knockdown reversed these effects in tube formation. Overall, type I IFN play an important role in the stability of EC by inhibiting Tie2 signalling, suggesting that these processes may be implicated in the cardiovascular events observed in patients with SLE.
Collapse
Affiliation(s)
- Carlos Rafael-Vidal
- Rheumatology and Immune-mediated Diseases Group, Galicia Sur Health Research Institute (IIS Galicia Sur), Vigo, Spain
- Rheumatology Department, University Hospital of Vigo, Vigo, Spain
| | - Sara Martínez-Ramos
- Rheumatology and Immune-mediated Diseases Group, Galicia Sur Health Research Institute (IIS Galicia Sur), Vigo, Spain
- Rheumatology Department, University Hospital of Vigo, Vigo, Spain
| | - Beatriz Malvar-Fernández
- Rheumatology and Immune-mediated Diseases Group, Galicia Sur Health Research Institute (IIS Galicia Sur), Vigo, Spain
- Rheumatology Department, University Hospital of Vigo, Vigo, Spain
| | - Irene Altabás-González
- Rheumatology and Immune-mediated Diseases Group, Galicia Sur Health Research Institute (IIS Galicia Sur), Vigo, Spain
- Rheumatology Department, University Hospital of Vigo, Vigo, Spain
| | - Coral Mouriño
- Rheumatology and Immune-mediated Diseases Group, Galicia Sur Health Research Institute (IIS Galicia Sur), Vigo, Spain
| | - Douglas J. Veale
- Molecular Rheumatology, Clinical Medicine, Trinity Biomedical Science Institute, Dublin, Ireland
- European Alliance of Associations for Rheumatology (EULAR) Centre for Arthritis and Rheumatic Diseases, St Vincent’s University Hospital, University College Dublin, Dublin, Ireland
| | | | - Ursula Fearon
- Molecular Rheumatology, Clinical Medicine, Trinity Biomedical Science Institute, Dublin, Ireland
- European Alliance of Associations for Rheumatology (EULAR) Centre for Arthritis and Rheumatic Diseases, St Vincent’s University Hospital, University College Dublin, Dublin, Ireland
| | - José María Pego Reigosa
- Rheumatology and Immune-mediated Diseases Group, Galicia Sur Health Research Institute (IIS Galicia Sur), Vigo, Spain
- Rheumatology Department, University Hospital of Vigo, Vigo, Spain
| | - Samuel García
- Rheumatology and Immune-mediated Diseases Group, Galicia Sur Health Research Institute (IIS Galicia Sur), Vigo, Spain
- Rheumatology Department, University Hospital of Vigo, Vigo, Spain
| |
Collapse
|
21
|
Miñano S, González-Correa C, Moleón J, Duarte J. Metabolic Modulators in Cardiovascular Complications of Systemic Lupus Erythematosus. Biomedicines 2023; 11:3142. [PMID: 38137363 PMCID: PMC10741086 DOI: 10.3390/biomedicines11123142] [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: 10/20/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a multifactorial disorder with contributions from hormones, genetics, and the environment, predominantly affecting young women. Cardiovascular disease is the primary cause of mortality in SLE, and hypertension is more prevalent among SLE patients. The dysregulation of both innate and adaptive immune cells in SLE, along with their infiltration into kidney and vascular tissues, is a pivotal factor contributing to the cardiovascular complications associated with SLE. The activation, proliferation, and differentiation of CD4+ T cells are intricately governed by cellular metabolism. Numerous metabolic inhibitors have been identified to target critical nodes in T cell metabolism. This review explores the existing evidence and knowledge gaps concerning whether the beneficial effects of metabolic modulators on autoimmunity, hypertension, endothelial dysfunction, and renal injury in lupus result from the restoration of a balanced immune system. The inhibition of glycolysis, mitochondrial metabolism, or mTORC1 has been found to improve endothelial dysfunction and prevent the development of hypertension in mouse models of SLE. Nevertheless, limited information is available regarding the potential vasculo-protective effects of drugs that act on immunometabolism in SLE patients.
Collapse
Affiliation(s)
- Sofía Miñano
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (S.M.); (C.G.-C.)
| | - Cristina González-Correa
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (S.M.); (C.G.-C.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
| | - Javier Moleón
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (S.M.); (C.G.-C.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
| | - Juan Duarte
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (S.M.); (C.G.-C.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| |
Collapse
|
22
|
Muduli S, Gupta MD, Mp G, Yadav R. Anti-inflammatory therapy in atherosclerotic cardiovascular disease: Current reappraisal. Indian Heart J 2023; 75:391-397. [PMID: 37890557 PMCID: PMC10774583 DOI: 10.1016/j.ihj.2023.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023] Open
Affiliation(s)
- Subrat Muduli
- Department of Cardiology, GB Pant Hospital, New Delhi, India
| | - Mohit D Gupta
- Department of Cardiology, GB Pant Hospital, New Delhi, India.
| | - Girish Mp
- Department of Cardiology, GB Pant Hospital, New Delhi, India
| | - Rakesh Yadav
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
23
|
Feugray G, Miranda S, Le Cam Duchez V, Bellien J, Billoir P. Endothelial Progenitor Cells in Autoimmune Disorders. Stem Cell Rev Rep 2023; 19:2597-2611. [PMID: 37676423 DOI: 10.1007/s12015-023-10617-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2023] [Indexed: 09/08/2023]
Abstract
Circulating endothelial progenitor cells (EPCs) were first described in 1997 by Asahara et al. as "putative endothelial cells" from human peripheral blood. The study of endothelial progenitors is also intensifying in several pathologies associated with endothelial damage, including diabetes, myocardial infarction, sepsis, pulmonary arterial hypertension, obstructive bronchopneumopathy and transplantation. EPCs have been studied in several autoimmune diseases with endothelial involvement such as systemic lupus erythematosus, thrombotic thrombocytopenic purpura, antineutrophil cytoplasmic antibodies, vasculitis, rheumatoid arthritis, Goujerot-Sjögren and antiphospholipid syndrome. Factors involved in endothelial damage are due to overexpression of pro-inflammatory cytokines and/or autoantibodies. Management of these pathologies, particularly the long-term use of glucocorticoids and methotrexate, promote atherosclerosis. A lack of standardized assessment of the number and function of EPCs represents a serious challenge for the use of EPCs as prognostic markers of cardiovascular diseases (CVD). The objective of this review was to describe EPCs, their properties and their involvement in several autoimmune diseases.
Collapse
Affiliation(s)
- Guillaume Feugray
- UNIROUEN, INSERM U1096 EnVI, CHU Rouen, Department of General Biochemistry, Normandie University, F-76000, Rouen, France
| | - Sébastien Miranda
- UNIROUEN, INSERM U1096, CHU Rouen. Department of Internal Medicine, Normandie University, Rouen, France
| | | | - Jérémy Bellien
- UNIROUEN, INSERM U1096 EnVI, CHU Rouen, Department of Pharmacology, Normandie University, F-76000, Rouen, France
| | - Paul Billoir
- UNIROUEN, INSERM U1096, CHU Rouen. Department of Internal Medicine, Normandie University, Rouen, France.
- Normandy Univ, U1096, Rouen University Hospital, Vascular Hemostasis Unit, Rouen, France.
| |
Collapse
|
24
|
Yung S, Chan TM. Endothelial cell activation and glycocalyx shedding - potential as biomarkers in patients with lupus nephritis. Front Immunol 2023; 14:1251876. [PMID: 37854589 PMCID: PMC10579905 DOI: 10.3389/fimmu.2023.1251876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 09/18/2023] [Indexed: 10/20/2023] Open
Abstract
Lupus nephritis (LN) is a common and severe manifestation of systemic lupus erythematosus and an important cause of acute and chronic kidney injury. Early diagnosis of LN and preventing relapses are key to preserving renal reserve. However, due to the complexity and heterogeneity of the disease, clinical management remains challenging. Kidney biopsy remains the gold standard for confirming the diagnosis of LN and subsequent assessment of kidney histopathology, but it is invasive and cannot be repeated frequently. Current clinical indicators of kidney function such as proteinuria and serum creatinine level are non-specific and do not accurately reflect histopathological changes, while anti-dsDNA antibody and C3 levels reflect immunological status but not kidney injury. Identification of novel and specific biomarkers for LN is prerequisite to improve management. Renal function deterioration is associated with changes in the endothelial glycocalyx, a delicate gel-like layer located at the interface between the endothelium and bloodstream. Inflammation induces endothelial cell activation and shedding of glycocalyx constituents into the circulation. This review discusses the potential role of soluble glycocalyx components as biomarkers of active LN, especially in patients in whom conventional serological and biochemical markers do not appear helpful.
Collapse
Affiliation(s)
- Susan Yung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Tak Mao Chan
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
25
|
Ferrigno S, Conigliaro P, Rizza S, Longo S, Nesi C, Carlucci F, Bergamini A, Mancino R, Nucci C, Federici M, Chimenti MS, Cesareo M. Relationship between retinal microvascular impairment and subclinical atherosclerosis in SLE. Lupus Sci Med 2023; 10:e000977. [PMID: 37852671 PMCID: PMC10603324 DOI: 10.1136/lupus-2023-000977] [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: 06/17/2023] [Accepted: 09/19/2023] [Indexed: 10/20/2023]
Abstract
OBJECTIVES Patients with SLE have higher cardiovascular (CV) risk compared with healthy controls (HC) and are characterised by accelerated atherosclerosis; intima media thickness (IMT), marker of subclinical atherosclerosis, is higher in patients with SLE than in HCs. Retinal microvascular impairment detected through optical coherence tomography angiography (OCTA) was investigated as a marker of systemic vascular involvement in SLE.The aim of the study was to evaluate the relationship between retinal vascular impairment and IMT in SLE. METHODS Cross-sectional study recruiting patients with SLE and HCs. Data of the study population were collected. CV risk was evaluated through the American College of Cardiology/American Heart Association (ACC/AHA) guidelines, Framingham and QRESEARCH risk estimator V.3 (QRISK3) scores. Both groups underwent OCTA and carotid ultrasound with IMT assessment.Statistical analysis was accomplished using Pearson/Spearman, t-test/Mann-Whitney or χ2 test. Variables statistically significant at univariate regression analysis were tested in an age-corrected and sex-corrected multivariate regression model. RESULTS 43 patients with SLE and 34 HCs were recruited. Patients with SLE showed higher triglycerides (p=0.019), Triglycerides-Glucose (TyG) Index (p=0.035), ACC/AHA guidelines (p=0.001), Framingham Risk Scores (p=0.008) and a reduced superficial (p<0.001) and deep (p=0.005) whole retinal vessel density (VD) compared with HCs.In SLE univariate analysis, deep whole VD showed a negative correlation with IMT (p=0.027), age (p=0.001), systolic blood pressure (p=0.011), QRISK3 Score (p<0.001), Systemic Lupus International Collaborating Clinics Damage Index (p=0.006) and apolipoprotein B (p=0.021), while a positive correlation was found with female sex (p=0.029). Age-adjusted and sex-adjusted multivariate analysis confirmed QRISK3 Score (p=0.049) and IMT (p=0.039) to be independent risk factors for reduced retinal VD. CONCLUSIONS Patients with SLE showed lower retinal VD and higher CV risk indicators compared with HCs. Among patients with SLE, QRISK3 Score and IMT were found to be independent risk factors for retinal vascular impairment, suggesting a role of OCTA in evaluating preclinical CV involvement in SLE. Moreover, TyG Index could represent a biomarker of CV risk in patients with SLE compared with HCs.
Collapse
Affiliation(s)
- Sara Ferrigno
- Rheumatology, Allergology and Clinical Immunology, Department of 'Medicina dei Sistemi', University of Rome Tor Vergata, Rome, Italy
| | - Paola Conigliaro
- Rheumatology, Allergology and Clinical Immunology, Department of 'Medicina dei Sistemi', University of Rome Tor Vergata, Rome, Italy
| | - Stefano Rizza
- Department of 'Medicina dei Sistemi', University of Rome Tor Vergata, Rome, Italy
| | - Susanna Longo
- Department of 'Medicina dei Sistemi', University of Rome Tor Vergata, Rome, Italy
| | - Carolina Nesi
- Ophtalmology, Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| | - Federico Carlucci
- Ophtalmology, Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| | - Alberto Bergamini
- Rheumatology, Allergology and Clinical Immunology, Department of 'Medicina dei Sistemi', University of Rome Tor Vergata, Rome, Italy
| | - Raffaele Mancino
- Ophtalmology, Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| | - Carlo Nucci
- Ophtalmology, Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| | - Massimo Federici
- Department of 'Medicina dei Sistemi', University of Rome Tor Vergata, Rome, Italy
| | - Maria Sole Chimenti
- Rheumatology, Allergology and Clinical Immunology, Department of 'Medicina dei Sistemi', University of Rome Tor Vergata, Rome, Italy
| | - Massimo Cesareo
- Ophtalmology, Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
26
|
Sieczkowska SM, Smaira FI, Mazzolani BC, Romero M, Pasoto SG, de Sá Pinto AL, Lima FR, De Oliveira VR, Ueda S, Benatti FB, Roschel H, Gualano B. A randomized controlled trial of an intervention promoting physical activity and healthy eating recommendations in systemic lupus erythematosus: the protocol study "Living Well with Lupus". Rheumatol Int 2023; 43:1799-1810. [PMID: 37354245 DOI: 10.1007/s00296-023-05370-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 06/08/2023] [Indexed: 06/26/2023]
Abstract
There is a paucity of studies assessing multidisciplinary interventions focused on tackling physical inactivity/sedentary behavior and poor dietary habits in SLE. The Living well with Lupus (LWWL) is a randomized controlled trial to investigate whether a six-month lifestyle change intervention will improve cardiometabolic risk factors (primary outcome) among systemic lupus erythematosus (SLE) patients with low disease activity (SLEDAI score ≤ 4) and with high cardiovascular risk. As secondary goals, we will evaluate: (1) the intervention's safety, efficacy, and feasibility in promoting lifestyle changes, and (2) the effects of the intervention on secondary outcomes (i.e., clinical parameters, functional capacity, fatigue, psychological aspects, sleep quality and health-related quality of life). Patients will be randomly allocated to either a control (i.e., standard care) or a lifestyle intervention group using a simple randomization (1:1 ratio, blocks of 20). Mixed Model analyses will be conducted for comparing groups following an intention-to-treat approach. A per protocol analysis will also be conducted. This study has the potential to generate new, clinically relevant data able to refine the multidisciplinary management of SLE patients. Protocol version number: NCT04431167 (first version).
Collapse
Affiliation(s)
- Sofia Mendes Sieczkowska
- Applied Physiology and Nutrition Research Group, Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de São Paulo, Av. Dr. Arnaldo, 455, 3° Andar, São Paulo, SP, 01246-903, Brazil
| | - Fabiana Infante Smaira
- Applied Physiology and Nutrition Research Group, Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de São Paulo, Av. Dr. Arnaldo, 455, 3° Andar, São Paulo, SP, 01246-903, Brazil
| | - Bruna Caruso Mazzolani
- Applied Physiology and Nutrition Research Group, Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de São Paulo, Av. Dr. Arnaldo, 455, 3° Andar, São Paulo, SP, 01246-903, Brazil
| | - Marina Romero
- Applied Physiology and Nutrition Research Group, Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de São Paulo, Av. Dr. Arnaldo, 455, 3° Andar, São Paulo, SP, 01246-903, Brazil
- School of Applied Sciences, Universidade Estadual de Campinas, Campinas, São Paulo, 13484-350, Brazil
| | - Sandra Gofinet Pasoto
- Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, Av. Dr. Arnaldo, 455, Sao Paulo, SP, 05403-900, Brazil
| | - Ana Lúcia de Sá Pinto
- Applied Physiology and Nutrition Research Group, Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de São Paulo, Av. Dr. Arnaldo, 455, 3° Andar, São Paulo, SP, 01246-903, Brazil
- Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, Av. Dr. Arnaldo, 455, Sao Paulo, SP, 05403-900, Brazil
| | - Fernanda Rodrigues Lima
- Applied Physiology and Nutrition Research Group, Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de São Paulo, Av. Dr. Arnaldo, 455, 3° Andar, São Paulo, SP, 01246-903, Brazil
- Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, Av. Dr. Arnaldo, 455, Sao Paulo, SP, 05403-900, Brazil
| | - Victor Rodrigues De Oliveira
- Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, Av. Dr. Arnaldo, 455, Sao Paulo, SP, 05403-900, Brazil
| | - Serli Ueda
- Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, Av. Dr. Arnaldo, 455, Sao Paulo, SP, 05403-900, Brazil
| | - Fabiana Braga Benatti
- Applied Physiology and Nutrition Research Group, Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de São Paulo, Av. Dr. Arnaldo, 455, 3° Andar, São Paulo, SP, 01246-903, Brazil
- School of Applied Sciences, Universidade Estadual de Campinas, Campinas, São Paulo, 13484-350, Brazil
| | - Hamilton Roschel
- Applied Physiology and Nutrition Research Group, Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de São Paulo, Av. Dr. Arnaldo, 455, 3° Andar, São Paulo, SP, 01246-903, Brazil
| | - Bruno Gualano
- Applied Physiology and Nutrition Research Group, Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de São Paulo, Av. Dr. Arnaldo, 455, 3° Andar, São Paulo, SP, 01246-903, Brazil.
- Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, Av. Dr. Arnaldo, 455, Sao Paulo, SP, 05403-900, Brazil.
| |
Collapse
|
27
|
Urbain F, Ponnaiah M, Ichou F, Lhomme M, Materne C, Galier S, Haroche J, Frisdal E, Mathian A, Durand H, Pha M, Hie M, Kontush A, Cluzel P, Lesnik P, Amoura Z, Guerin M, Cohen Aubart F, Le Goff W. Impaired metabolism predicts coronary artery calcification in women with systemic lupus erythematosus. EBioMedicine 2023; 96:104802. [PMID: 37725854 PMCID: PMC10518349 DOI: 10.1016/j.ebiom.2023.104802] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 08/23/2023] [Accepted: 09/03/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND Patients with systemic lupus erythematosus (SLE) exhibit a high risk for cardiovascular diseases (CVD) which is not fully explained by the classical Framingham risk factors. SLE is characterized by major metabolic alterations which can contribute to the elevated prevalence of CVD. METHODS A comprehensive analysis of the circulating metabolome and lipidome was conducted in a large cohort of 211 women with SLE who underwent a multi-detector computed tomography scan for quantification of coronary artery calcium (CAC), a robust predictor of coronary heart disease (CHD). FINDINGS Beyond traditional risk factors, including age and hypertension, disease activity and duration were independent risk factors for developing CAC in women with SLE. The presence of coronary calcium was associated with major alterations of circulating lipidome dominated by an elevated abundance of ceramides with very long chain fatty acids. Alterations in multiple metabolic pathways, including purine, arginine and proline metabolism, and microbiota-derived metabolites, were also associated with CAC in women with SLE. Logistic regression with bootstrapping of lipidomic and metabolomic variables were used to develop prognostic scores. Strikingly, combining metabolic and lipidomic variables with clinical and biological parameters markedly improved the prediction (area under the curve: 0.887, p < 0.001) of the presence of coronary calcium in women with SLE. INTERPRETATION The present study uncovers the contribution of disturbed metabolism to the presence of coronary artery calcium and the associated risk of CHD in SLE. Identification of novel lipid and metabolite biomarkers may help stratifying patients for reducing CVD morbidity and mortality in SLE. FUNDING INSERM and Sorbonne Université.
Collapse
Affiliation(s)
- Fanny Urbain
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Centre de Référence pour le Lupus, le Syndrome des Anti-phospholipides et Autres Maladies Auto-immunes Rares, Service de Médecine Interne 2, Paris, France
| | - Maharajah Ponnaiah
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), ICAN I/O Data Science (MPo), ICAN Omics (FI and ML), 75013, Paris, France
| | - Farid Ichou
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), ICAN I/O Data Science (MPo), ICAN Omics (FI and ML), 75013, Paris, France
| | - Marie Lhomme
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), ICAN I/O Data Science (MPo), ICAN Omics (FI and ML), 75013, Paris, France
| | - Clément Materne
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), UMR_S1166, F-75013, Paris, France
| | - Sophie Galier
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), UMR_S1166, F-75013, Paris, France
| | - Julien Haroche
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Centre de Référence pour le Lupus, le Syndrome des Anti-phospholipides et Autres Maladies Auto-immunes Rares, Service de Médecine Interne 2, Paris, France
| | - Eric Frisdal
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), UMR_S1166, F-75013, Paris, France
| | - Alexis Mathian
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Centre de Référence pour le Lupus, le Syndrome des Anti-phospholipides et Autres Maladies Auto-immunes Rares, Service de Médecine Interne 2, Paris, France
| | - Herve Durand
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), UMR_S1166, F-75013, Paris, France
| | - Micheline Pha
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Centre de Référence pour le Lupus, le Syndrome des Anti-phospholipides et Autres Maladies Auto-immunes Rares, Service de Médecine Interne 2, Paris, France
| | - Miguel Hie
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Centre de Référence pour le Lupus, le Syndrome des Anti-phospholipides et Autres Maladies Auto-immunes Rares, Service de Médecine Interne 2, Paris, France
| | - Anatol Kontush
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), UMR_S1166, F-75013, Paris, France
| | - Philippe Cluzel
- Cardiovascular and Interventional Radiology Department, Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Paris, F-75013, France
| | - Philippe Lesnik
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), UMR_S1166, F-75013, Paris, France
| | - Zahir Amoura
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Centre de Référence pour le Lupus, le Syndrome des Anti-phospholipides et Autres Maladies Auto-immunes Rares, Service de Médecine Interne 2, Paris, France; Sorbonne Université, Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013, Paris, France
| | - Maryse Guerin
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), UMR_S1166, F-75013, Paris, France
| | - Fleur Cohen Aubart
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Centre de Référence pour le Lupus, le Syndrome des Anti-phospholipides et Autres Maladies Auto-immunes Rares, Service de Médecine Interne 2, Paris, France.
| | - Wilfried Le Goff
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), UMR_S1166, F-75013, Paris, France.
| |
Collapse
|
28
|
Farina N, Webster J, Luo W, Garelick D, Pinto SM, Isenberg D, Rahman A. Factors associated with cardiovascular events in systemic lupus erythematosus in a monocentric cohort with up to 40 years of follow-up. Semin Arthritis Rheum 2023; 61:152226. [PMID: 37201214 DOI: 10.1016/j.semarthrit.2023.152226] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 04/23/2023] [Accepted: 05/08/2023] [Indexed: 05/20/2023]
Abstract
OBJECTIVES Systemic lupus erythematosus (SLE) is associated with an increased cardiovascular risk. Several traditional and disease-specific risk factors have been shown to correlate with the occurrence of cardiovascular events (CVE) in patients with SLE. However, results of previous studies are diverse. The objectives of this study were to report number, type and those factors associated with CVE in patients with SLE in a large, single-center, ethnically diverse cohort with a long follow-up duration. METHODS Medical records of patients treated at the Lupus Clinic at University College London Hospital (UCLH) between 1979 and 2020 were retrospectively reviewed. Data about CVE, traditional cardiovascular risk factors, demographic and disease features, and treatment history were collected. Only patients with complete available information were included in the study. Regression analyses were performed to identify factors associated with CVE. RESULTS Four hundred and nineteen patients were included in the study. Maximum follow-up length was 40 years. Seventy-one (17%) patients had at least one CVE. Multivariable analysis showed that only antiphospholipid antibody positivity (p-value<0.001) was associated with CVE. When analysing different types of CVE, antiphospholipid antibodies were specifically associated with both venous thromboembolic events (p-value<0.001) and cerebrovascular events (p-value=0.007). Dedicated subanalyses revealed that cumulative glucocorticoid dose (p-value=0.010) and a diagnosis of SLE before 2000 (p-value<0.001) were significantly associated with CVE. CONCLUSIONS Cardiovascular disease is highly prevalent among patients with SLE and is associated with antiphospholipid antibodies, glucocorticoid therapy, and diagnosis before 2000.
Collapse
Affiliation(s)
- Nicola Farina
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele, Milan, Italy; Centre for Rheumatology Research, Division of Medicine, University College London, UK
| | - Jemma Webster
- Centre for Rheumatology Research, Division of Medicine, University College London, UK
| | - Weike Luo
- Centre for Rheumatology Research, Division of Medicine, University College London, UK
| | - Daniela Garelick
- Rheumatology Unit, The Zabludowicz Center for Autoimmune Diseases, Tel-Hashomer, Ramat Gan, Israel
| | - Sara Moreira Pinto
- Internal Medicine Service, Centro Hospitalar Póvoa de Varzim e Vila do Conde, Portugal
| | - David Isenberg
- Centre for Rheumatology Research, Division of Medicine, University College London, UK
| | - Anisur Rahman
- Centre for Rheumatology Research, Division of Medicine, University College London, UK.
| |
Collapse
|
29
|
Alhammadi NA, Alqahtani H, Alshahrani LH, Al Qahtani SA, Al Qahtani AA, Alharthi A, Asiri LA, Abu Aqil MA. Cardiovascular Complications in Lupus Patients in the Aseer Region, Saudi Arabia. Cureus 2023; 15:e43501. [PMID: 37719528 PMCID: PMC10500381 DOI: 10.7759/cureus.43501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2023] [Indexed: 09/19/2023] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is an autoimmune disease with high morbidity. The objective of this study was to investigate the prevalence and types of cardiovascular complications among patients with SLE in the Aseer region of Saudi Arabia. METHOD This study is retrospective record-based research conducted at Aseer Central Hospital in the Aseer region from 2020 to 2022. We conducted a comprehensive review of the medical records of patients. RESULTS Out of the 189 patients diagnosed with SLE, 18.0% (34 out of 189) experienced cardiovascular complications. Of the patients who experienced cardiovascular complications, around two-fifths (15/34) fell between 35 and 44 years (44.12%), females represented (31/34) 91.20%, the majority were nonsmokers (32/34) 94.0%, (6/32) 17.65% were diabetic, (17/34) 50.0% had hypertension, and (13/34) 38.24% had vasculitis. Pericarditis and myocarditis are seen in (5/34) 14.7% of each of the cases, endocarditis accounts for (4/34) 11.7% of the cases, a myocardial infarction occurs in (7/34) 20.6% of patients, coronary artery disease is prevalent in (14/34) 38.2% of cases, and valvular lesions at (5/34) 14.7%. CONCLUSIONS Understanding the prevalence of cardiovascular complications in SLE patients is crucial for healthcare providers to tailor treatment plans and preventive measures to address this aspect of the disease.
Collapse
|
30
|
Dar S, Koirala S, Khan A, Bellary MD, Patel AV, Mathew B, Singh R, Baigam N, Razzaq W, Abdin ZU, Khawaja UA. A Comprehensive Literature Review on Managing Systemic Lupus Erythematosus: Addressing Cardiovascular Disease Risk in Females and Its Autoimmune Disease Associations. Cureus 2023; 15:e43725. [PMID: 37727166 PMCID: PMC10505685 DOI: 10.7759/cureus.43725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/18/2023] [Indexed: 09/21/2023] Open
Abstract
This review aimed to evaluate the mechanism of premature cardiovascular disease (CVD) in systemic lupus erythematosus (SLE) patients, particularly in the female population, and emphasize the need for early management interventions; explore the association between SLE and two autoimmune diseases, myasthenia gravis (MG) and antiphospholipid antibody syndrome (APS), and their management strategies; and evaluate the effectiveness of pharmacological and non-pharmacological interventions in managing SLE, focusing on premenopausal females, females of childbearing age, and pregnant patients. We conducted a comprehensive literature review to achieve these objectives using various databases, including PubMed, Google Scholar, and Cochrane. The collected data were analyzed and synthesized to provide an evidence-based overview of SLE, its management strategies as an independent disease, and some disease associations. The treatment should be focused on remission, preventing organ damage, and improving the overall quality of life (QOL). Extensive emphasis should also be focused on diagnosing SLE and concurrent underlying secondary diseases timely and managing them appropriately.
Collapse
Affiliation(s)
- Saleha Dar
- Department of Adult Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Sabina Koirala
- Department of Medicine, Gandaki Medical College, Pokhara, NPL
| | - Arooba Khan
- Department of Internal Medicine, Khyber Medical College, Peshawar, PAK
| | | | - Arya V Patel
- Department of Internal Medicine, Smt. Nathiba Hargovandas Lakhmichand (NHL) Municipal Medical College, Ahmedabad, IND
| | - Bejoi Mathew
- Department of Internal Medicine, Sri Devaraj Urs Medical College, Kolar, IND
| | - Rahul Singh
- Department of Medicine, Armed Forces Medical College, Pune, IND
| | - Nahida Baigam
- Department of Medicine, Association of Physicians of Pakistani Descent of North America (APPNA), Westmont, USA
| | - Waleed Razzaq
- Department of Internal Medicine, Services Hospital Lahore, Lahore, PAK
| | - Zain U Abdin
- Department of Medicine, District Head Quarter Hospital, Faisalabad, PAK
| | - Uzzam Ahmed Khawaja
- Department of Pulmonary and Critical Care Medicine, Jinnah Medical and Dental College, Karachi, PAK
- Department of Clinical and Translational Research, Dr. Ferrer BioPharma, South Miami, USA
| |
Collapse
|
31
|
Shahi T, Ghimire P, Khanal UP, Dhakal TR, Jha S. Fatal ascending aortic aneurysm in a patient with systemic lupus erythematosus: A case report. Clin Case Rep 2023; 11:e7696. [PMID: 37457996 PMCID: PMC10340078 DOI: 10.1002/ccr3.7696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/27/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023] Open
Abstract
Aortic aneurysm is a potentially life-threatening condition with higher incidence in patients with systemic lupus erythematosus(SLE). Patients usually present with nonspecific symptoms and diagnosis is typically made incidentally through imaging studies. Management strategies include medical therapy to control inflammation and hypertension, surgical intervention for large or symptomatic aneursyms, and close monitoring for early detection of complications. We present a case of a 49-year female with multiple joint pain and other nonspecific symptoms for 7 years. Anti-ds DNA and ANA titre were significantly high and CT angiogram showed ascending aortic aneurysm measuring 5.5 cm. Conservative management was started with steroids, hydroxychloroquine, and antihypertensives, while awaiting surgery. However she suddenly collapsed, probably due to aneurysm rupture and could not be revived. Our case report therefore emphasizes the importance of close surveillance and timely intervention to minimize the morbidity and mortality in these patients.
Collapse
Affiliation(s)
- Tejash Shahi
- Institute of Medicine, Tribhuwan University Teaching HospitalKathmanduNepal
| | - Prinska Ghimire
- Institute of Medicine, Tribhuwan University Teaching HospitalKathmanduNepal
| | | | - Tulsi Ram Dhakal
- Institute of Medicine, Tribhuwan University Teaching HospitalKathmanduNepal
| | - Saket Jha
- Institute of Medicine, Tribhuwan University Teaching HospitalKathmanduNepal
| |
Collapse
|
32
|
Frostegård J. Antibodies against Phosphorylcholine-Implications for Chronic Inflammatory Diseases. Metabolites 2023; 13:720. [PMID: 37367878 DOI: 10.3390/metabo13060720] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 06/28/2023] Open
Abstract
Atherosclerosis and its main consequence, cardiovascular disease (CVD) are nowadays regarded as chronic inflammatory disease conditions, and CVD is the main cause of death in the world. Other examples of chronic inflammation are rheumatic and other autoimmune conditions, but also diabetes, obesity, and even osteoarthritis among others. In addition, infectious diseases can have traits in common with these conditions. Systemic lupus erythematosus (SLE) is a prototypical autoimmune disease, where atherosclerosis is increased and the risk of CVD is very high. This is a clinical problem but could also shed light on the role of the immune system in atherosclerosis and CVD. Underlying mechanisms are of major interest and these are only partially known. Phosphorylcholine (PC) is a small lipid-related antigen, which is both a danger associated molecular pattern (DAMP), and a pathogen associated molecular pattern (PAMP). Antibodies against PC are ubiquitous and 5-10% of circulating IgM is IgM anti-PC. Anti-PC, especially IgM and IgG1 anti-PC, has been associated with protection in the chronic inflammatory conditions mentioned above, and develops during the first years of life, while being present at very low levels at birth. Animal experiments with immunization to raise anti-PC ameliorate atherosclerosis and other chronic inflammatory conditions. Potential mechanisms include anti-inflammatory, immune modulatory, clearance of dead cells and protection against infectious agents. An intriguing possibility is to raise anti-PC levels through immunization, to prevent and/or ameliorate chronic inflammation.
Collapse
Affiliation(s)
- Johan Frostegård
- IMM, Nobels Väg 13, Karolinska Institutet, 17165 Stockholm, Sweden
| |
Collapse
|
33
|
Cornwell MG, Bannoudi HE, Luttrell-Williams E, Engel A, Barrett TJ, Myndzar K, Izmirly P, Belmont HM, Clancy R, Ruggles KV, Buyon JP, Berger JS. Modeling of clinical phenotypes in systemic lupus erythematosus based on the platelet transcriptome and FCGR2a genotype. J Transl Med 2023; 21:247. [PMID: 37029410 PMCID: PMC10082503 DOI: 10.1186/s12967-023-04059-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 03/12/2023] [Indexed: 04/09/2023] Open
Abstract
BACKGROUND The clinical heterogeneity of SLE with its complex pathogenesis remains challenging as we strive to provide optimal management. The contribution of platelets to endovascular homeostasis, inflammation and immune regulation highlights their potential importance in SLE. Prior work from our group showed that the Fcγ receptor type IIa (FcγRIIa)-R/H131 biallelic polymorphism is associated with increased platelet activity and cardiovascular risk in SLE. The study was initiated to investigate the platelet transcriptome in patients with SLE and evaluate its association across FcγRIIa genotypes and distinct clinical features. METHODS Fifty-one patients fulfilling established criteria for SLE (mean age = 41.1 ± 12.3, 100% female, 45% Hispanic, 24% black, 22% Asian, 51% white, mean SLEDAI = 4.4 ± 4.2 at baseline) were enrolled and compared with 18 demographically matched control samples. The FCGR2a receptor was genotyped for each sample, and RNA-seq was performed on isolated, leukocyte-depleted platelets. Transcriptomic data were used to create a modular landscape to explore the differences between SLE patients and controls and various clinical parameters in the context of FCGR2a genotypes. RESULTS There were 2290 differentially expressed genes enriched for pathways involved in interferon signaling, immune activation, and coagulation when comparing SLE samples vs controls. When analyzing patients with proteinuria, modules associated with oxidative phosphorylation and platelet activity were unexpectedly decreased. Furthermore, genes that were increased in SLE and in patients with proteinuria were enriched for immune effector processes, while genes increased in SLE but decreased in proteinuria were enriched for coagulation and cell adhesion. A low-binding FCG2Ra allele (R131) was associated with decreases in FCR activation, which further correlated with increases in platelet and immune activation pathways. Finally, we were able to create a transcriptomic signature of clinically active disease that performed significantly well in discerning SLE patients with active clinical disease form those with inactive clinical disease. CONCLUSIONS In aggregate, these data demonstrate the platelet transcriptome provides insight into lupus pathogenesis and disease activity, and shows potential use as means of assessing this complex disease using a liquid biopsy.
Collapse
Affiliation(s)
- MacIntosh G Cornwell
- Division of Precision Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY, USA
| | - Hanane El Bannoudi
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Elliot Luttrell-Williams
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Alexis Engel
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, NYU Grossman School of Medicine, Medical Science Building 593, 530 First Avenue, New York, NY, 10016, USA
| | - Tessa J Barrett
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Center for the Prevention of Cardiovascular Disease, New York University Grossman School of Medicine, 530 First Avenue, Skirball 9R, New York, NY, 10016, USA
| | - Khrystyna Myndzar
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, NYU Grossman School of Medicine, Medical Science Building 593, 530 First Avenue, New York, NY, 10016, USA
| | - Peter Izmirly
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, NYU Grossman School of Medicine, Medical Science Building 593, 530 First Avenue, New York, NY, 10016, USA
| | - H Michael Belmont
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, NYU Grossman School of Medicine, Medical Science Building 593, 530 First Avenue, New York, NY, 10016, USA
| | - Robert Clancy
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, NYU Grossman School of Medicine, Medical Science Building 593, 530 First Avenue, New York, NY, 10016, USA
| | - Kelly V Ruggles
- Division of Precision Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA.
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY, USA.
| | - Jill P Buyon
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, NYU Grossman School of Medicine, Medical Science Building 593, 530 First Avenue, New York, NY, 10016, USA.
| | - Jeffrey S Berger
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA.
- Center for the Prevention of Cardiovascular Disease, New York University Grossman School of Medicine, 530 First Avenue, Skirball 9R, New York, NY, 10016, USA.
| |
Collapse
|
34
|
Shi H, Goo B, Kim D, Kress TC, Ogbi M, Mintz J, Wu H, Belin de Chantemèle EJ, Stepp D, Long X, Guha A, Lee R, Carbone L, Annex BH, Hui DY, Kim HW, Weintraub NL. Perivascular adipose tissue promotes vascular dysfunction in murine lupus. Front Immunol 2023; 14:1095034. [PMID: 37006244 PMCID: PMC10062185 DOI: 10.3389/fimmu.2023.1095034] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
Introduction Patients with systemic lupus erythematosus (SLE) are at elevated risk for Q10 cardiovascular disease (CVD) due to accelerated atherosclerosis. Compared to heathy control subjects, lupus patients have higher volumes and densities of thoracic aortic perivascular adipose tissue (PVAT), which independently associates with vascular calcification, a marker of subclinical atherosclerosis. However, the biological and functional role of PVAT in SLE has not been directly investigated. Methods Using mouse models of lupus, we studied the phenotype and function of PVAT, and the mechanisms linking PVAT and vascular dysfunction in lupus disease. Results and discussion Lupus mice were hypermetabolic and exhibited partial lipodystrophy, with sparing of thoracic aortic PVAT. Using wire myography, we found that mice with active lupus exhibited impaired endothelium-dependent relaxation of thoracic aorta, which was further exacerbated in the presence of thoracic aortic PVAT. Interestingly, PVAT from lupus mice exhibited phenotypic switching, as evidenced by "whitening" and hypertrophy of perivascular adipocytes along with immune cell infiltration, in association with adventitial hyperplasia. In addition, expression of UCP1, a brown/beige adipose marker, was dramatically decreased, while CD45-positive leukocyte infiltration was increased, in PVAT from lupus mice. Furthermore, PVAT from lupus mice exhibited a marked decrease in adipogenic gene expression, concomitant with increased pro-inflammatory adipocytokine and leukocyte marker expression. Taken together, these results suggest that dysfunctional, inflamed PVAT may contribute to vascular disease in lupus.
Collapse
Affiliation(s)
- Hong Shi
- Division of Rheumatology, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Brandee Goo
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - David Kim
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Taylor C. Kress
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Mourad Ogbi
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - James Mintz
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Hanping Wu
- Department of Radiology and Imaging, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Eric J. Belin de Chantemèle
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Division of Cardiology, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - David Stepp
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Xiaochun Long
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Division of Cardiology, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Avirup Guha
- Division of Cardiology, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Richard Lee
- Department of Surgery, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Laura Carbone
- Division of Rheumatology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Brian H. Annex
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Division of Cardiology, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - David Y. Hui
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Ha Won Kim
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Division of Cardiology, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Neal L. Weintraub
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Division of Cardiology, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
35
|
Mehta PK, Levit RD, Wood MJ, Aggarwal N, O'Donoghue ML, Lim SS, Lindley K, Gaignard S, Quesada O, Vatsa N, Leon A, Volgman AS, Malas W, Pepine CJ. Chronic rheumatologic disorders and cardiovascular disease risk in women. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2023; 27:100267. [PMID: 38511090 PMCID: PMC10945906 DOI: 10.1016/j.ahjo.2023.100267] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/18/2023] [Accepted: 02/02/2023] [Indexed: 03/22/2024]
Abstract
Cardiovascular disease (CVD) is a major health threat to women worldwide. In addition to traditional CVD risk factors, autoimmune conditions are increasingly being recognized as contributors to adverse CVD consequences in women. Chronic systemic autoimmune and inflammatory disorders can trigger premature and accelerated atherosclerosis, microvascular dysfunction, and thrombosis. The presence of comorbid conditions, duration of the autoimmune condition, disease severity, and treatment of underlying inflammation are all factors that impact CVD risk and progression. Early identification and screening of CVD risk factors in those with underlying autoimmune conditions may attenuate CVD in this population. Treatment with non-steroidal anti-inflammatory drugs, corticosteroids, disease modifying agents and biologics may influence CVD risk factors and overall risk. Multi-disciplinary and team-based care, clinical trials, and collaborative team-science studies focusing on systemic autoimmune conditions will be beneficial to advance care for women.
Collapse
Affiliation(s)
- Puja K. Mehta
- Emory Women's Heart Center, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Rebecca D. Levit
- Emory Women's Heart Center, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Malissa J. Wood
- Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA
| | - Niti Aggarwal
- Department of Cardiovascular Disease, Mayo Clinic, Rochester, MN, USA
| | - Michelle L. O'Donoghue
- TIMI Study Group, Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA
| | - S. Sam Lim
- Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Kate Lindley
- Cardiovascular Division, Washington University in St. Louis, USA
| | - Scott Gaignard
- J. Willis Hurst Internal Medicine Residency Program, Emory University, Atlanta, GA, USA
| | - Odayme Quesada
- Women's Heart Center, The Christ Hospital Heart and Vascular Institute, Cincinnati, OH, USA
| | - Nishant Vatsa
- J. Willis Hurst Internal Medicine Residency Program, Emory University, Atlanta, GA, USA
| | - Ana Leon
- Emory University School of Medicine, Atlanta, GA, USA
| | | | - Waddah Malas
- Loyola University Internal Medicine Residency Program, Chicago, IL, USA
| | - Carl J. Pepine
- Division of Cardiovascular Medicine, University of Florida, Gainesville, FL, USA
| | - American College of Cardiology Cardiovascular Disease in Women Committee
- Emory Women's Heart Center, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
- Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA
- Department of Cardiovascular Disease, Mayo Clinic, Rochester, MN, USA
- TIMI Study Group, Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA
- Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, USA
- Cardiovascular Division, Washington University in St. Louis, USA
- J. Willis Hurst Internal Medicine Residency Program, Emory University, Atlanta, GA, USA
- Women's Heart Center, The Christ Hospital Heart and Vascular Institute, Cincinnati, OH, USA
- Emory University School of Medicine, Atlanta, GA, USA
- Section Division of Cardiology, Rush University Medical Center, Chicago, IL, USA
- Loyola University Internal Medicine Residency Program, Chicago, IL, USA
- Division of Cardiovascular Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
36
|
Hurst C, Soto M, Vina ER, Rodgers KE. Renin-Angiotensin System-Modifying Antihypertensive Drugs Can Reduce the Risk of Cardiovascular Complications in Lupus: A Retrospective Cohort Study. Am J Med 2023; 136:284-293.e4. [PMID: 36495935 PMCID: PMC9957968 DOI: 10.1016/j.amjmed.2022.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Patients with systemic lupus erythematosus have a higher incidence of cardiovascular disease than the general population. Antihypertensive drugs that modify the renin-angiotensin system (RAS) are used to protect renal function in lupus nephritis and may also have extrarenal effects that lower cardiovascular disease risk due to their anti-inflammatory properties. In this study, we compared the effects of RAS vs non-RAS antihypertensive drugs on cardiovascular disease incidence in patients with lupus. METHODS Using a medical insurance claims dataset, 220,168 patients with lupus were identified, of which 31,647 patients (4018 patients prescribed RAS drugs, 27,629 patients prescribed non-RAS drugs) were eligible for the study. Patients had a mean age of 46.1 years, were 93.0% female, and healthy (96.9% Charlson Comorbidity Index score 0-4). Patients in the 2 drug groups were propensity score matched using demographic data, risk factors, and comorbidities. RESULTS Use of RAS vs non-RAS drugs lowered the relative risk (RR) of diagnosis of cardiovascular disease (RR 0.80; 95% confidence interval [CI], 0.74-0.87), which was more pronounced after propensity score matching (RR 0.62; 95% CI, 0.57-0.68). The decreased risk in cardiovascular disease occurred regardless of lupus nephritis status (with lupus nephritis: RR 0.51; 95% CI, 0.39-0.65; without lupus nephritis: RR 0.65; 95% CI, 0.59-0.72). RAS-modifying therapies significantly increased cardiovascular disease-free survival probability over a 5-year period (86.0% vs 78.3% probability). CONCLUSIONS RAS-modifying drugs reduced the risk of cardiovascular disease in patients with systemic lupus erythematosus in this dataset. These findings have the potential to impact clinical decision-making with regards to hypertension management in patients with lupus.
Collapse
Affiliation(s)
- Chelsie Hurst
- Department of Pharmacology, Center for Innovation in Brain Science, College of Medicine, University of Arizona, Tucson
| | - Maira Soto
- Department of Pharmacology, Center for Innovation in Brain Science, College of Medicine, University of Arizona, Tucson
| | - Ernest R Vina
- Section of Rheumatology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pa
| | - Kathleen E Rodgers
- Department of Pharmacology, Center for Innovation in Brain Science, College of Medicine, University of Arizona, Tucson.
| |
Collapse
|
37
|
Løgstrup BB. Heart Failure in Rheumatic Disease: Secular Trends and Novel Insights. Rheum Dis Clin North Am 2023; 49:67-79. [PMID: 36424027 DOI: 10.1016/j.rdc.2022.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
There is a significant increase in risk of heart failure in several rheumatic diseases. Common cardiovascular risk factors and inflammatory processes, present in both rheumatic diseases and heart failure, are contributing to this increase. The opportunities for using immune-based strategies to fight development of heart failure in rheumatic diseases are evolving. The diversity of inflammation calls for a tailored characterization of inflammation, enabling differentiation of inflammation and subsequent introduction of precision medicine using target-specific strategies and immunomodulatory therapy. As the field of rheuma-cardiology is still evolving, clear recommendations cannot be given yet.
Collapse
Affiliation(s)
- Brian Bridal Løgstrup
- Department of Cardiology, Institute of Clinical Medicine, Aarhus University Hospital, Palle Juul Jensens Boulevard 99, Aarhus N 8200, Denmark.
| |
Collapse
|
38
|
Misra DP, Hauge EM, Crowson CS, Kitas GD, Ormseth SR, Karpouzas GA. Atherosclerotic Cardiovascular Risk Stratification in the Rheumatic Diseases:: An Integrative, Multiparametric Approach. Rheum Dis Clin North Am 2023; 49:19-43. [PMID: 36424025 DOI: 10.1016/j.rdc.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cardiovascular disease (CVD) risk is increased in most inflammatory rheumatic diseases (IRDs), reiterating the role of inflammation in the initiation and progression of atherosclerosis. An inverse association of CVD risk with body weight and lipid levels has been described in IRDs. Coronary artery calcium scores, plaque burden and characteristics, and carotid plaques on ultrasound optimize CVD risk estimate in IRDs. Biomarkers of cardiac injury, autoantibodies, lipid biomarkers, and cytokines also improve risk assessment in IRDs. Machine learning and deep learning algorithms for phenotype and image analysis hold promise to improve CVD risk stratification in IRDs.
Collapse
Affiliation(s)
- Durga Prasanna Misra
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Rae Bareli Road, Lucknow 226014, India
| | - Ellen M Hauge
- Division of Rheumatology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99 DK-8200, Aarhus, Denmark
| | - Cynthia S Crowson
- Department of Quantitative Health Sciences and Division of Rheumatology, Mayo Clinic, 200 first St SW, Rochester, MN 55905, USA
| | | | - Sarah R Ormseth
- The Lundquist Institute and Harbor-UCLA Medical Center, 1124 West Carson Street, Building E4-R17, Torrance, CA 90502, USA
| | - George A Karpouzas
- The Lundquist Institute and Harbor-UCLA Medical Center, 1124 West Carson Street, Building E4-R17, Torrance, CA 90502, USA.
| |
Collapse
|
39
|
Sabio JM, Garcia-de Los Ríos C, Medina-Casado M, Del Mar Del Águila-García M, Cáliz-Cáliz R, Díaz-Chamorro A. High-sensitivity cardiac troponin I is a biomarker for increased arterial stiffness in systemic lupus erythematous women with normal kidney function. Rheumatol Int 2023; 43:253-263. [PMID: 36094601 DOI: 10.1007/s00296-022-05204-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/29/2022] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease (CVD) is a main cause of death in patients with systemic lupus erythematous (SLE). Algorithms for cardiovascular risk stratification in general population underestimate the risk for CVD in SLE. Our study aimed to determine whether serum high-sensitivity cardiac troponin I (hs-cTnI) might help to identify SLE patients with subclinical atherosclerosis. Arterial stiffness was assessed measuring the carotid-femoral pulse wave velocity (PWV) in 68 SLE women with a normal or almost normal kidney function and in 71 controls of similar characteristics. None of the participants had a history of an overt CVD. Serum hs-cTnI level was measured using the chemiluminescence method. Factors associated with an increased PWV (iPWV) were identified and multivariate analysis was performed. When detectable, patients tended to have had higher hs-cTnI levels than controls [2.9 (2.3-4.0) vs 2.4 (2.2-4.1); p = 0.098] and were more likely to have detectable hs-cTnI [50% vs 28%, odds ratio (OR) 7.0; 95% confidence interval (CI) 0.008-0.013]. Also, patients with iPWV were more likely to have detectable hs-cTnI than those with normal PWV (OR 6.4; 95% CI 0.019-0.026). In the multivariate analysis, the age at SLE diagnosis (OR 1.24; 95% CI 1.04-1.48), systolic blood pressure (OR 1.28; 95% CI 1.10-1.48) and detectable hs-cTnI level (OR 2.04; 95% CI 1.18-3.50) were independently associated with an iPWV. The negative predictive value of having an iPWV with undetectable hs-cTnI levels was 88%. Hs-cTnI may be a useful biomarker for the identification of SLE patients with iPWV as a surrogated marker of subclinical atherosclerosis. Specifically targeted prospective studies are needed to confirm this hypothesis.
Collapse
Affiliation(s)
- J M Sabio
- Systemic Autoimmune Diseases Unit, Department of Internal Medicine, Virgen de Las Nieves University Hospital, Granada, Spain
| | - Carlos Garcia-de Los Ríos
- Department of Internal Medicine, Virgen de Las Nieves University Hospital, 9th Floor, Avda. Fuerzas Armadas No. 2, 18012, Granada, Spain.
| | | | | | - Rafael Cáliz-Cáliz
- Department of Rheumatology, Virgen de Las Nieves University Hospital, Granada, Spain
| | - Antonio Díaz-Chamorro
- Department of Internal Medicine, Virgen de Las Nieves University Hospital, 9th Floor, Avda. Fuerzas Armadas No. 2, 18012, Granada, Spain
| |
Collapse
|
40
|
Santos-Moreno P, Arias-Aponte J, Rodríguez-Vargas GS, Nieto-Zambrano PD, Villarreal L, Ibatá L, Martinez S, Rubio-Rubio JA, Rodríguez P, Rojas-Villarraga A. Polyautoimmunity in systemic lupus erythematosus patients: New insights from a cross-sectional study. J Transl Autoimmun 2023; 6:100187. [PMID: 36654852 PMCID: PMC9841268 DOI: 10.1016/j.jtauto.2022.100187] [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: 11/30/2022] [Accepted: 12/29/2022] [Indexed: 01/05/2023] Open
Abstract
Objective To assess the frequency of polyautoimmunity (PolyA) in a cohort of Colombian patients with systemic lupus erythematosus (SLE) and to identify associated factors. Methods This is an analytical cross-sectional study in a specialized center., a comprehensive review of the medical records of SLE patients was performed from 2015 to 2020 in order to obtain demographic, clinical data, laboratory, and treatment information. Associations between PolyA, demographic, and characteristics of the disease were explored. Results A total of 463 patients were included in the analysis. The average age was 47.3 ± 15 years. Most of this population were female (87.4%), whom were diagnosed with SLE in a long-term SLE (10.6 ± 10.1 years). Out of the total patients, 34.7% were diagnosed with PolyA. Among the most frequent clinical criteria for SLICC, arthritis (65%), kidney impairment (39.5%), and alopecia (34.8%) were found. The most frequent SLE-associated PolyA were antiphospholipid syndrome (APS) and Sjögren's syndrome (SS) (16.63% and 10.58%, respectively). PolyA-associated factors were age, xerophthalmia, central nervous system occlusion, and deep vein thrombosis (DVT). In contrast, renal impairment was significantly less frequent in PolyA patients after multivariate analysis. Conclusion The results have showed associated factors with PolyA like age, xerophthalmia, central nervous system occlusion, and deep vein thrombosis in this cohort. On the other hand, lupus nephritis was less frequent in patients with PolyA. This study provides a spotlight of a specific SLE population as real-life evidence for a better characterization of PolyA in the future.
Collapse
Affiliation(s)
- Pedro Santos-Moreno
- Scientific Direction, Biomab Center for Rheumatoid Arthritis, Bogotá, D.C., 110221, Colombia,Corresponding author.
| | - Julián Arias-Aponte
- Research Vice-rectory, Fundación Universitaria de Ciencias de La Salud-FUCS, Bogotá D.C, 111411, Colombia
| | | | | | - Laura Villarreal
- Medical Direction, Biomab Center for Rheumatoid Arthritis, Bogotá, D.C., 110221, Colombia
| | - Linda Ibatá
- Epidemiology, Biomab IPS, Bogotá, D.C, 110221, Colombia
| | | | - Jaime-Andrés Rubio-Rubio
- Research Institute, Fundación Universitaria de Ciencias de La Salud FUCS, Bogotá, D.C, 111411, Colombia
| | - Pedro Rodríguez
- Scientific Direction, Biomab Center for Rheumatoid Arthritis, Bogotá, D.C., 110221, Colombia
| | - Adriana Rojas-Villarraga
- Research Institute, Fundación Universitaria de Ciencias de La Salud FUCS, Bogotá, D.C, 111411, Colombia
| |
Collapse
|
41
|
Moore J, Lakshmanan S, Manubolu VS, Kinninger A, Stojan G, Goldman DW, Petri M, Budoff M, Karpouzas GA. Coronary plaque progression is greater in systemic lupus erythematosus than rheumatoid arthritis. Coron Artery Dis 2023; 34:52-58. [PMID: 36421035 DOI: 10.1097/mca.0000000000001205] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) are associated with a high incidence of cardiovascular disease. Coronary atherosclerosis, particularly total plaque and noncalcified plaque on coronary computed tomography angiography (CCTA) has been correlated with cardiovascular events. We compared baseline coronary plaque burden and progression by serial CCTA in SLE and RA patients. METHODS We prospectively evaluated 44 patients who underwent serial CCTA examinations to quantify coronary plaque progression, 22 SLE patients, and 22 age- and sex-matched RA patients. Semiautomated plaque software was used for quantitative plaque assessment. Linear regression examined the effect of SLE diagnosis (versus RA) on annualized change in natural log-transformed total normalized atheroma volume (ln-TAV norm ) for low-attenuation, fibrofatty, fibrous, total noncalcified, densely calcified, and total plaque. RESULTS No quantitative differences for any plaque types were observed at baseline between SLE and RA patients ( P = 0.330-0.990). After adjustment for baseline plaque and cardiovascular risk factors, the increase in ln-TAV norm was higher in SLE than RA patients for fibrous [Exp-β: 0.202 (0.398), P = 0.0003], total noncalcified [Exp-β: 0.179 (0.393), P = 0.0001], and total plaque volume [Exp-β: 0.154 (0.501), P = 0.0007], but not for low-attenuation, fibrofatty, or densely calcified plaque ( P = 0.103-0.489). Patients with SLE had 80% more fibrous, 82% more noncalcified, and 85% more total plaque increase than those with RA. CONCLUSION Coronary plaque volume was similar in RA and SLE at baseline. Progression was greater in SLE, which may explain the greater cardiovascular risk in this disease. Further research to evaluate screening and management strategies for cardiovascular disease in these high-risk patients is warranted.
Collapse
Affiliation(s)
- Jeff Moore
- Chronic Disease Clinical Research Center, The Lundquist Institute for Biomedical Innovation at the Harbor University of California Los Angeles Medical Center, Torrance
- School of Exercise and Nutritional Sciences, San Diego State University, San Diego, California
| | - Suvasini Lakshmanan
- Chronic Disease Clinical Research Center, The Lundquist Institute for Biomedical Innovation at the Harbor University of California Los Angeles Medical Center, Torrance
| | - Venkat Sanjay Manubolu
- Chronic Disease Clinical Research Center, The Lundquist Institute for Biomedical Innovation at the Harbor University of California Los Angeles Medical Center, Torrance
| | - April Kinninger
- Chronic Disease Clinical Research Center, The Lundquist Institute for Biomedical Innovation at the Harbor University of California Los Angeles Medical Center, Torrance
| | - George Stojan
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniel W Goldman
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Matthew Budoff
- Chronic Disease Clinical Research Center, The Lundquist Institute for Biomedical Innovation at the Harbor University of California Los Angeles Medical Center, Torrance
| | - George A Karpouzas
- Chronic Disease Clinical Research Center, The Lundquist Institute for Biomedical Innovation at the Harbor University of California Los Angeles Medical Center, Torrance
| |
Collapse
|
42
|
Taylor T, Anastasiou C, Ja C, Rush S, Trupin L, Dall'Era M, Katz P, Barbour KE, Greenlund KJ, Yazdany J, Gianfrancesco MA. Causes of Death Among Individuals With Systemic Lupus Erythematosus by Race and Ethnicity: A Population-Based Study. Arthritis Care Res (Hoboken) 2023; 75:61-68. [PMID: 35904969 PMCID: PMC9797422 DOI: 10.1002/acr.24988] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 07/10/2022] [Accepted: 07/26/2022] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Non-White populations are at higher risk of developing systemic lupus erythematosus (SLE) and have more severe outcomes, including mortality. The present study was undertaken to examine how specific causes of death vary by race and ethnicity, including Asian and Hispanic individuals. METHODS The California Lupus Surveillance Project included SLE cases identified among residents of San Francisco County, CA during January 1, 2007 to December 31, 2009. Cases were matched to the National Death Index over a 10-year period. Logistic regression examined age-adjusted differences in causes of death by race, ethnicity, and sex. Age-standardized mortality ratios between individuals with SLE and the corresponding general population were calculated for the leading cause of death, and observed versus expected deaths were estimated. RESULTS The study included 812 individuals of White (38%), Asian (36%), Black (20%), and mixed/other/unknown (5%) race; 15% identified as Hispanic. One hundred thirty-five deaths were recorded, with a mean ± SD age at death of 62.2 ± 15.6 years. Cardiovascular disease (CVD) was the leading cause of death overall (33%), and across all racial and ethnic groups, followed by rheumatic disease (18%) and hematologic/oncologic conditions (18%). CVD as the underlying cause of death was 3.63 times higher among SLE cases than in the general population. CVD deaths for those with SLE were nearly 4 and 6 times higher for Asian and Hispanic individuals with SLE, respectively, compared to the general population. CONCLUSION Individuals with SLE experience a disproportionate burden of CVD mortality compared to the general population, which is magnified for Asian and Hispanic groups.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kamil E Barbour
- Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | | | | |
Collapse
|
43
|
Abstract
The prognosis in systemic lupus erythematosus (SLE) has improved due to better treatment and care, but cardiovascular disease (CVD) still remains an important clinical problem, since the risk of CVD in SLE is much higher than among controls. Atherosclerosis is the main cause of CVD in the general population, and in SLE, increased atherosclerosis, especially the prevalence of atherosclerotic plaques, has been demonstrated. Atherosclerosis is an inflammatory condition, where immunity plays an important role. Interestingly, oxidized low-density lipoprotein, defective clearance of dead cells, and inflammation, with a pro-inflammatory T-cell profile are characteristics of both atherosclerosis and SLE. In addition to atherosclerosis as an underlying cause of CVD in SLE, there are also other non-mutually exclusive mechanisms, and the most important of these are antiphospholipid antibodies (aPL) leading to the antiphospholipid antibody syndrome with both arterial and venous thrombosis. aPL can cause direct pro-inflammatory and prothrombotic effects on endothelial and other cells and also interfere with the coagulation, for example, by inhibiting annexin A5 from its antithrombotic and protective effects. Antibodies against phosphorylcholine (anti-PC) and other small lipid-related epitopes, sometimes called natural antibodies, are negatively associated with CVD and atherosclerosis in SLE. Taken together, a combination of traditional risk factors such as hypertension and dyslipidemia, and nontraditional ones, especially aPL, inflammation, and low anti-PC are implicated in the increased risk of CVD in SLE. Close monitoring of both traditional risk factors and nontraditional ones, including treatment of disease manifestations, not lest renal disease in SLE, is warranted.
Collapse
Affiliation(s)
- Johan Frostegård
- Section of Immunology and Chronic Disease, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
44
|
Wang SXY, Horomanski A, Tooley JE, Reejhsinghani R, White AA. A change of heart. J Hosp Med 2022; 18:444-448. [PMID: 36479928 DOI: 10.1002/jhm.13016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 10/31/2022] [Accepted: 11/04/2022] [Indexed: 12/13/2022]
Affiliation(s)
- Samantha X Y Wang
- Department of Medicine, Division of Hospital Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Audra Horomanski
- Department of Medicine, Division of Immunology & Rheumatology, Stanford University School of Medicine, Stanford, California, USA
| | - James E Tooley
- Department of Medicine, Division of Cardiology, UCSF School of Medicine, San Francisco, California, USA
| | - Risheen Reejhsinghani
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Andrew A White
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
45
|
Abstract
Sarcoidosis is a heterogeneous disease, which can affect virtually every body organ, even though lungs and intra thoracic lymph nodes are almost universally affected. The presence of noncaseating granulomas is the histopathological hallmark of the disease, and clinical picture depends on the organs affected. Data about interaction between sarcoidosis and comorbidities, such as cardiovascular and pulmonary diseases, autoimmune disorders, malignancy and drug-related adverse events are limited. Several lung conditions can be associated with sarcoidosis, such as pulmonary hypertension and fibrosis, making it difficult sometimes the differentiation between complications and distinctive pathologies. Their coexistence may complicate the diagnosis of sarcoidosis and contribute to the highly variable and unpredictable natural history, particularly if several diseases are recognised. A thorough assessment of specific disorders that can be associated with sarcoidosis should always be carried out, and future studies will need to evaluate sarcoidosis not only as a single disorder, but also in the light of possible concomitant conditions.Key messagesComorbidities in sarcoidosis are common, especially cardiovascular and pulmonary diseases.In the diagnostic workup, a distinction must be made between sarcoidosis-related complaints and complaints caused by other separate disorders. It can be very difficult to distinguish between complications of sarcoidosis and other concomitant conditions.The coexistence of multiple conditions may complicate the diagnosis of sarcoidosis, affect its natural course and response to treatment.
Collapse
Affiliation(s)
- Claudio Tana
- Geriatrics Clinic, Medicine Department, SS Annunziata Hospital of Chieti, Chieti, Italy
| | - Marjolein Drent
- Department of Pharmacology and Toxicology, Faculty of Health, Medicine and Life Science, Maastricht University, Maastricht, The Netherlands.,ILD Center of Excellence, Department of Respiratory Medicine, St. Antonius Hospital, Nieuwegein, The Netherlands.,ILD Care Foundation Research Team, Ede, The Netherlands
| | - Hilario Nunes
- AP-HP, Hôpital Avicenne, Service de Pneumologie, Centre de Référence des Maladies Pulmonaires Rares de l'adulte, Université Sorbonne Paris Nord, Bobigny, France
| | - Vasilis Kouranos
- Interstitial Lung Disease Unit, Royal Brompton Hospital, National Heart and Lung Institute, Imperial College London, London, UK
| | - Francesco Cinetto
- Rare Diseases Referral Center, Internal Medicine 1, Ca' Foncello Hospital - AULSS2 Marca Trevigiana and Department of Medicine - DIMED, University of Padova, Italy
| | - Naomi T Jessurun
- ILD Care Foundation Research Team, Ede, The Netherlands.,Netherlands Pharmacovigilance Centre Lareb, 's-Hertogenbosch, The Netherlands
| | - Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| |
Collapse
|
46
|
Yang Y, Chen Y, Li Y, Feng Y, Hu N, Xue L. Expression and Significance of Programmed Death-1 and Its Ligands in the Accelerated Formation of Atherosclerosis in an Induced Murine Lupus Model. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6255383. [PMID: 39050559 PMCID: PMC11268968 DOI: 10.1155/2022/6255383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 09/30/2022] [Accepted: 10/19/2022] [Indexed: 07/27/2024]
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease that occurs in artery walls, which seriously affects the survival and prognosis of patients with systemic lupus erythematosus (SLE). Immune and inflammatory responses have notable effects on all stages of AS. In this study, we modeled SLE combined with AS in vivo via intraperitoneal injection of pristane (2,6,10,14-tetramethylpentadecane) into apolipoprotein E-knockout (ApoE-/- ) mice that had accelerated atherosclerotic lesions compared with wild-type (WT) ApoE-/- mice. In pristane-induced ApoE-/- mice, expression of programmed death-1 (PD-1) and programmed death-ligand 1 (PD-L1) in peripheral blood and on the surfaces of atherosclerotic lesions significantly increased, and levels of proinflammatory cytokines, namely, interferon-gamma (IFN-γ) and tumor necrosis factor alpha (TNF-α) in peripheral blood were elevated. We did not detect expression of programmed death-ligand 2 (PD-L2) in the arterial plaques of either pristane-induced or WT ApoE-/- mice, nor did we observe any significant difference in PD-L2 expression in peripheral blood between the two groups. Taken together, these results suggested that PD-1/PD-L1 signaling pathway might play an important regulatory role in the progression of AS in an induced murine lupus model which implies a potential target for treatment of AS in SLE.
Collapse
Affiliation(s)
- Yue Yang
- Department of Rheumatology, Yueyang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yueying Chen
- Department of Rheumatology, Yueyang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yongming Li
- Department of Rheumatology, Yueyang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yiyi Feng
- Department of Rheumatology, Yueyang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Na Hu
- Science and Technology Experimental Center, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Luan Xue
- Department of Rheumatology, Yueyang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| |
Collapse
|
47
|
Guzmán-Martínez G, Marañón C. Immune mechanisms associated with cardiovascular disease in systemic lupus erythematosus: A path to potential biomarkers. Front Immunol 2022; 13:974826. [PMID: 36420265 PMCID: PMC9677819 DOI: 10.3389/fimmu.2022.974826] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/13/2022] [Indexed: 10/29/2023] Open
Abstract
Systemic lupus erythematosus (SLE) patients display an increased risk of cardiovascular disease (CVD). With the improved clinical management of other classical severe manifestation of the disease, CVD is becoming one of the most relevant complications of SLE, and it is an important factor causing morbidity and mortality. Several immune constituents have been shown to be involved in the pathogenesis of atherosclerosis and endothelial damage in SLE patients, including specific circulating cell populations, autoantibodies, and inflammatory mediators. In this review, we summarize the presentation of CVD in SLE and the role of the autoimmune responses present in SLE patients in the induction of atherogenesis, endothelial impairment and cardiac disease. Additionally, we discuss the utility of these immune mediators as early CVD biomarkers and targets for clinical intervention in SLE patients.
Collapse
Affiliation(s)
- Gabriela Guzmán-Martínez
- Atrys Health, Madrid, Spain
- Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
- Department of Cardiology, La Paz University Hospital, IdiPaz, Madrid, Spain
| | - Concepción Marañón
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| |
Collapse
|
48
|
Ramipril Improves Endothelial Function and Increases the Number of Endothelial Progenitor Cells in Patients With Systemic Lupus Erythematosus. J Clin Rheumatol 2022; 28:349-353. [PMID: 35662232 DOI: 10.1097/rhu.0000000000001869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND/OBJECTIVES Endothelial dysfunction and reduced number of endothelial progenitor cells (EPCs) in peripheral blood are contributing factors to cardiovascular disease in systemic lupus erythematosus (SLE) patients. Endothelial progenitor cell proliferation is regulated by vascular endothelial growth factor (VEGF). Angiotensin-converting enzyme inhibitors reduce cardiovascular mortality in patients with coronary heart disease. METHODS This was a randomized trial including 37 female SLE patients without cardiovascular risk factors allocated into 2 groups: 19 patients received ramipril 10 mg/d for 12 weeks (IG) and 18 patients maintained without ramipril (CG). Endothelial function was assessed by brachial artery ultrasound measuring flow-mediated dilation, and EPCs were quantified by flow cytometry and cell culture, at baseline and after 12 weeks. Serum VEGF levels were measured by enzyme-linked immunosorbent assay. Statistical analysis was intention to treat. p < 0.05 was considered significant. RESULTS After 12 weeks, higher flow-mediated dilation (6.17% vs. 11.14%, p < 0.001) was observed in IG, without change in CG (5.37% vs. 5.02%, p = 0.630). Higher number of EPC colony-forming units was also observed in IG (21.3 ± 10.4 vs. 31.6 ± 8.5, p < 0.001), without difference in CG ( p = 0.714). No difference was found in EPCs evaluated by flow cytometry. Vascular endothelial growth factor level increased after 12 weeks in IG ( p = 0.048), with no difference in CG ( p = 0.661). CONCLUSION Ramipril improved endothelial function and increased the numbers of EPCs evaluated by cell culture and VEGF levels in SLE patients without cardiovascular risk factors. These data suggest that angiotensin-converting enzyme inhibitor bring an extra benefit beyond the hypotensive action and should be considered as a preferred antihypertensive drug in SLE patients.
Collapse
|
49
|
Su L, Qi Z, Guan S, Wei L, Zhao Y. Exploring the risk factors for ischemic cerebrovascular disease in systemic lupus erythematosus: A single-center case-control study. Front Immunol 2022; 13:978910. [PMID: 36238309 PMCID: PMC9552613 DOI: 10.3389/fimmu.2022.978910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectivesIschemic cerebrovascular disease (ICVD) is one of the most common and severe complications in systemic lupus erythematosus (SLE). We aim to explore the risk factors for ICVD in SLE and to assess their associated clinical characteristics.MethodsIn this study, 44 lupus patients with ICVD (ICVD-SLE) and 80 age- and sex-matched lupus patients without ICVD (non-ICVD-SLE) who were hospitalized in our center between 2014 and 2021 were enrolled. A comprehensive set of clinical and socio-demographic data was recorded. In the ICVD-SLE group, the modified Rankin score (mRS) at 90 days after the occurrence of ICVD, the brain MRI, and arterial ultrasonography findings were collected. Group comparisons were made with continuous variables using an independent t-test or the Mann–Whitney test, and with categorical variables using the chi-square test or Fisher exact test. Multivariate logistic regression analysis was performed to identify the risk factors for ICVD in SLE. Patients with ICVD-SLE were divided into three subgroups according to the gradations of intracranial arterial stenosis (ICAS). The subgroup comparisons were performed by one-way ANOVA test or Kruskal–Wallis test.ResultsOf the 44 patients with ICVD, 45% had a large-vessel ischemic stroke, 50% had a symptomatic lacunar stroke, and 9% had a transient ischemic attack. 2 (4.5%) had both large-vessel ischemic stroke and symptomatic lacunar stroke. Multivariate logistic regression analysis showed that cutaneous vasculitis (OR=7.36, 95% CI=2.11–25.65), anticardiolipin antibody (aCL) (OR=4.38, 95% CI=1.435–13.350), and lupus anticoagulant (LA) (OR=7.543,95% CI=1.789–31.808) were the risk factors, and hydroxychloroquine (HCQ) therapy (OR=0.198, 95% CI=0.078–0.502) was the protective factor, after controlling for confounders. During the analysis of the subgroups, no significant difference was observed between the patients in the group without internal carotid arterial occlusion (ICAS) and those with severe ICAS except for diagnostic delay. However, patients in the moderate ICAS group were older when SLE occurred (P<0.01), had a longer diagnostic delay (P<0.01), a lower percentage of hypocomplementemia (P=0.05) and steroids and HCQ therapy (P=0.01, P=0.05, respectively), a trend toward lower mRS score, but a higher incidence of carotid atherosclerotic plaque (P<0.01), when compared with the other two subgroups.ConclusionCutaneous vasculitis and antiphospholipid antibodies (aPLs) are associated with an increased risk of ICVD, while HCQ therapy may provide protection against ICVD in SLE. The ICVD in younger lupus patients is associated with complement-mediated inflammation and poorer outcome, and require immunosuppressive therapy, whereas the ICVD in elderly patients are characterized by moderate ICAS and carotid atherosclerotic plaques.
Collapse
Affiliation(s)
- Li Su
- Department of Rheumatology and Allergy, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhigang Qi
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Shaochen Guan
- Evidence-Based Medical Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Lian Wei
- Department of Rheumatology and Allergy, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yi Zhao
- Department of Rheumatology and Allergy, Xuanwu Hospital, Capital Medical University, Beijing, China
- *Correspondence: Yi Zhao,
| |
Collapse
|
50
|
Conrad N, Verbeke G, Molenberghs G, Goetschalckx L, Callender T, Cambridge G, Mason JC, Rahimi K, McMurray JJV, Verbakel JY. Autoimmune diseases and cardiovascular risk: a population-based study on 19 autoimmune diseases and 12 cardiovascular diseases in 22 million individuals in the UK. Lancet 2022; 400:733-743. [PMID: 36041475 DOI: 10.1016/s0140-6736(22)01349-6] [Citation(s) in RCA: 141] [Impact Index Per Article: 70.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/01/2022] [Accepted: 07/11/2022] [Indexed: 02/09/2023]
Abstract
BACKGROUND Some autoimmune diseases are associated with an increased risk of cardiovascular disease. We aimed to determine whether or not this is true, and to what extent, for a broad range of autoimmune conditions. METHODS In this population-based study, we used linked primary and secondary care records from the Clinical Practice Research Datalink (CPRD), GOLD and Aurum datasets, to assemble a cohort of individuals across the UK who were newly diagnosed with any of 19 autoimmune diseases between Jan 1, 2000, and Dec 31, 2017, younger than 80 years at diagnosis, and free of cardiovascular diseases up to 12 months after diagnosis. We also assembled a matched cohort with up to five individuals matched on age, sex, socioeconomic status, region, and calendar year, who were free of autoimmune disease and free of cardiovascular diseases up to 12 months after study entry. Both cohorts were followed up until June 30, 2019. We investigated the incidence of 12 cardiovascular outcomes and used Cox proportional hazards models to examine differences in patients with and without autoimmune diseases. FINDINGS Of 22 009 375 individuals identified from the CPRD databases, we identified 446 449 eligible individuals with autoimmune diseases and 2 102 830 matched controls. In the autoimmune cohort, mean age at diagnosis was 46·2 years (SD 19·8), and 271 410 (60·8%) were women and 175 039 (39·2%) were men. 68 413 (15·3%) people with and 231 410 (11·0%) without autoimmune diseases developed incident cardiovascular disease during a median of 6·2 years (IQR 2·7-10·8) of follow-up. The incidence rate of cardiovascular disease was 23·3 events per 1000 patient-years among patients with autoimmune disease and 15·0 events per 1000 patient-years among those without an autoimmune disease (hazard ratio [HR] 1·56 [95% CI 1·52-1·59]). An increased risk of cardiovascular disease with autoimmune disease was seen for every individual cardiovascular disease and increased progressively with the number of autoimmune diseases present (one disease: HR 1·41 [95% CI 1·37-1·45]; two diseases: 2·63 [2·49-2·78]); three or more diseases: 3·79 [3·36-4·27]), and in younger age groups (age <45 years: 2·33 [2·16-2·51]; 55-64 years: 1·76 [1·67-1·85]; ≥75 years: 1·30 [1·24-1·36]). Among autoimmune diseases, systemic sclerosis (3·59 [2·81-4·59]), Addison's disease (2·83 [1·96-4·09]), systemic lupus erythematosus (2·82 [2·38-3·33]), and type 1 diabetes (2·36 [2·21-2·52]) had the highest overall cardiovascular risk. INTERPRETATION These findings warrant targeted cardiovascular prevention measures, in particular in younger patients with autoimmune diseases, and further research into pathophysiological mechanisms underlying these complications. FUNDING Horizon 2020 Marie Skłodowska-Curie Actions and European Society of Cardiology.
Collapse
Affiliation(s)
- Nathalie Conrad
- EPI-Centre, Academic Centre for General Practice, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium.
| | - Geert Verbeke
- Interuniversity Institute for Biostatistics and statistical Bioinformatics (I-BioStat), Hasselt University and KU Leuven, Leuven, Belgium
| | - Geert Molenberghs
- Interuniversity Institute for Biostatistics and statistical Bioinformatics (I-BioStat), Hasselt University and KU Leuven, Leuven, Belgium
| | - Laura Goetschalckx
- EPI-Centre, Academic Centre for General Practice, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Thomas Callender
- Department of Respiratory Medicine, University College London, London, UK
| | - Geraldine Cambridge
- Division of Medicine, Department of Rheumatology, University College London, London, UK
| | - Justin C Mason
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London UK
| | - Kazem Rahimi
- Deep Medicine, Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | - John J V McMurray
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Jan Y Verbakel
- EPI-Centre, Academic Centre for General Practice, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium; Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| |
Collapse
|