1
|
Porsch F, Binder CJ. Autoimmune diseases and atherosclerotic cardiovascular disease. Nat Rev Cardiol 2024; 21:780-807. [PMID: 38937626 DOI: 10.1038/s41569-024-01045-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/28/2024] [Indexed: 06/29/2024]
Abstract
Autoimmune diseases are associated with a dramatically increased risk of atherosclerotic cardiovascular disease and its clinical manifestations. The increased risk is consistent with the notion that atherogenesis is modulated by both protective and disease-promoting immune mechanisms. Notably, traditional cardiovascular risk factors such as dyslipidaemia and hypertension alone do not explain the increased risk of cardiovascular disease associated with autoimmune diseases. Several mechanisms have been implicated in mediating the autoimmunity-associated cardiovascular risk, either directly or by modulating the effect of other risk factors in a complex interplay. Aberrant leukocyte function and pro-inflammatory cytokines are central to both disease entities, resulting in vascular dysfunction, impaired resolution of inflammation and promotion of chronic inflammation. Similarly, loss of tolerance to self-antigens and the generation of autoantibodies are key features of autoimmunity but are also implicated in the maladaptive inflammatory response during atherosclerotic cardiovascular disease. Therefore, immunomodulatory therapies are potential efficacious interventions to directly reduce the risk of cardiovascular disease, and biomarkers of autoimmune disease activity could be relevant tools to stratify patients with autoimmunity according to their cardiovascular risk. In this Review, we discuss the pathophysiological aspects of the increased cardiovascular risk associated with autoimmunity and highlight the many open questions that need to be answered to develop novel therapies that specifically address this unmet clinical need.
Collapse
Affiliation(s)
- Florentina Porsch
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
2
|
Mangoni AA, Wiese MD, Woodman RJ, Sotgia S, Zinellu A, Carru C, Hulin JA, Shanahan EM, Tommasi S. Methotrexate, blood pressure and arterial function in rheumatoid arthritis: study protocol. Future Cardiol 2024:1-13. [PMID: 39387403 DOI: 10.1080/14796678.2024.2411167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 09/27/2024] [Indexed: 10/15/2024] Open
Abstract
This article discusses the rationale and design of the study "Methotrexate, blood pressure, and arterial function in rheumatoid arthritis". The recognition that immune activation and excess inflammation favor atherosclerosis has stimulated a significant body of research not only to identify new drugs targeting these pathways but also to repurpose (reposition) existing immunomodulatory medications as atheroprotective agents. Observational studies in patients with rheumatoid arthritis have reported that treatment with methotrexate, a traditional disease-modifying antirheumatic drug, is associated with a significantly lower risk of cardiovascular morbidity and mortality when compared with other disease-modifying antirheumatic drugs. One potential mechanism accounting for the reduced cardiovascular risk associated with methotrexate is the lowering effect on arterial blood pressure. However, such effect has only been observed in cross-sectional and observational studies. Given the established role of hypertension as a leading cardiovascular risk factor, these observations justify an intervention comparison study, the focus of this article, investigating the temporal effects of methotrexate on blood pressure and various surrogate markers of atherosclerosis in patients with rheumatoid arthritis. The results of this study might lead to the repurposing of methotrexate for cardiovascular prevention in patients with and without autoimmune disorders.Clinical Trial Registration: NCT03254589 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Arduino A Mangoni
- College of Medicine & Public Health, Flinders Health & Medical Research Institute, Flinders University, Adelaide, Australia
- Department of Clinical Pharmacology, Finders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia
| | - Michael D Wiese
- Centre for Pharmaceutical Innovation, Clinical & Health Sciences, University of South Australia, Adelaide, Australia
| | - Richard J Woodman
- Centre for Epidemiology & Biostatistics, College of Medicine & Public Health, Flinders University, Adelaide, Australia
| | - Salvatore Sotgia
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Medical Oncology Unit, University Hospital (AOUSS), Sassari, Italy
| | - Julie-Ann Hulin
- College of Medicine & Public Health, Flinders Health & Medical Research Institute, Flinders University, Adelaide, Australia
| | - E Michael Shanahan
- College of Medicine & Public Health, Flinders Health & Medical Research Institute, Flinders University, Adelaide, Australia
- Department of Rheumatology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia
| | - Sara Tommasi
- College of Medicine & Public Health, Flinders Health & Medical Research Institute, Flinders University, Adelaide, Australia
- Department of Clinical Pharmacology, Finders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia
| |
Collapse
|
3
|
Rossdale J, Graby J, Harris M, Jones C, Greenish D, Bartlett J, Gilroy A, Sanghera J, Pauling JD, Skeoch S, Flower V, Mackenzie Ross R, Suntharalingam J, Rodrigues JCL. Coronary artery calcification is prevalent in systemic sclerosis and is associated with adverse prognosis. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2024; 9:192-202. [PMID: 39386266 PMCID: PMC11459481 DOI: 10.1177/23971983241264090] [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: 04/12/2024] [Accepted: 06/02/2024] [Indexed: 10/12/2024]
Abstract
Objective Coronary artery calcification assessed on thoracic computed tomography represents the calcific component of established coronary artery disease, is a biomarker of total atheromatous plaque burden and predicts mortality. Systemic sclerosis is a pro-inflammatory condition, and inflammation is also a driver of coronary artery disease. We assessed coronary artery calcification prevalence, mortality risk and potential clinical impact on primary prevention in a cohort of patients with systemic sclerosis, differentiated by clinical phenotype including the presence of interstitial lung disease and pulmonary arterial hypertension. Methods Retrospective analysis of 258 computed tomographies in systemic sclerosis patients from three prospectively maintained clinical and research databases at a single tertiary rheumatology/pulmonary hypertension (PH) service between March 2007 and September 2020 (mean age = 65 ± 12, 14% male). Co-morbidities, statin prescription and all-cause mortality were recorded. Patients were subtyped according to underlying systemic sclerosis complications. Computed tomographies were re-reviewed for coronary artery calcification; severity was graded using a 4-point scale per vessel and summed for total coronary artery calcification score. The impact of reporting coronary artery calcification was assessed against pre-existing statin prescriptions. Results Coronary artery calcification was present in 58% (149/258). Coronary artery calcification was more prevalent in systemic sclerosis-pulmonary arterial hypertension than in systemic sclerosis subgroups with interstitial lung disease or without pulmonary arterial hypertension, controlling for age, sex, co-morbidities and smoking status (71%; χ 2(13) = 81.4; p < 0.001). The presence and severity of coronary artery calcification were associated with increased risk of mortality independently of age and co-morbidities (hazard ratio = 2.8; 95% confidence interval = 1.2-6.6; p = 0.018). The 'number needed to report' coronary artery calcification presence to potentially impact management was 3. Conclusions Coronary artery calcification is common in systemic sclerosis. Coronary artery calcification predicts mortality independently of age and confounding co-morbidities which suggests this finding has clinical relevance and is a potential target for screening and therapeutic intervention.
Collapse
Affiliation(s)
- Jennifer Rossdale
- Respiratory Department, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
- Department of Life Sciences, University of Bath, Bath, UK
| | - John Graby
- Cardiology Department, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
- Department for Health, University of Bath, Bath, UK
| | - Maredudd Harris
- Radiology Department, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
| | - Calum Jones
- Radiology Department, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
| | - Davyd Greenish
- Radiology Department, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
| | - Jessica Bartlett
- Radiology Department, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
| | - Andrew Gilroy
- Radiology Department, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
| | - Jamie Sanghera
- Respiratory Department, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
| | - John D Pauling
- Royal National Hospital for Rheumatic Diseases, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
- Rheumatology Department, North Bristol NHS Trust, Bristol, UK
| | - Sarah Skeoch
- Royal National Hospital for Rheumatic Diseases, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
| | - Victoria Flower
- Department of Life Sciences, University of Bath, Bath, UK
- Royal National Hospital for Rheumatic Diseases, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
| | - Rob Mackenzie Ross
- Respiratory Department, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
| | - Jay Suntharalingam
- Respiratory Department, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
- Department of Life Sciences, University of Bath, Bath, UK
| | - Jonathan CL Rodrigues
- Department for Health, University of Bath, Bath, UK
- Radiology Department, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
| |
Collapse
|
4
|
Vrints C, Andreotti F, Koskinas KC, Rossello X, Adamo M, Ainslie J, Banning AP, Budaj A, Buechel RR, Chiariello GA, Chieffo A, Christodorescu RM, Deaton C, Doenst T, Jones HW, Kunadian V, Mehilli J, Milojevic M, Piek JJ, Pugliese F, Rubboli A, Semb AG, Senior R, Ten Berg JM, Van Belle E, Van Craenenbroeck EM, Vidal-Perez R, Winther S. 2024 ESC Guidelines for the management of chronic coronary syndromes. Eur Heart J 2024; 45:3415-3537. [PMID: 39210710 DOI: 10.1093/eurheartj/ehae177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
|
5
|
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
|
6
|
Chourabi S, Sayhi S, Ben Ameur S, Chourabi C, Mahfoudhi H, Fehri W, Ben Abdelhafidh N. Cardiac involvement in systemic lupus erythematosus: Interest of 2D global longitudinal strain. Lupus 2024; 33:1100-1108. [PMID: 39041544 DOI: 10.1177/09612033241266990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a chronic multisystem autoimmune disease of undetermined etiology. Cardiac involvement is common in SLE and constitutes one of the main causes of mortality. More recently, new ultrasound imaging techniques, such as transthoracic ultrasound (TTE) with strain evaluation, have appeared and seem promising for the detection of cardiac involvement. The objective of our work was to study the frequency and characteristics of ultrasound abnormalities found in lupus patients and to study the benefit of ultrasound with global longitudinal strain (GLS) for early management. METHODS It was an observational study of patients followed for SLE at the internal medicine and cardiology department of the HMPIT for 6 months (May-November 2023). The definition of cardiac involvement was by ultrasound. All patients benefited from TTE coupled with 2D-strain. We divided the workforce into two groups: the first group (patients with heart disease) and the second group (patients without heart disease). RESULTS In a series of 40 lupus patients including 33 women and seven men, cardiac manifestations were reported in 60% of patients. In the first group, 29% had palpitations, 25% had chest pain, 67% had dyspnea, 37% had pericarditis, 8% had pulmonary arterial hypertension (PAH) and 12% had myocarditis. The comparative study showed that patients in the first group presented significantly more frequently with dyspnea (p = 0.02), chest pain (p = 0.03) and serositis (p = 0.01) compared to those in the second group. The mean left ventricular ejection fraction (LVEF) did not show a significant difference between the two groups. On the other hand, the average Global Longitudinal Strain (GLS) was significantly altered in the first group (p = 0.01). Furthermore, the frequency of pathological GLS was significantly higher in patients with lupus heart disease (p < 0.01). CONCLUSION Cardiac involvement during SLE is a frequent and most often asymptomatic complication. A systematic search for this impairment using a high-performance echocardiography examination, namely the 2D GLS, is essential for early treatment.
Collapse
Affiliation(s)
- Sana Chourabi
- Department of Internal Medicine, HMPIT, Tunis, Tunisia
| | - Sameh Sayhi
- Department of Internal Medicine, HMPIT, Tunis, Tunisia
| | | | | | | | - Wafa Fehri
- Department of Internal Medicine, HMPIT, Tunis, Tunisia
| | | |
Collapse
|
7
|
Sysojev AÖ, Alfredsson L, Klareskog L, Silberberg GN, Saevarsdottir S, Padyukov L, Magnusson PKE, Askling J, Westerlind H. Minor Genetic Overlap Among Rheumatoid Arthritis, Myocardial Infarction, and Myocardial Infarction Risk Determinants. Arthritis Rheumatol 2024; 76:1344-1352. [PMID: 38782598 DOI: 10.1002/art.42918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/22/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVE The aim of this study was to investigate whether a shared genetic susceptibility exists between individuals with rheumatoid arthritis (RA) and individuals with myocardial infarction (MI)-including major MI risk factors-and to quantify the degree of any such overlap. METHODS Genome-wide association study (GWAS) data for individuals with RA were constructed from a sample of 26,637 Swedish patients with RA and controls without RA. For patients with MI, GWAS data were obtained from a previously published meta-analysis. Genome-wide genetic correlation was estimated via linkage disequilibrium score regression. LAVA was employed to estimate local genetic correlations in ~2,500 nonoverlapping loci, including the major histocompatibility complex. The controls without RA were used for reference panel data. We also assessed stratified estimates of both genome-wide and local genetic correlation based on subsamples of individuals with seropositive RA and those with seronegative RA. Furthermore, genome-wide genetic correlation was estimated between RA and selected cardiovascular risk factors to elucidate pleiotropic relationships. RESULTS Following quality control, our GWAS of patients with RA consisted of 25,826 individuas. Genome-wide genetic correlation between patients with RA and MI was estimated to 0.13 (95% confidence interval -0.03 to 0.29). Six regions exhibited significant local genetic correlation, though none harbored any known risk single-nucleotide polymorphisms for either of the two traits. Estimates were similar in both individuals with seropositive RA and those with seronegative RA. No statistically significant genetic correlations were observed between RA risk factors and any of the MI risk factors. CONCLUSION Our findings indicate that genetic overlap between patients with RA and MI is minor. Furthermore, genetic overlap between RA and MI risk factors seem unlikely to provide a major contribution to the increased risk of MI observed in patients with RA.
Collapse
Affiliation(s)
| | | | - Lars Klareskog
- Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Gilad N Silberberg
- Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Saedis Saevarsdottir
- Karolinska Institute, Stockholm, Sweden, and deCODE genetics, Reykjavik, Iceland. Members of the Swedish Rheumatology Quality Register Biobank Group are shown in Appendix A
| | - Leonid Padyukov
- Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | | | - Johan Askling
- Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | | |
Collapse
|
8
|
Carvalho LM, Carvalho BG, Souza LL, da Mota JC, Ribeiro AA, Nicoletti CF. Obesity as an aggravating factor of systemic lupus erythematosus disease: What we already know and what we must explore? - A rapid scoping review. Nutrition 2024; 128:112559. [PMID: 39244807 DOI: 10.1016/j.nut.2024.112559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/10/2024]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease that can affect various organs and systems. Symptoms of SLE can vary widely from person to person and over time, including fatigue, joint pain, skin rashes, fever, and inflammation of multiple organs. The association between SLE and excess body weight has been the subject of study, with evidence suggesting that overweight and obesity can worsen the disease´s clinical presentation. Obesity is linked to a state of low-grade chronic inflammation, which can exacerbate the inflammation present in SLE. Additionally, obesity may negatively impact treatment response, disease progression, and patient prognosis. Patients with SLE and obesity may face additional challenges in managing the disease, such as increased symptom severity, higher risk of cardiovascular and renal complications, and a reduced response to conventional treatments. Obesity can also influence the quality of life of patients with SLE, making a holistic approach that considers the individual's nutritional status essential. Therefore, understanding the relationship between obesity and SLE is crucial for optimizing treatment, improving clinical outcomes, and enhancing patients' quality of life. Further research is needed to elucidate the underlying pathophysiological mechanisms, develop more precise and personalized management strategies, and identify biomarkers that can predict disease prognosis and treatment response.
Collapse
Affiliation(s)
- Lucas M Carvalho
- Applied Physiology and Nutrition Research Group - School of Physical Education and Sport and Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil; Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Beatriz G Carvalho
- Applied Physiology and Nutrition Research Group - School of Physical Education and Sport and Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil; Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Leticia L Souza
- Applied Physiology and Nutrition Research Group - School of Physical Education and Sport and Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil; Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Jhulia Cnl da Mota
- Applied Physiology and Nutrition Research Group - School of Physical Education and Sport and Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil; Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Amanda A Ribeiro
- Applied Physiology and Nutrition Research Group - School of Physical Education and Sport and Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil; Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Carolina F Nicoletti
- Applied Physiology and Nutrition Research Group - School of Physical Education and Sport and Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil; Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil; Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo, Brazil.
| |
Collapse
|
9
|
Olivera PA, Dignass A, Dubinsky MC, Peretto G, Kotze PG, Dotan I, Kobayashi T, Ghosh S, Magro F, Faria-Neto JR, Siegmund B, Danese S, Peyrin-Biroulet L. Preventing and managing cardiovascular events in patients with inflammatory bowel diseases treated with small-molecule drugs, an international Delphi consensus. Dig Liver Dis 2024; 56:1270-1280. [PMID: 38584033 DOI: 10.1016/j.dld.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 03/08/2024] [Accepted: 03/17/2024] [Indexed: 04/09/2024]
Abstract
Janus kinase (JAK) inhibitors and sphingosine 1 phosphate (S1P) receptor modulators are small molecule drugs (SMDs) approved for IBD treatment. Their use in clinical practice might be limited due to cardiovascular concerns. We aimed to provide guidance on risk assessment, monitoring, and management strategies, aiming to minimize potential cardiovascular risks of SMDs and to facilitate an adequate shared decision-making. A systematic literature search was conducted, and proposed statements were prepared. A virtual consensus meeting was held, in which eleven IBD physicians and two cardiovascular specialists from ten countries attended. Proposed statements were voted upon in an anonymous manner. Agreement was defined as at least 75 % of participants voting as 'agree' with each statement. Consensus was reached for eighteen statements. Available evidence does not show a higher risk of cardiovascular events with JAK inhibitors in the overall IBD population, although it might be increased in patients with an unfavorable cardiovascular profile. S1P receptor modulators may be associated with a risk of bradycardia, atrioventricular blocks, and hypertension. Cardiovascular risk stratification should be done before initiation of SMDs. Although the risk of cardiovascular events in patients with IBD on SMDs appears to be low overall, caution should still be taken in certain scenarios.
Collapse
Affiliation(s)
- Pablo A Olivera
- IBD Unit, Gastroenterology Section, Department of Internal Medicine, Centro de Educación Médica e Investigación Clínica (CEMIC), Buenos Aires, Argentina; Zane Cohen Centre for Digestive Diseases, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Axel Dignass
- Department of Medicine I, Agaplesion Markus Hospital, Goethe-University, Frankfurt Am Main, Germany
| | - Marla C Dubinsky
- The Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Giovanni Peretto
- Myocarditis Disease Unit, Department of Cardiac Electrophysiology and Arrhythmology, IRCCS San Raffaele Scientific Institute, Milan, Italy; School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Paulo G Kotze
- IBD outpatient clinics, Colorectal Surgery Unit, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Brazil
| | - Iris Dotan
- Sackler Faculty of Medicine, Tel Aviv University, Israel; Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel
| | - Taku Kobayashi
- Center for Advanced IBD Research and Treatment, Kitasato University Kitasato Institute Hospital, Tokyo, Japan
| | - Subrata Ghosh
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Fernando Magro
- CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
| | - Jose Rocha Faria-Neto
- School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Brazil
| | - Britta Siegmund
- Division of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Silvio Danese
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele and University Vita-Salute San Raffaele, Milano, Italy
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France; INSERM, NGERE, University of Lorraine, F-54000 Nancy, France; INFINY Institute, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France; FHU-CURE, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France; Groupe Hospitalier Privé Ambroise Paré - Hartmann, Paris IBD center, 92200 Neuilly sur Seine, France; Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, Quebec, Canada.
| |
Collapse
|
10
|
Rydell E, Jacobsson LTH, Turesson C. Circulating Interleukin 17A and Other Inflammatory Proteins May Predict Cardiovascular Disease in Early Rheumatoid Arthritis. J Rheumatol 2024; 51:752-758. [PMID: 38692670 DOI: 10.3899/jrheum.2023-1078] [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: 04/12/2024] [Indexed: 05/03/2024]
Abstract
OBJECTIVE The objective of this study was to investigate the impact of 92 inflammatory proteins on the risk of cardiovascular disease (CVD) in patients with early rheumatoid arthritis (RA). METHODS This study included consecutive patients with early RA recruited between 1995 and 2002. Stored plasma samples were analyzed for 92 inflammatory proteins. CVD diagnoses were retrieved from national in-patient and cause-of-death registries. Statistical analyses were predesignated as hypothesis-driven or exploratory. For the latter, proteins were selected based on principal component analysis (ie, factor loading > 0.5 within main components). Potential predictors of CVD and coronary artery disease (CAD) were assessed using Cox regression. RESULTS Data on baseline levels of proteins and CVD were available for 163 patients. As hypothesized, levels of interleukin 17A (IL-17A) were associated with CVD (hazard ratio 1.35, 95% CI 1.02-1.78, adjusted for age, sex, hypertension, diabetes, smoking, and erythrocyte sedimentation rate [ESR]), although not significantly with CAD. Osteoprotegerin (OPG) levels were significantly associated with both outcomes, but only in crude models. No associations were observed for IL-6, tumor necrosis factor, monocyte chemotactic protein-1, or IL-8. In the exploratory analyses, MCP-3 in particular had significant associations with both outcomes in crude models. CONCLUSION Circulating IL-17A at RA diagnosis predicted future CVD, although we cannot exclude the possibility that this finding is due to multiple testing. The association was independent of traditional CVD risk factors, and of ESR at the time of diagnosis. Further, OPG may be a predictor of CVD. We also identified some novel potential biomarkers for CVD in RA.
Collapse
Affiliation(s)
- Emil Rydell
- E. Rydell, MD, Rheumatology, Department of Clinical Sciences, Malmö, Lund University, Malmö;
| | - Lennart T H Jacobsson
- L.T.H. Jacobsson, MD, PhD, Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at Gothenburg University, Göteborg
| | - Carl Turesson
- C. Turesson, MD, PhD, Rheumatology, Department of Clinical Sciences, Lund University, and Department of Rheumatology, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
11
|
Karpouzas GA, Ormseth SR, van Riel PLCM, Gonzalez-Gay MA, Corrales A, Rantapää-Dahlqvist S, Sfikakis PP, Dessein P, Tsang L, Hitchon C, El-Gabalawy H, Pascual-Ramos V, Contreras-Yáñez I, Colunga-Pedraza IJ, Galarza-Delgado DA, Azpiri-Lopez JR, Semb AG, Misra DP, Hauge EM, Kitas G. Biological use influences the impact of inflammation on risk of major adverse cardiovascular events in rheumatoid arthritis. RMD Open 2024; 10:e004546. [PMID: 39043615 PMCID: PMC11268070 DOI: 10.1136/rmdopen-2024-004546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/05/2024] [Indexed: 07/25/2024] Open
Abstract
OBJECTIVES Chronic inflammation promotes cardiovascular risk in rheumatoid arthritis (RA). Biological disease-modifying antirheumatic drugs (bDMARDs) improve disease activity and cardiovascular disease outcomes. We explored whether bDMARDs influence the impact of disease activity and inflammatory markers on long-term cardiovascular risk in RA. METHODS We studied 4370 participants without cardiovascular disease in a 10-country observational cohort of patients with RA. Endpoints were (1) major adverse cardiovascular events (MACE) encompassing myocardial infarction, stroke and cardiovascular death; and (2) any ischaemic cardiovascular events (iCVE) including MACE plus revascularisation, angina, transient ischaemic attack and peripheral arterial disease. RESULTS Over 26 534 patient-years, 239 MACE and 362 iCVE occurred. The interaction between 28-joint Disease Activity Score with C-reactive protein (DAS28-CRP) and bDMARD use was significant for MACE (p=0.017), suggesting the effect of DAS28-CRP on MACE risk differed among bDMARD users (n=515) and non-users (n=3855). DAS28-CRP (per unit increase) is associated with MACE risk in bDMARD non-users (HR 1.21 (95% CI 1.07 to 1.37)) but not users (HR 0.69 (95% CI 0.40 to 1.20)). The interaction between CRP (per log unit increase) and bDMARD use was also significant for MACE (p=0.011). CRP associated with MACE risk in bDMARD non-users (HR 1.16 (95% CI 1.04 to 1.30)), but not users (HR 0.65 (95% CI 0.36 to 1.17)). No interaction was observed between bDMARD use and DAS28-CRP (p=0.167) or CRP (p=0.237) for iCVE risk. CONCLUSIONS RA activity and inflammatory markers associated with risk of MACE in bDMARD non-users but not users suggesting the possibility of biological-specific benefits locally on arterial wall independently of effects on systemic inflammation.
Collapse
Affiliation(s)
- George Athanasios Karpouzas
- Internal Medicine - Rheumatology, The Lundquist Institute, Torrance, California, USA
- Rheumatology, Harbor-UCLA Medical Center, Torrance, California, USA
| | | | | | - Miguel A Gonzalez-Gay
- Rheumatology, Hospital Universitario Marques de Valdecilla, Santander, Cantabria, Spain
- IIS-Fundacion Jimenez Diaz, Madrid, Spain
| | - Alfonso Corrales
- Hospital Universitario Marques de Valdecilla, Santander, Cantabria, Spain
| | | | - Petros P Sfikakis
- First Dept. of Propedeutic Medicine, University of Athens, Athens, Attica, Greece
| | - Patrick Dessein
- School of Physiology, University of the Witwatersrand Johannesburg, Johannesburg, South Africa
| | - Linda Tsang
- Vrije Universiteit Brussel, Brussel, Belgium
| | - Carol Hitchon
- Rheumatology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Hani El-Gabalawy
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Virginia Pascual-Ramos
- Immunology and Rheumatology, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico City, Mexico
| | - Irazú Contreras-Yáñez
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Ciudad de Mexico, Mexico
| | - Iris J Colunga-Pedraza
- Rheumatology, Hospital Universitario Dr José Eleuterio González, Monterrey, Nuevo León, Mexico
| | | | | | - Anne Grete Semb
- Rheumatology and Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Durga Prasanna Misra
- Clinical Immunology and Rheumatology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Ellen-Margrethe Hauge
- Department of Joint and Connective Tissue Diseases, Aarhus Universitetshospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus Universitet, Aarhus, Midtjylland, Denmark
| | - George Kitas
- Department of Rheumatology, The Dudley Group NHS Foundation Trust, Dudley, West Midlands, UK
| |
Collapse
|
12
|
Zhang X, Wang Z, Lin GL, Wei FZ, Zhuang YP, Xu WL, Zhang Q, Wu HT, He ZM, Yin XY, Liu Y, Mi L, Gong AM. Analysis of status and influencing factors of mental health in patients with systemic lupus erythematosus. World J Psychiatry 2024; 14:829-837. [PMID: 38984348 PMCID: PMC11230090 DOI: 10.5498/wjp.v14.i6.829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/23/2024] [Accepted: 05/06/2024] [Indexed: 06/19/2024] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune disorder with varied clinical courses and prognoses, not only did the patients suffer from physical impairment, but also various physical and psychiatric comorbidities. Growing evidence have suggested that mental disorders in SLE patients, can lead to various adverse consequences. AIM To explored the features and influencing factors of mental health in patients with SLE and clarifying the correlations between mental health and personality characteristics and perceived social support. The results would provide a basis for psychological intervention in patients with SLE. METHODS The clinical data of 168 patients with SLE admitted at the First Affiliated Hospital of Hainan Medical University between June 2020 and June 2022 were collected. Psychological assessment and correlation analysis were conducted using the Symptom Checklist-90 (SCL-90) and Perceived Social Support Scale, and the collected data were compared with the national norms in China. The relevant factors influencing mental health were identified by statistical analysis. A general information questionnaire, the Revised Life Orientation Test, and Short-Form 36-Item Health Survey were employed to assess optimism level and quality of life (QoL), respectively. RESULTS Patients with SLE obtained higher scores for the somatization, depression, anxiety, and phobic anxiety subscales than national norms (P < 0.05). A correlation was identified between total social support and total SCL-90 score or each subscale (P < 0.05). The factors significantly affecting patients' mental health were hormone dosage and disease activity index (DAI) (P < 0.05). The average optimism score of patients with SLE was 14.36 ± 4.42, and 30 cases were in the middle and lower levels. A positive correlation was found between optimism level and QoL scores. CONCLUSION Patients with SLE develop psychological disorders at varying degrees, which are significantly influenced by hormone dosage and DAI. Patients' mental health should be closely monitored during clinical diagnosis and treatment and provided adequate support in establishing positive, healthy thinking and behavior patterns and improving their optimism level and QoL.
Collapse
Affiliation(s)
- Xuan Zhang
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou 571101, Hainan Province, China
| | - Zhe Wang
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou 571101, Hainan Province, China
| | - Gui-Ling Lin
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou 571101, Hainan Province, China
| | - Fang-Zhi Wei
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou 571101, Hainan Province, China
| | - Yan-Ping Zhuang
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou 571101, Hainan Province, China
| | - Wen-Lu Xu
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou 571101, Hainan Province, China
| | - Qi Zhang
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou 571101, Hainan Province, China
| | - Hui-Tao Wu
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou 571101, Hainan Province, China
| | - Zi-Man He
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou 571101, Hainan Province, China
| | - Xi-Yu Yin
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou 571101, Hainan Province, China
| | - Ying Liu
- Department of Rheumatology and Immunology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou 570311, Hainan Province, China
| | - Long Mi
- Department of Radiology, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 570100, Hainan Province, China
| | - Ai-Min Gong
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou 571101, Hainan Province, China
| |
Collapse
|
13
|
Kondratyeva LV, Popkova TV, Nasonov EL. [The effect of hypothyroidism on cardiovascular events and type 2 diabetes mellitus developing in rheumatoid arthritis]. TERAPEVT ARKH 2024; 96:459-464. [PMID: 38829806 DOI: 10.26442/00403660.2024.05.202700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 06/05/2024]
Abstract
AIM To compare the frequency of cardiovascular events (CVE), to assess the risk of cardiovascular death using the mSCORE and the development of type 2 diabetes mellitus (DM) using the FINDRISC in patients with rheumatoid arthritis (RA) with and without hypothyroidism. MATERIALS AND METHODS The study included 149 patients (125 women, 24 men) with RA (median age - 57 [52; 61] years). In all patients, traditional factors of cardiovascular risk and glucose metabolism disorders (age, smoking status, total blood cholesterol, blood pressure, overweight, abdominal obesity - AO, heredity burdened by diabetes, insufficient physical activity, the lack of the necessary amount of berries, fruits and vegetables in the daily diet, history of hyperglycemia episodes), the 10-year risk of death from cardiovascular causes according to the mSCORE and the risk of developing type 2 DM according to the FINDRISС were assessed, a history of CVE (myocardial infarctions, and its revascularization, stroke) was recorded. RESULTS Hypothyroidism was diagnosed in 17.4% of RA patients. Patients with hypothyroidism (group 1) were more likely to have AO and less likely to consume unsufficient dietary fiber than patients with euthyroidism (group 2). Moderate, high and very high risk of development according to the mSCORE and FINDRISC was detected in 61.5% of hypothyroid patients and 48.8% euthyroid patients, according to mSCORE alone - in 30.8 and 44.7%, according to FINDRISC - in 0 and 2.4%, respectively (p>0.05 in all cases); 11.5% of patients in group 1 and 6.5% in group 2 suffered from CVE (OR 1.875, 95% CI 0.462-7.607; p=0.63). CONCLUSION It is necessary to evaluate the thyroid gland function, especially in patients with AO due to the high frequency of hypothyroidism in RA. Hypothyroidism did not have an independent effect on the severe CVЕ rates, as well as risk assessment according to the score and FINDRISC in RA patients. Theses, with and without hypothyroidism, were predominantly in the moderate, high, very high risk groups according to both scales.
Collapse
Affiliation(s)
| | | | - E L Nasonov
- Nasonova Research Institute of Rheumatology
- Sechenov First Moscow State Medical University (Sechenov University)
| |
Collapse
|
14
|
Galajda NÁ, Meznerics FA, Mátrai P, Fehérvári P, Lengyel AS, Kolonics MV, Sipos Z, Kemény LV, Csupor D, Hegyi P, Bánvölgyi A, Holló P. Reducing cardiovascular risk in immune-mediated inflammatory diseases: Tumour necrosis factor inhibitors compared to conventional therapies-A systematic review and meta-analysis. J Eur Acad Dermatol Venereol 2024; 38:1070-1088. [PMID: 38433519 DOI: 10.1111/jdv.19900] [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: 10/02/2023] [Accepted: 01/19/2024] [Indexed: 03/05/2024]
Abstract
Immune-mediated inflammatory disease (IMID) patients including psoriasis, inflammatory arthritides and bowel diseases have a higher risk of developing cardiovascular (CV) diseases compared to the general population. The increased CV risk may be promoted by tumour necrosis factor (TNF)-α-mediated immunological processes, which are present both in the pathomechanism of IMIDs and atherosclerosis. Our objective was to comprehensively investigate the effect of TNF inhibitors (TNFi) on CV risk compared with conventional therapies in IMIDs. The systematic literature search was conducted in three databases (MEDLINE, EMBASE, Cochrane Library) on 14 November 2022. Randomized controlled trials, cohort and case-control studies were eligible for inclusion. Outcomes consisted of the incidence of CV events, with major adverse cardiovascular events (MACE) as a main endpoint. A random-effects meta-analysis was performed by pooling fully adjusted multivariate hazard ratios (HR) and incidence rate ratios (IRR) with a 95% confidence interval (CI) comparing TNFis with conventional systemic non-biologicals (CSNBs). Of a total of 8724 search results, 56 studies were included overall, of which 29 articles were eligible for the meta-analysis, and 27 were involved in the systematic review. Including all IMIDs, the TNFi group showed a significantly reduced risk of MACE compared with the CSNB group (HR = 0.74, 95% confidence interval (CI) 0.58-0.95, p = 0.025; IRR = 0.77, 95% CI 0.67-0.88, p < 0.001). Subgroup analysis of Pso, PsA patients by pooling IRRs also confirmed the significantly decreased risk of MACE in TNFi-treated patients compared with CSNB groups (IRR = 0.79, 95% CI 0.64-0.98). The observational nature of most included studies leading to high heterogeneity represents a limitation. Based on the results, TNFis may reduce the risk of CV events compared to CSNBs. Therefore, earlier use of TNFis compared to conventional systemic agents in the therapeutic sequence may benefit CV risk in IMID patients.
Collapse
Affiliation(s)
- N Á Galajda
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - F A Meznerics
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - P Mátrai
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - P Fehérvári
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Department of Biostatistics, University of Veterinary Medicine, Budapest, Hungary
| | - A S Lengyel
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- HCEMM-SU Translational Dermatology Research Group, Department of Physiology, Semmelweis University, Budapest, Hungary
| | - M V Kolonics
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Z Sipos
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - L V Kemény
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- HCEMM-SU Translational Dermatology Research Group, Department of Physiology, Semmelweis University, Budapest, Hungary
| | - D Csupor
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Institute of Clinical Pharmacy, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - P Hegyi
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
- Institute of Pancreatic Diseases, Semmelweis University, Budapest, Hungary
| | - A Bánvölgyi
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - P Holló
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
15
|
Zaragoza-García O, Briceño O, Villafan-Bernal JR, Gutiérrez-Pérez IA, Rojas-Delgado HU, Alonso-Silverio GA, Alarcón-Paredes A, Navarro-Zarza JE, Morales-Martínez C, Rodríguez-García R, Guzmán-Guzmán IP. Levels of sCD163 in women rheumatoid arthritis: Relationship with cardiovascular risk markers. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024:S0214-9168(24)00039-1. [PMID: 38729859 DOI: 10.1016/j.arteri.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 05/12/2024]
Abstract
AIM The soluble scavenger receptor differentiation antigen 163 (sCD163), a monocyte/macrophage activation marker, is related to cardiovascular mortality in the general population. This study aimed to evaluate their relationship between serum levels of sCD163 with cardiovascular risk indicators in rheumatoid arthritis (RA). METHODS A cross-sectional study was performed on 80 women diagnosed with RA. The cardiovascular risks were determined using the lipid profile, metabolic syndrome, and QRISK3 calculator. For the assessment of RA activity, we evaluated the DAS28 with erythrocyte sedimentation rate (DAS28-ESR). The serum levels of sCD163 were determined by the ELISA method. Logistic regression models and receiver operating characteristics (ROC) curve were used to assess the association and predictive value of sCD163 with cardiovascular risk in RA patients. RESULTS Levels of sCD163 were significantly higher in RA patients with high sensitivity protein C-reactive to HDL-c ratio (CHR)≥0.121 (p=0.003), total cholesterol/HDL-c ratio>7% (p=0.004), LDL-c/HDL-c ratio>3% (p=0.035), atherogenic index of plasma>0.21 (p=0.004), cardiometabolic index (CMI)≥1.70 (p=0.005), and high DAS28-ESR (p=0.004). In multivariate analysis, levels of sCD163≥1107.3ng/mL were associated with CHR≥0.121 (OR=3.43, p=0.020), CMI≥1.70 (OR=4.25, p=0.005), total cholesterol/HDL-c ratio>7% (OR=6.63, p=0.044), as well as with DAS28-ESR>3.2 (OR=8.10, p=0.008). Moreover, levels of sCD163 predicted CHR≥0.121 (AUC=0.701), cholesterol total/HDL ratio>7% (AUC=0.764), and DAS28-ESR>3.2 (AUC=0.720). CONCLUSION Serum levels of sCD163 could be considered a surrogate of cardiovascular risk and clinical activity in RA.
Collapse
Affiliation(s)
- Oscar Zaragoza-García
- Laboratory of Multidisciplinary Research and Biomedical Innovation, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Olivia Briceño
- Infectious Diseases Research Center, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - José Rafael Villafan-Bernal
- Laboratory of Immunogenomics and Metabolic Diseases, Instituto Nacional de Medicina Genomica, Mexico City, Mexico
| | - Ilse Adriana Gutiérrez-Pérez
- Laboratory of Multidisciplinary Research and Biomedical Innovation, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | | | - Gustavo Adolfo Alonso-Silverio
- Laboratory of Multidisciplinary Research and Biomedical Innovation, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Antonio Alarcón-Paredes
- Laboratory of Multidisciplinary Research and Biomedical Innovation, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | | | | | - Rubén Rodríguez-García
- Laboratorio de Clínico, Instituto Mexicano del Seguro Social, Hospital General Regional, Cuernavaca, Morelos, Mexico
| | - Iris Paola Guzmán-Guzmán
- Laboratory of Multidisciplinary Research and Biomedical Innovation, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico.
| |
Collapse
|
16
|
Su QY, Li HC, Jiang XJ, Jiang ZQ, Zhang Y, Zhang HY, Zhang SX. Exploring the therapeutic potential of regulatory T cell in rheumatoid arthritis: Insights into subsets, markers, and signaling pathways. Biomed Pharmacother 2024; 174:116440. [PMID: 38518605 DOI: 10.1016/j.biopha.2024.116440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 03/24/2024] Open
Abstract
Rheumatoid arthritis (RA) is a complex autoimmune inflammatory rheumatic disease characterized by an imbalance between immunological reactivity and immune tolerance. Regulatory T cells (Tregs), which play a crucial role in controlling ongoing autoimmunity and maintaining peripheral tolerance, have shown great potential for the treatment of autoimmune inflammatory rheumatic diseases such as RA. This review aims to provide an updated summary of the latest insights into Treg-targeting techniques in RA. We focus on current therapeutic strategies for targeting Tregs based on discussing their subsets, surface markers, suppressive function, and signaling pathways in RA.
Collapse
Affiliation(s)
- Qin-Yi Su
- The Second Hospital of Shanxi Medical University, Department of Rheumatology, Taiyuan, China; Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - Huan-Cheng Li
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - Xiao-Jing Jiang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - Zhong-Qing Jiang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - Yan Zhang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - He-Yi Zhang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - Sheng-Xiao Zhang
- The Second Hospital of Shanxi Medical University, Department of Rheumatology, Taiyuan, China; Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China.
| |
Collapse
|
17
|
Dhital R, Baer RJ, Bandoli G, Guma M, Poudel DR, Kalunian K, Weisman MH, Chambers C. Maternal cardiovascular events in autoimmune rheumatic diseases and antiphospholipid syndrome pregnancies. Am J Obstet Gynecol MFM 2024; 6:101319. [PMID: 38428526 PMCID: PMC11378732 DOI: 10.1016/j.ajogmf.2024.101319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/15/2024] [Accepted: 02/21/2024] [Indexed: 03/03/2024]
Affiliation(s)
- Rashmi Dhital
- Division of Rheumatology, Department of Medicine, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0656.
| | - Rebecca J Baer
- Division of Environmental Science and Health, Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA; The California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA
| | - Gretchen Bandoli
- Division of Environmental Science and Health, Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA; Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, CA
| | - Monica Guma
- Division of Rheumatology, Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA; Department of Rheumatology, Veteran Affairs San Diego Healthcare System, San Diego, CA
| | - Dilli R Poudel
- Department of Medicine, Indiana Regional Medical Center, Indiana, PA
| | - Kenneth Kalunian
- Division of Rheumatology, Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA
| | - Michael H Weisman
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Palo Alto, CA
| | - Christina Chambers
- Division of Environmental Science and Health, Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA; Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, CA
| |
Collapse
|
18
|
Ikdahl E, Stensrud MJ. Re-evaluating the mythical divide between traditional and novel cardiovascular risk factors in rheumatoid arthritis. RMD Open 2024; 10:e003954. [PMID: 38428975 PMCID: PMC10910651 DOI: 10.1136/rmdopen-2023-003954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/16/2024] [Indexed: 03/03/2024] Open
Abstract
Cardiovascular (CV) risk factors for rheumatoid arthritis (RA) are conventionally classified as 'traditional' and 'novel'. We argue that this classification is obsolete and potentially counterproductive. Further, we discuss problems with the common practice of adjusting for traditional CV risk factors in statistical analyses. These analyses do not target well-defined effects of RA on CV risk. Ultimately, we propose a future direction for cardiorheumatology research that prioritises optimising current treatments and identifying novel therapeutic targets over further categorisation of well-known risk factors.
Collapse
Affiliation(s)
- Eirik Ikdahl
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Mats Julius Stensrud
- Department of Mathematics, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
19
|
Di Muzio C, Di Cola I, Shariat Panahi A, Ursini F, Iagnocco A, Giacomelli R, Cipriani P, Ruscitti P. The effects of suppressing inflammation by tofacitinib may simultaneously improve glycaemic parameters and inflammatory markers in rheumatoid arthritis patients with comorbid type 2 diabetes: a proof-of-concept, open, prospective, clinical study. Arthritis Res Ther 2024; 26:14. [PMID: 38178250 PMCID: PMC10765862 DOI: 10.1186/s13075-023-03249-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND A consistent connection has been increasingly reported between rheumatoid arthritis (RA), insulin resistance (IR), and type 2 diabetes (T2D). The β-cell apoptosis induced by pro-inflammatory cytokines, which could be exaggerated in the context of RA, is associated with increased expression pro-apoptotic proteins, which is dependent on JAnus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) activation. On these bases, we aimed to evaluate if the administration of tofacitinib, a potent and selective JAK inhibitor, could simultaneously improve glycaemic parameters and inflammatory markers in patients with RA and comorbid T2D. METHODS The primary endpoint was the change in the 1998-updated homeostatic model assessment of IR (HOMA2-IR) after 6 months of treatment with tofacitinib in RA patients with T2D. Consecutive RA patients with T2D diagnosis were included in this proof-of-concept, open, prospective, clinical study, which was planned before the recent emergence of safety signals about tofacitinib. Additional endpoints were also assessed regarding RA disease activity and metabolic parameters. RESULTS Forty consecutive RA patients with T2D were included (female sex 68.9%, mean age of 63.4 ± 9.9 years). During 6-month follow-up, a progressive reduction of HOMA2-IR was observed in RA patients with T2D treated with tofacitinib. Specifically, a significant effect of tofacitinib was shown on the overall reduction of HOMA2-IR (β = - 1.1, p = 0.019, 95%CI - 1.5 to - 0.76). Also, HOMA2-β enhanced in these patients highlighting an improvement of insulin sensitivity. Furthermore, although a longer follow-up is required, a trend in glycated haemoglobin reduction was also recorded. The administration of tofacitinib induced an improvement in RA disease activity, and a significant reduction of DAS28-CRP and SDAI was observed; 76.8% of patients achieved a good clinical response. In this study, no major adverse events (AEs) were retrieved without the identification of new safety signals. Specifically, no life-threatening AEs and cardiovascular and/or thromboembolic events were recorded. CONCLUSIONS The administration of tofacitinib in RA with T2D led to a simultaneous improvement of IR and inflammatory disease activity, inducing a "bidirectional" benefit in these patients. However, further specific designed and powered studies are warranted to entirely evaluate the metabolic effects of tofacitinib in RA patients with T2D.
Collapse
Affiliation(s)
- Claudia Di Muzio
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, PO box 67100, L'Aquila, Italy
| | - Ilenia Di Cola
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, PO box 67100, L'Aquila, Italy
| | - Azadeh Shariat Panahi
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, PO box 67100, L'Aquila, Italy
| | - Francesco Ursini
- Medicine and Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Annamaria Iagnocco
- Academic Rheumatology Centre, Dipartimento di Scienze Cliniche e Biologiche Università di Torino - AO Mauriziano di Torino, Turin, Italy
| | - Roberto Giacomelli
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Campus Bio-Medico, Via Álvaro del Portillo 200, 00128, Rome, Italy
- Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Paola Cipriani
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, PO box 67100, L'Aquila, Italy
| | - Piero Ruscitti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, PO box 67100, L'Aquila, Italy.
| |
Collapse
|
20
|
Karakasis P, Patoulias D, Stachteas P, Lefkou E, Dimitroulas T, Fragakis N. Accelerated Atherosclerosis and Management of Cardiovascular Risk in Autoimmune Rheumatic Diseases: An Updated Review. Curr Probl Cardiol 2023; 48:101999. [PMID: 37506959 DOI: 10.1016/j.cpcardiol.2023.101999] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 07/24/2023] [Indexed: 07/30/2023]
Abstract
Even though diagnosis and management pathways have been substantially improved over the last years, autoimmune rheumatic diseases (AIRDs) such as rheumatoid arthritis, systemic sclerosis, systemic lupus erythematosus, antiphospholipid syndrome, Sjögren's syndrome, and systemic vasculitides have been linked to elevated rates of cardiovascular morbidity and mortality, primarily secondary to accelerated atherosclerosis. This phenomenon can be partially attributed to the presence of established cardiovascular risk factors but may also be a result of other inflammatory and autoimmune mechanisms that are enhanced in AIRDs. According to the current guidelines, the recommendations regarding cardiovascular disease prevention in patients with AIRDs are not significantly different from those applied to the general population. Herein, we present a review of the current literature on the risk of accelerated atherosclerosis in AIRDs and provide a summary of available recommendations for the management of cardiovascular risk in rheumatic diseases.
Collapse
Affiliation(s)
- Paschalis Karakasis
- Second Department of Cardiology, Aristotle University of Thessaloniki, General Hospital "Hippokration," Thessaloniki, Greece.
| | - Dimitrios Patoulias
- Second Department of Cardiology, Aristotle University of Thessaloniki, General Hospital "Hippokration," Thessaloniki, Greece; Outpatient Department of Cardiometabolic Medicine, Second Department of Cardiology, Aristotle University of Thessaloniki, General Hospital "Hippokration," Thessaloniki, Greece; Second Department of Internal Medicine, European Interbalkan Medical Center, Thessaloniki, Greece
| | - Panagiotis Stachteas
- Second Department of Cardiology, Aristotle University of Thessaloniki, General Hospital "Hippokration," Thessaloniki, Greece
| | - Eleftheria Lefkou
- Second Department of Cardiology, Aristotle University of Thessaloniki, General Hospital "Hippokration," Thessaloniki, Greece; Perigenesis, Institute of Obstetric Haematology, Thessaloniki, Greece
| | - Theodoros Dimitroulas
- Second Department of Cardiology, Aristotle University of Thessaloniki, General Hospital "Hippokration," Thessaloniki, Greece; Fourth Department of Internal Medicine, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Fragakis
- Second Department of Cardiology, Aristotle University of Thessaloniki, General Hospital "Hippokration," Thessaloniki, Greece
| |
Collapse
|
21
|
Guo Y, Chung W, Shan Z, Zhu Z, Costenbader KH, Liang L. Genome-Wide Assessment of Shared Genetic Architecture Between Rheumatoid Arthritis and Cardiovascular Diseases. J Am Heart Assoc 2023; 12:e030211. [PMID: 37947095 PMCID: PMC10727280 DOI: 10.1161/jaha.123.030211] [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: 03/16/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Patients with rheumatoid arthritis (RA) have a 2- to 10-fold increased risk of cardiovascular disease (CVD), but the biological mechanisms and existence of causality underlying such associations remain to be investigated. We aimed to investigate the genetic associations and underlying mechanisms between RA and CVD by leveraging large-scale genomic data and genetic cross-trait analytic approaches. METHODS AND RESULTS Within UK Biobank data, we examined the genetic correlation, shared genetics, and potential causality between RA (Ncases=6754, Ncontrols=452 384) and cardiovascular diseases (CVD, Ncases=44 238, Ncontrols=414 900) using linkage disequilibrium score regression, cross-trait meta-analysis, and Mendelian randomization. We observed significant genetic correlations of RA with myocardial infarction (rg:0.40 [95% CI, 0.24-0.56), angina (rg:0.42 [95% CI, 0.28-0.56]), coronary heart diseases (rg:0.41 [95% CI, 0.27-0.55]), and CVD (rg:0.43 [95% CI, 0.29-0.57]) after correcting for multiple testing (P<0.05/5). When stratified by frequent use of analgesics, we found increased genetic correlation between RA and CVD among participants without aspirin usage (rg:0.54 [95% CI, 0.30-0.78] for angina; Pvalue=6.69×10-6) and among participants with paracetamol usage (rg:0.75 [95% CI, 0.20-1.29] for myocardial infarction; Pvalue=8.90×10-3), whereas others remained similar. Cross-trait meta-analysis identified 9 independent shared loci between RA and CVD, including PTPN22 at chr1p13.2, BCL2L11 at chr2q13, and CCR3 at chr3p21.31 (Psingle trait<1×10-3 and Pmeta<5×10-8), highlighting potential shared pathogenesis including accelerating atherosclerosis, upregulating oxidative stress, and vascular permeability. Finally, Mendelian randomization estimates showed limited evidence of causality between RA and CVD. CONCLUSIONS Our results supported shared genetic pathogenesis rather than causality in explaining the observed association between RA and CVD. The identified shared genetic factors provided insights into potential novel therapeutic target for RA-CVD comorbidities.
Collapse
Affiliation(s)
- Yanjun Guo
- Department of Occupational and Environmental Health, School of Public HealthTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Program in Genetic Epidemiology and Statistical Genetics, Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMAUSA
- Division of Preventive MedicineBrigham and Women’s HospitalBostonMA
- Harvard Medical SchoolBostonMA
| | - Wonil Chung
- Program in Genetic Epidemiology and Statistical Genetics, Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMAUSA
- Department of Statistics and Actuarial ScienceSoongsil UniversitySeoulKorea
| | - Zhilei Shan
- Department of NutritionHarvard T.H. Chan School of Public HealthBostonMA
| | - Zhaozhong Zhu
- Program in Genetic Epidemiology and Statistical Genetics, Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMAUSA
| | - Karen H. Costenbader
- Division of Preventive MedicineBrigham and Women’s HospitalBostonMA
- Division of Rheumatology, Inflammation and Immunity, Department of MedicineBrigham and Women’s HospitalBostonMA
| | - Liming Liang
- Program in Genetic Epidemiology and Statistical Genetics, Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMAUSA
- Department of BiostatisticsHarvard T.H. Chan School of Public HealthBostonMA
| |
Collapse
|
22
|
Mengozzi A, de Ciuceis C, Dell'oro R, Georgiopoulos G, Lazaridis A, Nosalski R, Pavlidis G, Tual-Chalot S, Agabiti-Rosei C, Anyfanti P, Camargo LL, Dąbrowska E, Quarti-Trevano F, Hellmann M, Masi S, Mavraganis G, Montezano AC, Rios FJ, Winklewski PJ, Wolf J, Costantino S, Gkaliagkousi E, Grassi G, Guzik TJ, Ikonomidis I, Narkiewicz K, Paneni F, Rizzoni D, Stamatelopoulos K, Stellos K, Taddei S, Touyz RM, Triantafyllou A, Virdis A. The importance of microvascular inflammation in ageing and age-related diseases: a position paper from the ESH working group on small arteries, section of microvascular inflammation. J Hypertens 2023; 41:1521-1543. [PMID: 37382158 DOI: 10.1097/hjh.0000000000003503] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
Microcirculation is pervasive and orchestrates a profound regulatory cross-talk with the surrounding tissue and organs. Similarly, it is one of the earliest biological systems targeted by environmental stressors and consequently involved in the development and progression of ageing and age-related disease. Microvascular dysfunction, if not targeted, leads to a steady derangement of the phenotype, which cumulates comorbidities and eventually results in a nonrescuable, very high-cardiovascular risk. Along the broad spectrum of pathologies, both shared and distinct molecular pathways and pathophysiological alteration are involved in the disruption of microvascular homeostasis, all pointing to microvascular inflammation as the putative primary culprit. This position paper explores the presence and the detrimental contribution of microvascular inflammation across the whole spectrum of chronic age-related diseases, which characterise the 21st-century healthcare landscape. The manuscript aims to strongly affirm the centrality of microvascular inflammation by recapitulating the current evidence and providing a clear synoptic view of the whole cardiometabolic derangement. Indeed, there is an urgent need for further mechanistic exploration to identify clear, very early or disease-specific molecular targets to provide an effective therapeutic strategy against the otherwise unstoppable rising prevalence of age-related diseases.
Collapse
Affiliation(s)
- Alessandro Mengozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa
| | - Carolina de Ciuceis
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia
| | - Raffaella Dell'oro
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Georgios Georgiopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Athens
| | - Antonios Lazaridis
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - Ryszard Nosalski
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute; University of Edinburgh, University of Edinburgh, Edinburgh, UK
- Department of Internal Medicine
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - George Pavlidis
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2 Cardiology Department, Attikon Hospital, Athens
- Medical School, National and Kapodistrian University of Athens, Greece
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | | | - Panagiota Anyfanti
- Second Medical Department, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Livia L Camargo
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | - Edyta Dąbrowska
- Department of Hypertension and Diabetology, Center of Translational Medicine
- Center of Translational Medicine
| | - Fosca Quarti-Trevano
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Marcin Hellmann
- Department of Cardiac Diagnostics, Medical University, Gdansk, Poland
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Institute of Cardiovascular Science, University College London, London, UK
| | - Georgios Mavraganis
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Athens
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | - Francesco J Rios
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | | | - Jacek Wolf
- Department of Hypertension and Diabetology, Center of Translational Medicine
| | - Sarah Costantino
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- University Heart Center, Cardiology, University Hospital Zurich
| | - Eugenia Gkaliagkousi
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - Guido Grassi
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Tomasz J Guzik
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute; University of Edinburgh, University of Edinburgh, Edinburgh, UK
- Department of Internal Medicine
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - Ignatios Ikonomidis
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2 Cardiology Department, Attikon Hospital, Athens
- Medical School, National and Kapodistrian University of Athens, Greece
| | | | - Francesco Paneni
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- University Heart Center, Cardiology, University Hospital Zurich
- Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Damiano Rizzoni
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia
- Division of Medicine, Spedali Civili di Brescia, Montichiari, Brescia, Italy
| | - Kimon Stamatelopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Athens
| | - Konstantinos Stellos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site
- Department of Cardiology, University Hospital Mannheim, Heidelberg University, Manheim, Germany
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | - Areti Triantafyllou
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - Agostino Virdis
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
23
|
Mercader-Salvans J, García-González M, Gómez-Bernal F, Quevedo-Abeledo JC, de Vera-González A, González-Delgado A, López-Mejías R, Martín-González C, González-Gay MÁ, Ferraz-Amaro I. Relationship between Disease Characteristics and Circulating Interleukin 6 in a Well-Characterized Cohort of Patients with Systemic Lupus Erythematosus. Int J Mol Sci 2023; 24:14006. [PMID: 37762312 PMCID: PMC10531425 DOI: 10.3390/ijms241814006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Interleukin-6 (IL-6) is a proinflammatory cytokine that mediates pleiotropic functions in immune responses and inflammatory diseases. The literature lacks studies, with a clinical perspective, on the relationship between IL-6 serum levels and the characteristics of the disease in patients with systemic lupus erythematosus (SLE). In the present work, we aimed to analyze the association between circulating IL-6 and disease manifestations in a well-characterized series of patients with SLE. Serum IL-6 levels and disease activity (SLEDAI-2K), severity (Katz) and damage index (SLICC-DI), complete lipid profile, and subclinical carotid atherosclerosis were evaluated in 284 patients with SLE. In addition, a complete characterization of the complement system was performed in samples from patients with SLE. A multivariate linear regression analysis was carried out to study the relationship between clinical and laboratory characteristics of the disease and IL-6 levels. Age (beta coef. 0.07 [95%CI 0.01-0.1] pg/mL, p = 0.014), C-reactive protein (beta coef. 0.21 [95%CI 0.16-0.25] pg/mL, p < 0.01), and male gender (beta coef. 2 [95%CI 0.3-0.5] pg/mL, p = 0.024), were positively associated with higher IL-6 levels in SLE patients. Most disease characteristics and damage and activity indices did not show significant relationships with IL-6. However, after multivariate analysis, IL-6 was associated with lower serum levels of HDL cholesterol (beta coef. -0.04 [95%CI -0.08-(-0.1)] pg/mL, p = 0.011), and apolipoprotein A1 (beta coef. -0.02 [95%CI -0.04-(-0.001)] pg/mL, p = 0.035). In contrast, the alternative complement cascade, C1inh, and C3a were all positively and independently associated with higher serum levels of IL-6. Moreover, stratification of the Systematic Coronary Risk Assessment 2 (SCORE2) results according to different categories of cardiovascular risk was associated with higher circulating serum IL-6 levels (beta coef. 0.2 [95%CI 0.02-0.4], pg/mL, p = 0.028). In conclusion, in a large series of SLE patients, IL-6 was not associated with disease-related features of SLE, including damage, severity, or activity indices. However, an association was found between serum IL-6 levels and circulating C3a and cardiovascular risk. Our study emphasizes the importance that IL-6 could have in cardiovascular disease and complement system disruption of SLE patients. Therapies targeting IL-6 could have a role in these two clinical manifestations of patients with SLE.
Collapse
Affiliation(s)
| | - María García-González
- Division of Rheumatology, Hospital Universitario de Canarias, 38320 Tenerife, Spain;
| | - Fuensanta Gómez-Bernal
- Division of Central Laboratory, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (F.G.-B.); (A.d.V.-G.); (A.G.-D.)
| | | | - Antonia de Vera-González
- Division of Central Laboratory, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (F.G.-B.); (A.d.V.-G.); (A.G.-D.)
| | - Alejandra González-Delgado
- Division of Central Laboratory, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (F.G.-B.); (A.d.V.-G.); (A.G.-D.)
| | - Raquel López-Mejías
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, IDIVAL, 39011 Santander, Spain;
| | - Candelaria Martín-González
- Division of Internal Medicine, Hospital Universitario de Canarias, 38320 Tenerife, Spain;
- Department of Internal Medicine, University of La Laguna (ULL), 38200 Tenerife, Spain
| | - Miguel Á. González-Gay
- Division of Rheumatology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
- Department of Medicine, University of Cantabria, 39005 Santander, Spain
| | - Iván Ferraz-Amaro
- Division of Rheumatology, Hospital Universitario de Canarias, 38320 Tenerife, Spain;
- Department of Internal Medicine, University of La Laguna (ULL), 38200 Tenerife, Spain
| |
Collapse
|
24
|
Merino de Paz N, García-González M, Gómez-Bernal F, Quevedo-Abeledo JC, de Vera-González A, López-Mejias R, Abreu-González P, Martín-González C, González-Gay MÁ, Ferraz-Amaro I. Relationship between Malondialdehyde Serum Levels and Disease Features in a Full Characterized Series of 284 Patients with Systemic Lupus Erythematosus. Antioxidants (Basel) 2023; 12:1535. [PMID: 37627530 PMCID: PMC10451961 DOI: 10.3390/antiox12081535] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/20/2023] [Accepted: 07/29/2023] [Indexed: 08/27/2023] Open
Abstract
Malondialdehyde (MDA) is a marker of oxidative stress and antioxidant status. Oxidative stress has been observed to be increased in systemic lupus erythematosus (SLE). Some studies have shown that MDA is upregulated in SLE compared to controls. However, the literature lacks reports regarding the relationship of MDA to disease manifestations. This is relevant since SLE is a multisystemic disease which may affect virtually any organ in the body. In this study, we set out to analyze how MDA serum levels are associated with disease expression in a large series of SLE patients who were fully characterized in clinical and laboratory terms. A total of 284 patients with SLE were recruited. Serum levels of MDA, and the activity (SLEDAI), severity (Katz) and damage index (SLICC-DI) scores, full lipid profile, and carotid subclinical atherosclerosis were assessed. In addition, a full characterization of the complement system was performed in SLE patients' samples. Multivariable linear regression analysis was executed to study the relationship between clinical and laboratory disease characteristics and MDA. A statistically significant negative relationship was found between disease duration and MDA. In contrast, the presence of anti-nucleosome antibodies was positively associated with MDA. Regarding the SLICC-DI areas, both the musculoskeletal domain and the cutaneous domain were significantly related to higher serum MDA values. Furthermore, after adjustment for confounding factors, lower levels of the classical complement pathway, which denotes activation, were associated with higher serum levels of MDA. In conclusion, cumulative musculoskeletal and skin damage in SLE patients is associated with superior serum levels of MDA. In addition, activation of the complement system is also related to higher circulating MDA levels.
Collapse
Affiliation(s)
- Nayra Merino de Paz
- Division of Dermatology, Dermamedicin Clínicas, 38004 Santa Cruz de Tenerife, Spain;
| | - María García-González
- Division of Rheumatology, Hospital Universitario de Canarias, 38320 Santa Cruz de Tenerife, Spain;
| | - Fuensanta Gómez-Bernal
- Division of Central Laboratory, Hospital Universitario de Canarias, 38320 Santa Cruz de Tenerife, Spain; (F.G.-B.); (A.d.V.-G.)
| | | | - Antonia de Vera-González
- Division of Central Laboratory, Hospital Universitario de Canarias, 38320 Santa Cruz de Tenerife, Spain; (F.G.-B.); (A.d.V.-G.)
| | - Raquel López-Mejias
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, IDIVAL, 39011 Santander, Spain;
| | - Pedro Abreu-González
- Unit of Physiology, Department of Basic Medical Sciences, University of La Laguna, 38200 Santa Cruz de Tenerife, Spain;
| | - Candelaria Martín-González
- Division of Internal Medicine, Hospital Universitario de Canarias, 38320 Santa Cruz de Tenerife, Spain;
- Department of Internal Medicine, University of La Laguna (ULL), 38200 Santa Cruz de Tenerife, Spain
| | - Miguel Á. González-Gay
- Division of Rheumatology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
- Department of Medicine, University of Cantabria, 39005 Santander, Spain
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Iván Ferraz-Amaro
- Division of Rheumatology, Hospital Universitario de Canarias, 38320 Santa Cruz de Tenerife, Spain;
- Department of Internal Medicine, University of La Laguna (ULL), 38200 Santa Cruz de Tenerife, Spain
| |
Collapse
|
25
|
Patoulias D, Dimosiari A, Michailidis T. Should lipoprotein(a) be routinely measured after COVID-19? Comment on the article by Stahl et al. Arthritis Rheumatol 2023; 75:1296. [PMID: 36691804 DOI: 10.1002/art.42453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/03/2023] [Indexed: 01/25/2023]
Affiliation(s)
| | - Athina Dimosiari
- Second Department of Internal Medicine, European Interbalkan Medical Center
| | - Theodoros Michailidis
- Second Department of Internal Medicine, Aristotle University of Thessaloniki, General Hospital "Hippokration", Thessaloniki, Greece
| |
Collapse
|
26
|
Triantafyllias K, Thiele LE, Cavagna L, Baraliakos X, Bertsias G, Schwarting A. Arterial Stiffness as a Surrogate Marker of Cardiovascular Disease and Atherosclerosis in Patients with Arthritides and Connective Tissue Diseases: A Literature Review. Diagnostics (Basel) 2023; 13:diagnostics13111870. [PMID: 37296720 DOI: 10.3390/diagnostics13111870] [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: 04/13/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
The increased cardiovascular (CV) risk among patients with autoimmune rheumatic diseases, such as arthritides and connective tissue diseases, has been extensively documented. From a pathophysiological standpoint, systemic inflammation in the context of the disease can lead to endothelial dysfunction, accelerated atherosclerosis, and structural changes in vessel walls, which, in turn, are associated with exaggerated CV morbidity and mortality. In addition to these abnormalities, the increased prevalence of traditional CV risk factors, such as obesity, dyslipidemia, arterial hypertension, and impaired glucose metabolism, can further worsen the status of and overall prognosis for CV in rheumatic patients. However, data on appropriate CV screening methods for patients with systemic autoimmune diseases are scarce, and traditional algorithms may lead to an underestimation of the true CV risk. The reason for this is that these calculations were developed for the general population and thus do not take into account the effect of the inflammatory burden, as well as other chronic-disease-associated CV risk factors. In recent years, different research groups, including ours, have examined the value of different CV surrogate markers, including carotid sonography, carotid-femoral pulse wave velocity, and flow-mediated arterial dilation, in the assessment of CV risk in healthy and rheumatic populations. In particular, arterial stiffness has been thoroughly examined in a number of studies, showing high diagnostic and predictive value for the occurrence of CV events. To this end, the present narrative review showcases a series of studies examining aortic and peripheral arterial stiffness as surrogates of all-cause CV disease and atherosclerosis in patients with rheumatoid and psoriatic arthritis, as well as in systemic lupus erythematosus and systemic sclerosis. Moreover, we discuss the associations of arterial stiffness with clinical, laboratory, and disease-specific parameters.
Collapse
Affiliation(s)
- Konstantinos Triantafyllias
- Rheumatology Center Rhineland-Palatinate, Kaiser-Wilhelm-Str. 9-11, 55543 Bad Kreuznach, Germany
- Department of Internal Medicine I, Division of Rheumatology and Clinical Immunology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany
| | - Leif-Erik Thiele
- Rheumatology Center Rhineland-Palatinate, Kaiser-Wilhelm-Str. 9-11, 55543 Bad Kreuznach, Germany
| | - Lorenzo Cavagna
- Department of Rheumatology, University and IRCCS Policlinico S. Matteo Foundation Pavia, 27100 Pavia, Italy
| | - Xenofon Baraliakos
- Rheumazentrum Ruhrgebiet Herne, Ruhr-University Bochum, 44649 Herne, Germany
| | - George Bertsias
- Department of Internal Medicine and Rheumatology, School of Medicine, University of Crete, 71500 Heraklion, Greece
| | - Andreas Schwarting
- Rheumatology Center Rhineland-Palatinate, Kaiser-Wilhelm-Str. 9-11, 55543 Bad Kreuznach, Germany
- Department of Internal Medicine I, Division of Rheumatology and Clinical Immunology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany
| |
Collapse
|
27
|
Bergkamp SC, Wahadat MJ, Salah A, Kuijpers TW, Smith V, Tas SW, van den Berg JM, Kamphuis S, Schonenberg-Meinema D. Dysregulated endothelial cell markers in systemic lupus erythematosus: a systematic review and meta-analysis. J Inflamm (Lond) 2023; 20:18. [PMID: 37194071 DOI: 10.1186/s12950-023-00342-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 04/28/2023] [Indexed: 05/18/2023] Open
Abstract
OBJECTIVES To perform a systematic literature review and meta-analysis on endothelial cell (EC) markers that are involved and dysregulated in systemic lupus erythematosus (SLE) in relation to disease activity, as EC dysregulation plays a major role in the development of premature atherosclerosis in SLE. METHODS Search terms were entered into Embase, MEDLINE, Web of Science, Google Scholar and Cochrane. Inclusion criteria were 1) studies published after 2000 reporting measurements of EC markers in serum and/or plasma of SLE patients (diagnosed according to ACR/SLICC criteria), 2) English language peer reviewed articles, and 3) disease activity measurement. For meta-analysis calculations, the Meta-Essentials tool by Erasmus Research Institute and of Management (ERIM) was used. Only those EC markers, which were 1) reported in at least two articles and 2) reported a correlation coefficient (i.e. Spearman's rank or Pearson's) between the measured levels of the EC marker and disease activity were included. For meta-analyses, a fixed effect model was used. RESULTS From 2133 hits, 123 eligible articles were selected. The identified SLE-related endothelial markers were involved in EC activation, EC apoptosis, disturbed angiogenesis, defective vascular tone control, immune dysregulation and coagulopathy. Meta-analyses of primarily cross-sectional studies showed significant associations between marker levels and disease activity for the following endothelial markers: Pentraxin-3, Thrombomodulin, VEGF, VCAM-1, ICAM-1, IP-10 and MCP-1. Dysregulated EC markers without associations with disease activity were: Angiopoeitin-2, vWF, P-Selectin, TWEAK and E-Selectin. CONCLUSIONS We provide a complete literature overview for dysregulated EC markers in SLE comprising a wide range of different EC functions. SLE-induced EC marker dysregulation was seen with, but also without, association with disease activity. This study provides some clarity in the eminent complex field of EC markers as biomarkers for SLE. Longitudinal data on EC markers in SLE are now needed to guide us more in unravelling the pathophysiology of premature atherosclerosis and cardiovascular events in SLE patients.
Collapse
Affiliation(s)
- S C Bergkamp
- Department of Paediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Centres (AUMC), University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - M J Wahadat
- Department of Paediatric Rheumatology, Sophia Children's Hospital, Erasmus University Medical Centre, Rotterdam, The Netherlands
- Department of Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - A Salah
- Department of Paediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Centres (AUMC), University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - T W Kuijpers
- Department of Paediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Centres (AUMC), University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - V Smith
- Department of Internal Medicine, Ghent University, Ghent, Belgium
- Department of Rheumatology, Ghent University Hospital, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB Inflammation Research Centre (IRC), Ghent, Belgium
| | - S W Tas
- Department of Rheumatology and Clinical Immunology, and Laboratory for Experimental Immunology, Amsterdam Rheumatology and Immunology Centre, Amsterdam University Medical Centres (AUMC), University of Amsterdam, Amsterdam, The Netherlands
| | - J M van den Berg
- Department of Paediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Centres (AUMC), University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - S Kamphuis
- Department of Paediatric Rheumatology, Sophia Children's Hospital, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - D Schonenberg-Meinema
- Department of Paediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Centres (AUMC), University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
28
|
Cabana-Puig X, Lu R, Geng S, Michaelis JS, Oakes V, Armstrong C, Testerman JC, Liao X, Alajoleen R, Appiah M, Zhang Y, Reilly CM, Li L, Luo XM. CX 3CR1 modulates SLE-associated glomerulonephritis and cardiovascular disease in MRL/lpr mice. Inflamm Res 2023; 72:1083-1097. [PMID: 37060359 PMCID: PMC10748465 DOI: 10.1007/s00011-023-01731-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/31/2023] [Accepted: 04/05/2023] [Indexed: 04/16/2023] Open
Abstract
OBJECTIVE Patients with systemic lupus erythematosus (SLE) often develop multi-organ damages including heart and kidney complications. We sought to better define the underlying mechanisms with a focus on the chemokine receptor CX3CR1. METHODS We generated Cx3cr1-deficient MRL/lpr lupus-prone mice through backcrossing. We then employed heterozygous intercross to generate MRL/lpr littermates that were either sufficient or deficient of CX3CR1. The mice were also treated with either Lactobacillus spp. or a high-fat diet (HFD) followed by assessments of the kidney and heart, respectively. RESULTS Cx3cr1-/- MRL/lpr mice exhibited a distinct phenotype of exacerbated glomerulonephritis compared to Cx3cr1+/+ littermates, which was associated with a decrease of spleen tolerogenic marginal zone macrophages and an increase of double-negative T cells. Interestingly, upon correction of the gut microbiota with Lactobacillus administration, the phenotype of exacerbated glomerulonephritis was reversed, suggesting that CX3CR1 controls glomerulonephritis in MRL/lpr mice through a gut microbiota-dependent mechanism. Upon treatment with HFD, Cx3cr1-/- MRL/lpr mice developed significantly more atherosclerotic plaques that were promoted by Ly6C+ monocytes. Activated monocytes expressed ICOS-L that interacted with ICOS-expressing follicular T-helper cells, which in turn facilitated a germinal center reaction to produce more autoantibodies. Through a positive feedback mechanism, the increased circulatory autoantibodies further promoted the activation of Ly6C+ monocytes and their display of ICOS-L. CONCLUSIONS We uncovered novel, Cx3cr1 deficiency-mediated pathogenic mechanisms contributing to SLE-associated glomerulonephritis and cardiovascular disease.
Collapse
Affiliation(s)
- Xavier Cabana-Puig
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, USA
| | - Ran Lu
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Shuo Geng
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Jacquelyn S Michaelis
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, USA
| | - Vanessa Oakes
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, USA
| | - Caitlin Armstrong
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| | - James C Testerman
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, USA
| | - Xiaofeng Liao
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, USA
| | - Razan Alajoleen
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, USA
| | - Michael Appiah
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, USA
| | - Yao Zhang
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| | | | - Liwu Li
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA.
| | - Xin M Luo
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
29
|
Fasano S, Milone A, Nicoletti GF, Isenberg DA, Ciccia F. Precision medicine in systemic lupus erythematosus. Nat Rev Rheumatol 2023; 19:331-342. [PMID: 37041269 DOI: 10.1038/s41584-023-00948-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2023] [Indexed: 04/13/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that has diverse clinical manifestations, ranging from restricted cutaneous involvement to life-threatening systemic organ involvement. The heterogeneity of pathomechanisms that lead to SLE contributes to between-patient variation in clinical phenotype and treatment response. Ongoing efforts to dissect cellular and molecular heterogeneity in SLE could facilitate the future development of stratified treatment recommendations and precision medicine, which is a considerable challenge for SLE. In particular, some genes involved in the clinical heterogeneity of SLE and some phenotype-related loci (STAT4, IRF5, PDGF genes, HAS2, ITGAM and SLC5A11) have an association with clinical features of the disease. An important part is also played by epigenetic varation (in DNA methylation, histone modifications and microRNAs) that influences gene expression and affects cell function without modifying the genome sequence. Immune profiling can help to identify an individual's specific response to a therapy and can potentially predict outcomes, using techniques such as flow cytometry, mass cytometry, transcriptomics, microarray analysis and single-cell RNA sequencing. Furthermore, the identification of novel serum and urinary biomarkers would enable the stratification of patients according to predictions of long-term outcomes and assessments of potential response to therapy.
Collapse
Affiliation(s)
- Serena Fasano
- Rheumatology Unit, Department of Precision Medicine, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy.
| | - Alessandra Milone
- Rheumatology Unit, Department of Precision Medicine, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy
| | - Giovanni Francesco Nicoletti
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy
| | - David A Isenberg
- Department of Rheumatology, Division of Medicine, University College London, London, UK
| | - Francesco Ciccia
- Rheumatology Unit, Department of Precision Medicine, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy.
| |
Collapse
|
30
|
Bello N, Meyers KJ, Workman J, Hartley L, McMahon M. Cardiovascular events and risk in patients with systemic lupus erythematosus: Systematic literature review and meta-analysis. Lupus 2023; 32:325-341. [PMID: 36547368 PMCID: PMC10012401 DOI: 10.1177/09612033221147471] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is an autoimmune disease that typically affects women aged 16-55 years. Cardiovascular disease (CVD) is a well-recognized complication of SLE. This systematic literature review and meta-analysis evaluated the relative risk (RR; compared with non-SLE controls), absolute risk (AR; as incidence proportion, n/N), and incidence rate (IR) of CVD events (including stroke, myocardial infarction [MI], and CVD [composite or undefined]) in adult patients with SLE. The RR of CV risk factors (including hypertension, diabetes, and metabolic syndrome [MetS]) was also examined. METHODS PubMed and Embase were searched on September 10, 2020. Observational studies published between January 2010 and September 2020 that reported RR, AR, and/or IR of CVD events, or RR of CV risk factors, were eligible. Pooled risk estimates were calculated using a random-effects model. RESULTS Forty-six studies (16 cross-sectional, 15 retrospective cohort, 14 prospective cohort, and 1 case-control) were included in meta-analyses. Most studies were considered high quality (Critical Appraisal Skills Programme checklists). Compared with adults without SLE, patients with SLE had statistically significantly higher RRs (95% CIs) of stroke (2.51 [2.03-3.10]; 12 studies), MI (2.92 [2.45-3.48]; 11 studies), CVD (2.24 [1.94-2.59]; 8 studies), and hypertension (2.70 [1.48-4.92]; 7 studies). RRs of diabetes (1.24 [0.78-1.96]; 3 studies) and MetS (1.49 [0.95-2.33]; 7 studies) were elevated but not significant. RRs of stroke and MI were generally higher in younger versus older patients with SLE. In patients with SLE, the pooled estimate of AR (95% CI) was 0.03 (0.02-0.05), 0.01 (0.00-0.02), and 0.06 (0.03-0.10) for stroke (7 studies), MI (6 studies), and CVD (8 studies), respectively. The pooled estimate of IR per 1000 person-years (95% CI) was 4.72 (3.35-6.32), 2.81 (1.61-4.32), and 11.21 (8.48-14.32) for stroke (10 studies), MI (6 studies), and CVD (8 studies), respectively. Although heterogeneity (based on I2 value) was high in most analyses, sensitivity analyses confirmed the robustness of the pooled estimates. CONCLUSIONS This meta-analysis found an increased risk of stroke, MI, CVD, and hypertension in patients with SLE compared with the general population, despite substantial heterogeneity across the included studies.
Collapse
Affiliation(s)
| | | | | | | | - Maureen McMahon
- Division of Rheumatology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
31
|
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
|
32
|
Kridin K, Vorobyev A, Papara C, De Luca DA, Bieber K, Ludwig RJ. Risk factors and sequelae of epidermolysis bullosa acquisita: A propensity-matched global study in 1,344 patients. Front Immunol 2023; 13:1103533. [PMID: 36776391 PMCID: PMC9910332 DOI: 10.3389/fimmu.2022.1103533] [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: 11/20/2022] [Accepted: 12/28/2022] [Indexed: 01/27/2023] Open
Abstract
Identification of risk factors and sequelae of any given disease is of key importance. For common diseases, primary prevention and disease management are based on this knowledge. For orphan diseases, identification of risk factors and sequelae has been challenging. With the advent of large databases, e.g., TriNetX, this can now be addressed. We used TriNetX to identify risk factors and sequelae of epidermolysis bullosa acquisita (EBA), a severe and orphan autoimmune disease. To date, there is only enigmatic information on EBA comorbidity. We recruited 1,344 EBA patients in the Global Collaborative Network of TriNetX. Using the "explore outcomes" function we identified 55 diagnoses with a different prevalence between EBA and no-EBA patients. We next performed propensity-matched, retrospective cohort studies in which we determined the risk of EBA development following any of the identified 55 diseases. Here, 31/55 diseases were identified as risk factors for subsequent EBA. Importantly, the highest risk for EBA were other chronic inflammatory diseases (CID), especially lupus erythematosus and lichen planus. Lastly, we determined the risk to develop any of the identified diseases after EBA diagnosis. Here, 38/55 diseases were identified as sequelae. Notably, EBA patients showed an increased risk for metabolic and cardiovascular disease, and thrombosis. Furthermore, the risk for CIDs, especially lupus erythematosus and lichen planus, was elevated. These insights into risk factors and sequelae of EBA are not only of clinical relevance, e.g., optimizing cardiovascular disease risk, but in addition, point to shared pathogenetic pathways between EBA and other inflammatory diseases.
Collapse
Affiliation(s)
- Khalaf Kridin
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany,Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel,Unit of Dermatology and Skin Research Laboratory, Barch Padeh Medical Center, Tiberias, Israel
| | - Artem Vorobyev
- Department of Dermatology, University Hospital Schleswig-Holstein Lübeck, Lübeck, Germany
| | - Cristian Papara
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - David A. De Luca
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Katja Bieber
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Ralf J. Ludwig
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany,Department of Dermatology, University Hospital Schleswig-Holstein Lübeck, Lübeck, Germany,*Correspondence: Ralf J. Ludwig,
| |
Collapse
|
33
|
Mangoni AA, Sotgia S, Zinellu A, Carru C, Pintus G, Damiani G, Erre GL, Tommasi S. Methotrexate and cardiovascular prevention: an appraisal of the current evidence. Ther Adv Cardiovasc Dis 2023; 17:17539447231215213. [PMID: 38115784 PMCID: PMC10732001 DOI: 10.1177/17539447231215213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/02/2023] [Indexed: 12/21/2023] Open
Abstract
New evidence continues to accumulate regarding a significant association between excessive inflammation and dysregulated immunity (local and systemic) and the risk of cardiovascular events in different patient cohorts. Whilst research has sought to identify novel atheroprotective therapies targeting inflammation and immunity, several marketed drugs for rheumatological conditions may serve a similar purpose. One such drug, methotrexate, has been used since 1948 for treating cancer and, more recently, for a wide range of dysimmune conditions. Over the last 30 years, epidemiological and experimental studies have shown that methotrexate is independently associated with a reduced risk of cardiovascular disease, particularly in rheumatological patients, and exerts several beneficial effects on vascular homeostasis and blood pressure control. This review article discusses the current challenges with managing cardiovascular risk and the new frontiers offered by drug discovery and drug repurposing targeting inflammation and immunity with a focus on methotrexate. Specifically, the article critically appraises the results of observational, cross-sectional and intervention studies investigating the effects of methotrexate on overall cardiovascular risk and individual risk factors. It also discusses the putative molecular mechanisms underpinning the atheroprotective effects of methotrexate and the practical advantages of using methotrexate in cardiovascular prevention, and highlights future research directions in this area.
Collapse
Affiliation(s)
- Arduino A. Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
- Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Bedford Park, SA 5042, Australia
| | - Salvatore Sotgia
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy; Quality Control Unit, University Hospital (AOUSS), Sassari, Italy
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy; Quality Control Unit, University Hospital (AOUSS), Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Quality Control Unit, University Hospital (AOUSS), Sassari, Italy
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy; Quality Control Unit, University Hospital (AOUSS), Sassari, Italy
| | - Giovanni Damiani
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
- Italian Centre of Precision Medicine and Chronic Inflammation, Milan, Italy
| | - Gian Luca Erre
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University Hospital (AOUSS) and University of Sassari, Sassari, Italy
| | - Sara Tommasi
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, SA, Australia
| |
Collapse
|
34
|
Lee C, Drobni ZD, Zafar A, Gongora CA, Zlotoff DA, Alvi RM, Taron J, Rambarat PK, Schoenfeld S, Mosarla RC, Raghu VK, Hartmann SE, Gilman HK, Murphy SP, Sullivan RJ, Faje A, Hoffmann U, Zhang L, Mayrhofer T, Reynolds KL, Neilan TG. Pre-Existing Autoimmune Disease Increases the Risk of Cardiovascular and Noncardiovascular Events After Immunotherapy. JACC CardioOncol 2022; 4:660-669. [PMID: 36636443 PMCID: PMC9830202 DOI: 10.1016/j.jaccao.2022.11.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 11/03/2022] [Accepted: 11/08/2022] [Indexed: 12/24/2022] Open
Abstract
Background The use of immune checkpoint inhibitors (ICI) is associated with cardiovascular (CV) events, and patients with pre-existing autoimmune disease are at increased CV risk. Objectives The aim of this study was to characterize the risk for CV events in patients with pre-existing autoimmune disease post-ICI. Methods This was a retrospective study of 6,683 patients treated with ICIs within an academic network. Autoimmune disease prior to ICI was confirmed by chart review. Baseline characteristics and risk for CV and non-CV immune-related adverse events were compared with a matched control group (1:1 ratio) of ICI patients without autoimmune disease. Matching was based on age, sex, history of coronary artery disease, history of heart failure, and diabetes mellitus. CV events were a composite of myocardial infarction, percutaneous coronary intervention, coronary artery bypass graft, stroke, transient ischemic attack, deep venous thrombosis, pulmonary embolism, or myocarditis. Univariable and multivariable Cox proportional hazards models were used to determine the association between autoimmune disease and CV events. Results Among 502 patients treated with ICIs, 251 patients with and 251 patients without autoimmune disease were studied. During a median follow-up period of 205 days, there were 45 CV events among patients with autoimmune disease and 22 CV events among control subjects (adjusted HR: 1.77; 95% CI: 1.04-3.03; P = 0.0364). Of the non-CV immune-related adverse events, there were increased rates of psoriasis (11.2% vs 0.4%; P < 0.001) and colitis (24.3% vs 16.7%; P = 0.045) in patients with autoimmune disease. Conclusions Patients with autoimmune disease have an increased risk for CV and non-CV events post-ICI.
Collapse
Key Words
- CABG, coronary artery bypass graft
- CTLA-4, cytotoxic T lymphocyte–associated antigen-4
- CV, cardiovascular
- DVT, deep venous thrombosis
- ICI, immune checkpoint inhibitor
- MI, myocardial infarction
- PCI, percutaneous coronary intervention
- PD-1, programmed death-1
- PD-L1, programmed death-ligand 1
- PE, pulmonary embolism
- SMD, standardized mean difference
- TIA, transient ischemic attack
- coronary artery disease
- immunotherapy
- irAE, immune-related adverse event
- myocarditis
- thrombosis
Collapse
Affiliation(s)
- Charlotte Lee
- Division of Cardiology, Columbia University Irving Medical Center, NewYork-Presbyterian Hospital, New York, New York, USA
| | - Zsofia D. Drobni
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Amna Zafar
- Division of Cardiovascular Diseases and Hypertension, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Carlos A. Gongora
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Daniel A. Zlotoff
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Raza M. Alvi
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jana Taron
- Department of Radiology, University Hospital Freiburg, Freiburg, Germany
| | - Paula K. Rambarat
- Department of Internal Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sara Schoenfeld
- Division of Rheumatology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ramya C. Mosarla
- Division of Cardiology, New York University, New York, New York, USA
| | - Vineet K. Raghu
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sarah E. Hartmann
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Hannah K. Gilman
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sean P. Murphy
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ryan J. Sullivan
- Division of Oncology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alexander Faje
- Division of Endocrinology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Udo Hoffmann
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Lili Zhang
- Department of Cardiology, Department of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Thomas Mayrhofer
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kerry L. Reynolds
- Division of Oncology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Tomas G. Neilan
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
35
|
Hernández-Negrín H, Ricci M, Mancebo-Sevilla JJ, Sanz-Cánovas J, López-Sampalo A, Cobos-Palacios L, Romero-Gómez C, Pérez de Pedro I, Ayala-Gutiérrez MDM, Gómez-Huelgas R, Bernal-López MR. Obesity, Diabetes, and Cardiovascular Risk Burden in Systemic Lupus Erythematosus: Current Approaches and Knowledge Gaps-A Rapid Scoping Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:14768. [PMID: 36429489 PMCID: PMC9690813 DOI: 10.3390/ijerph192214768] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 06/16/2023]
Abstract
Obesity, diabetes mellitus, and cardiovascular risk are real challenges in systemic lupus erythematosus (SLE) clinical practice and research. The evidence of the burden of these health problems in SLE patients is determined by the methods used to assess them. Therefore, the aim of this scoping review is to map current approaches in assessing obesity, diabetes mellitus, and cardiovascular risk burden in SLE patients and to identify existing knowledge gaps in this field. This rapid scoping review was conducted according to the Joanna Briggs Institute methodology and identified 274 articles, of which 73 were included. Most studies were conducted at European institutions and patients were recruited from specialist hospital clinics, the majority of whom were women. The burden of obesity and diabetes mellitus for SLE patients was assessed mainly in terms of prevalence, impact on disease activity, and cardiometabolic risk. The burden of cardiovascular risk was assessed using multiple approaches, mainly imaging and laboratory methods, and risk factor-based scores, although there is great heterogeneity and uncertainty between the methods used. This review highlights the importance of improving and standardizing the approach to obesity, diabetes, and cardiovascular risk in SLE patients through a holistic assessment that includes lifestyle, clinical, biological, and social aspects.
Collapse
Affiliation(s)
- Halbert Hernández-Negrín
- Internal Medicine Clinical Management Unit, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Avenida Carlos Haya S/N, 29010 Malaga, Spain
- Faculty of Medicine, Universidad de Málaga, Campus Teatinos, 29010 Malaga, Spain
| | - Michele Ricci
- Internal Medicine Clinical Management Unit, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Avenida Carlos Haya S/N, 29010 Malaga, Spain
- Faculty of Medicine, Universidad de Málaga, Campus Teatinos, 29010 Malaga, Spain
| | - Juan José Mancebo-Sevilla
- Internal Medicine Clinical Management Unit, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Avenida Carlos Haya S/N, 29010 Malaga, Spain
- Faculty of Medicine, Universidad de Málaga, Campus Teatinos, 29010 Malaga, Spain
| | - Jaime Sanz-Cánovas
- Internal Medicine Clinical Management Unit, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Avenida Carlos Haya S/N, 29010 Malaga, Spain
- Faculty of Medicine, Universidad de Málaga, Campus Teatinos, 29010 Malaga, Spain
| | - Almudena López-Sampalo
- Internal Medicine Clinical Management Unit, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Avenida Carlos Haya S/N, 29010 Malaga, Spain
- Faculty of Medicine, Universidad de Málaga, Campus Teatinos, 29010 Malaga, Spain
| | - Lidia Cobos-Palacios
- Internal Medicine Clinical Management Unit, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Avenida Carlos Haya S/N, 29010 Malaga, Spain
- Faculty of Medicine, Universidad de Málaga, Campus Teatinos, 29010 Malaga, Spain
| | - Carlos Romero-Gómez
- Internal Medicine Clinical Management Unit, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Avenida Carlos Haya S/N, 29010 Malaga, Spain
| | - Iván Pérez de Pedro
- Internal Medicine Clinical Management Unit, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Avenida Carlos Haya S/N, 29010 Malaga, Spain
| | - María del Mar Ayala-Gutiérrez
- Internal Medicine Clinical Management Unit, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Avenida Carlos Haya S/N, 29010 Malaga, Spain
| | - Ricardo Gómez-Huelgas
- Internal Medicine Clinical Management Unit, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Avenida Carlos Haya S/N, 29010 Malaga, Spain
- Faculty of Medicine, Universidad de Málaga, Campus Teatinos, 29010 Malaga, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María Rosa Bernal-López
- Internal Medicine Clinical Management Unit, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Avenida Carlos Haya S/N, 29010 Malaga, Spain
- Faculty of Medicine, Universidad de Málaga, Campus Teatinos, 29010 Malaga, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
36
|
Omair MA, Alkhelb SA, Ezzat SE, Boudal AM, Bedaiwi MK, Almaghlouth I. Venous Thromboembolism in Rheumatoid Arthritis: The Added Effect of Disease Activity to Traditional Risk Factors. Open Access Rheumatol 2022; 14:231-242. [PMID: 36276408 PMCID: PMC9586712 DOI: 10.2147/oarrr.s284757] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022] Open
Abstract
Many epidemiological studies have shown an increased risk of venous thromboembolism (VTE) in patients with rheumatoid arthritis (RA). RA and VTE share some background factors, such as increasing age, smoking, and obesity. At the same time, other VTE factors, such as knee replacement and oral contraceptive pills, occur commonly in RA patients. In addition, the chronic inflammatory state of RA might hypothetically lead to endothelial injury and a hypercoagulable state. Two critical pathophysiological pathways lead to VTE. Recently, concerns increased about the increased risk of VTE in patients using Janus Kinase inhibitors. This review aims at reviewing the risk of VTE in RA and the role of traditional risk factors and disease-related inflammation and develops a conceptual framework that describes the interaction between these factors.
Collapse
Affiliation(s)
- Mohammed A Omair
- Rheumatology Unit, Department of Medicine, King Saud University, Riyadh, Saudi Arabia,Correspondence: Mohammed A Omair, Rheumatology Unit, Department of Medicine, King Saud University, Riyadh, Saudi Arabia, Tel +966505270513, Email
| | - Sara A Alkhelb
- Rheumatology Unit, Department of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Sadeen E Ezzat
- Rheumatology Unit, Department of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Ayah M Boudal
- Rheumatology Unit, Department of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed K Bedaiwi
- Rheumatology Unit, Department of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Ibrahim Almaghlouth
- Rheumatology Unit, Department of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
37
|
Laura RP, Jonatan GC, Banwell G, Belén OÁA, Gabriel GN, Andrés RC. EXPERIENCES OF PATIENTS WITH RHEUMATOID ARTHRITIS DURING AND AFTER COVID-19 INDUCED QUARANTINE IN TERMS OF PHYSICAL ACTIVITY AND HEALTH STATUS: A QUALITATIVE STUDY. J Nurs Manag 2022; 30:2568-2576. [PMID: 36057281 PMCID: PMC9538776 DOI: 10.1111/jonm.13784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/26/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022]
Abstract
Aim The aim of this study was to explore experiences of people with rheumatoid arthritis during and after COVID‐19‐induced quarantine in terms of physical activity and health status. Background Rheumatoid arthritis affects multiple facets of the person, both physically and psychologically. Physical activity is considered a safe and effective intervention to improve symptoms and systemic manifestations of rheumatoid arthritis. In the context of the COVID‐19, countries like Spain were forced to impose restrictions on mobility, prohibiting outings even to perform physical activity. Methods Structured interviews were conducted and developed using the Tampa Scale for Kinesiophobia‐11 questionnaire. Data were analysed using a six‐step thematic analysis. Results The results make it clear that even though the patients declared that physical activity is essential for them to deal with their disease, most of the participants affirmed that they significatively reduced their levels of physical activity during the pandemic. Conclusions Physical activity should be promoted in people, even in difficult times, to improve disease outcomes, well‐being and mental health. Implications for Nursing Management Knowing the experiences of these patients enables nursing managers to develop interventions that ensure the delivery of comprehensive nursing care regarding physical activity and health status, in future situations like this pandemic.
Collapse
Affiliation(s)
- Ramos-Petersen Laura
- Department of Podiatry. Faculty of Health Sciences, Universidad Católica San Antonio de Murcia. Campus de Los Jerónimos. Guadalupe, Murcia, Spain
| | - García-Campos Jonatan
- Department of Behavioral Sciences and Health, Faculty of Medicine, Miguel Hernandez University, Alicante, Spain. Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - George Banwell
- Department of Nursing and Podiatry, Faculty of Health Sciences, University of Malaga, Malaga, Spain
| | - Ortega Ávila Ana Belén
- Instituto de Investigación Biomédica de Málaga (IBIMA), Department of Nursing and Podiatry, Faculty of Health Sciences, University of Malaga, Malaga, Spain
| | - Gijon-Nogueron Gabriel
- Instituto de Investigación Biomédica de Málaga (IBIMA), Department of Nursing and Podiatry, Faculty of Health Sciences, University of Malaga, Malaga, Spain
| | - Reinoso-Cobo Andrés
- Instituto de Investigación Biomédica de Málaga (IBIMA), Department of Nursing and Podiatry, Faculty of Health Sciences, University of Malaga, Malaga, Spain
| |
Collapse
|
38
|
Huang S, Huang F, Mei C, Tian F, Fan Y, Bao J. Systemic lupus erythematosus and the risk of cardiovascular diseases: A two-sample Mendelian randomization study. Front Cardiovasc Med 2022; 9:896499. [PMID: 36119739 PMCID: PMC9478435 DOI: 10.3389/fcvm.2022.896499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background Previous observational studies have suggested that the causal role of systemic lupus erythematosus (SLE) in the risk of cardiovascular diseases (CVDs) remained inconsistent. In this study, we aimed to investigate the causal relationship between SLE and CVDs by two-sample Mendelian randomization (MR) analysis. Methods Genetic instruments for SLE were obtained from a public genome-wide association study (GWAS) with 4,036 patients with SLE and 6,959 controls. Summary statistical data for CVDs, including coronary artery disease (CAD), myocardial infarction (MI), atrial fibrillation (AF), ischemic stroke (IS), and its subtypes, were identified from other available GWAS meta-analyses. The inverse-variance weighted (IVW) method was used as the primary method to estimate the causal effect. The simple- and weighted-median method, MR-Egger method, and MR pleiotropy residual sum and outlier (MR-PRESSO) were provided as a supplement to the IVW method. Besides, we performed sensitivity analyses, including Cochran's Q test, MR-Egger intercept test, and leave-one-out analysis, to evaluate the robustness of the results. Results A total of 15 single-nucleotide polymorphisms (SNPs) were identified after excluding linkage disequilibrium (LD) and potential confounding factors. According to the IVW results, our MR study indicated that genetically predicted SLE was not causally connected with the risk of CVDs [CAD: odds ratio (OR) = 1.005, 95% confidence interval (CI) = 0.986–1.024, p-value = 0.619; MI: OR = 1.002, 95% CI = 0.982–1.023, p-value = 0.854; AF: OR = 0.998, 95% CI = 0.982–1.014, p-value = 0.795; IS: OR = 1.006, 95% CI = 0.984–1.028, p-value = 0.621; cardioembolic stroke (CES): OR = 0.992, 95% CI = 0.949–1.036, p-value = 0.707; small vessel stroke (SVS): OR = 1.014, 95% CI = 0.964–1.067, p-value = 0.589; large artery stroke (LAS): OR = 1.030, 95% CI = 0.968–1.096, p-value = 0.352]. Analogical findings could be observed in supplementary MR methods. Sensitivity analyses suggested that the causal estimates were robust. Conclusion Our two-sample MR analysis provided no evidence that genetically determined SLE was causally associated with the risk of CVDs.
Collapse
Affiliation(s)
- Shuo Huang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fugang Huang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chunyun Mei
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fengyuan Tian
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yongsheng Fan
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Yongsheng Fan
| | - Jie Bao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Jie Bao
| |
Collapse
|
39
|
Samões B, Zen M, Abelha-Aleixo J, Gatto M, Doria A. Caveats and pitfalls in defining low disease activity in systemic lupus erythematosus. Autoimmun Rev 2022; 21:103165. [PMID: 35931316 DOI: 10.1016/j.autrev.2022.103165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 07/31/2022] [Indexed: 11/02/2022]
Abstract
The treat-to-target strategy has been recently suggested in the management of Systemic Lupus Erythematosus (SLE). Lupus Low Disease Activity State (LLDAS) and Definitions Of Remission In SLE (DORIS) remission were outlined as two concentric targets. The achievement of LLDAS was shown to be associated with lower frequency of SLE flare, decreased damage progression, better quality of life, and reduced mortality. In addition, LLDAS has successfully been tested in post-hoc analyses of a number of randomized controlled trials. However, it has been recently underlined that LLDAS includes a high proportion of patients in remission, raising the question if these endpoints are sufficiently distinct to consider their separation clinically relevant. Some studies suggest that the protective effect of LLDAS on damage might be due to the inclusion of patients who are in remission. Notably, clinical low disease activity (LDA) seems to be uncommon in SLE due to the relapsing-remitting pattern of the disease, in which low level of activity only occurs transiently. Moreover, since the domains included in LLDAS have several limitations, such as the use of a binomial disease activity index, the exclusion of some mild manifestations and the consideration of items subjected to variability (physician global assessment and glucocorticoids dose), not all patients in LDA are adequately represented by LLDAS.
Collapse
Affiliation(s)
- Beatriz Samões
- Rheumatology Department, Centro Hospitalar de Vila Nova de Gaia e Espinho, Rua Conceição Fernandes, s/n, 4434-502 Vila Nova de Gaia, Portugal.
| | - Margherita Zen
- Division of Rheumatology, Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy.
| | - Joana Abelha-Aleixo
- Rheumatology Department, Centro Hospitalar de Vila Nova de Gaia e Espinho, Rua Conceição Fernandes, s/n, 4434-502 Vila Nova de Gaia, Portugal.
| | - Mariele Gatto
- Division of Rheumatology, Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy.
| | - Andrea Doria
- Division of Rheumatology, Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy.
| |
Collapse
|
40
|
Colaci M, Zanoli L, Lo Gullo A, Sambataro D, Sambataro G, Aprile ML, Castellino P, Malatino L. The Impaired Elasticity of Large Arteries in Systemic Sclerosis Patients. J Clin Med 2022; 11:jcm11123256. [PMID: 35743327 PMCID: PMC9224949 DOI: 10.3390/jcm11123256] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 11/30/2022] Open
Abstract
(1) Background: Systemic sclerosis (SSc) is an autoimmune disease characterized by endothelial dysfunction and fibrosis of skin and visceral organs. In the last decade, attention has been focused on the macrovascular involvement of the disease. In particular, the observation of increased arterial stiffness represented an interesting aspect of the disease, as predictor of cardiovascular risk. (2) Methods: We recruited 60 SSc patients (52 ± 12 years old, 90% females) and 150 age/sex-matched healthy controls in order to evaluate both intima-media thickness of the right common carotid artery and arterial stiffness using the B-mode echography and the SphygmoCor system® tonometer. (3) Results: The carotid-femoral pulse wave velocity (PWV) was higher in SSc patients than in controls (8.6 ± 1.7 vs. 7.8 ± 1.5 m/s; p < 0.001), as was the carotid-radial PWV (7.8 ± 1.1 vs. 6.7 ± 1.4 m/s; p < 0.001). The intima-media thickness was higher in SSc than in controls (654 ± 108 vs. 602 ± 118 µm; p = 0.004). The other parameters measured at carotid (radial strain, Young’s modulus, compliance and distensibility) all indicated that arterial stiffness in tension was more pronounced in SSc. Of interest, the direct correlation between PWV and age corresponded closely in SSc. Moreover, a significant difference between SSc and controls as regards the carotid parameters was evident in younger subjects. (4) Conclusions: SSc patients showed an increased arterial stiffness compared to healthy controls. In particular, an SSc-related pathologic effect was suggested by the more pronounced increase in PWV with age and lower values of carotid elasticity in younger SSc patients than in age-matched controls.
Collapse
Affiliation(s)
- Michele Colaci
- Rheumatology Clinic, Internal Medicine Unit, AOE Cannizzaro, 95126 Catania, Italy; (M.L.A.); (L.M.)
- Department of Clinical and Experimental Medicine, University of Catania, 95100 Catania, Italy; (L.Z.); (D.S.); (G.S.); (P.C.)
- Correspondence:
| | - Luca Zanoli
- Department of Clinical and Experimental Medicine, University of Catania, 95100 Catania, Italy; (L.Z.); (D.S.); (G.S.); (P.C.)
- Internal Medicine Unit, Policlinico Rodolico—S. Marco, 95123 Catania, Italy
| | | | - Domenico Sambataro
- Department of Clinical and Experimental Medicine, University of Catania, 95100 Catania, Italy; (L.Z.); (D.S.); (G.S.); (P.C.)
| | - Gianluca Sambataro
- Department of Clinical and Experimental Medicine, University of Catania, 95100 Catania, Italy; (L.Z.); (D.S.); (G.S.); (P.C.)
| | - Maria Letizia Aprile
- Rheumatology Clinic, Internal Medicine Unit, AOE Cannizzaro, 95126 Catania, Italy; (M.L.A.); (L.M.)
| | - Pietro Castellino
- Department of Clinical and Experimental Medicine, University of Catania, 95100 Catania, Italy; (L.Z.); (D.S.); (G.S.); (P.C.)
- Internal Medicine Unit, Policlinico Rodolico—S. Marco, 95123 Catania, Italy
| | - Lorenzo Malatino
- Rheumatology Clinic, Internal Medicine Unit, AOE Cannizzaro, 95126 Catania, Italy; (M.L.A.); (L.M.)
- Department of Clinical and Experimental Medicine, University of Catania, 95100 Catania, Italy; (L.Z.); (D.S.); (G.S.); (P.C.)
| |
Collapse
|
41
|
Sahin S, Romano M, Guzel F, Piskin D, Poddighe D, Sezer S, Kasapcopur O, Appleton CT, Yilmaz I, Demirkaya E. Assessment of Surrogate Markers for Cardiovascular Disease in Familial Mediterranean Fever-Related Amyloidosis Patients Homozygous for M694V Mutation in MEFV Gene. Life (Basel) 2022; 12:life12050631. [PMID: 35629299 PMCID: PMC9146909 DOI: 10.3390/life12050631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/16/2022] [Accepted: 04/21/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular disease (CVD) remains underestimated in familial Mediterranean fever-associated AA amyloidosis (FMF-AA). We aimed to compare early markers of endothelial dysfunction and atherosclerosis in FMF-AA with a homozygous M694V mutation (Group 1 = 76 patients) in the Mediterranean fever (MEFV) gene and in patients with other genotypes (Group 2 = 93 patients). Measures of increased risk for future CVD events and endothelial dysfunction, including flow-mediated dilatation (FMD), pentraxin-3 (PTX3), and carotid intima-media thickness (cIMT), and fibroblast growth factor 23 (FGF23) as a marker of atherosclerotic vascular disease were compared between groups. The frequency of clinical FMF manifestations did not differ between the two groups apart from arthritis (76.3% in Group 1 and 59.1% in Group 2, p < 0.05). FMD was significantly lower in Group 1 when compared with Group 2 (MD [95% CI]: −0.6 [(−0.89)−(−0.31)]). cIMT, FGF23, and PTX3 levels were higher in Group 1 (cIMT MD [95% CI]: 0.12 [0.08−0.16]; FGF23 MD [95% CI]: 12.8 [5.9−19.6]; PTX3 MD [95% CI]: 13.3 [8.9−17.5]). In patients with FMF-AA, M694V homozygosity is associated with lower FMD values and higher cIMT, FGF23, and PTX3 levels, suggesting increased CVD risk profiles. These data suggest that a genotype−phenotype association exists in terms of endothelial dysfunction and atherosclerosis in patients with FMF-AA.
Collapse
Affiliation(s)
- Sezgin Sahin
- Department of Paediatric Rheumatology, Istanbul University-Cerrahpasa, Istanbul 34098, Turkey;
- Correspondence:
| | - Micol Romano
- Division of Paediatric Rheumatology, Department of Paediatrics, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (M.R.); (E.D.)
- Canadian Behcet and Autoinflammatory Disease Center (CAN-BE-AID), University of Western Ontario, London, ON N6A 5C1, Canada;
| | - Ferhat Guzel
- Molecular Genetics Laboratories, Genetics Research and Genome Center, Department of Research and Development, Ant Biotechnology, Istanbul 34775, Turkey;
| | - David Piskin
- Lawson Health Research Institute, London Health Sciences Center, London, ON N6C 2R5, Canada;
- Department of Epidemiology and Biostatistics, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Dimitri Poddighe
- Department of Medicine, Nazarbayev University School of Medicine, Nur-Sultan 010000, Kazakhstan;
- Clinical Academic Department of Pediatrics, National Research Center of Maternal and Child Health, University Medical Center, Nur-Sultan 010000, Kazakhstan
| | - Siren Sezer
- Division of Nephrology, Atilim University Faculty of Medicine, Ankara 06830, Turkey;
| | - Ozgur Kasapcopur
- Department of Paediatric Rheumatology, Istanbul University-Cerrahpasa, Istanbul 34098, Turkey;
| | - C. Thomas Appleton
- Canadian Behcet and Autoinflammatory Disease Center (CAN-BE-AID), University of Western Ontario, London, ON N6A 5C1, Canada;
- Lawson Health Research Institute, London Health Sciences Center, London, ON N6C 2R5, Canada;
- Division of Rheumatology, Department of Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Ilker Yilmaz
- Epigenetic Health Solutions, Unit of Nephrology, Ankara 06810, Turkey;
| | - Erkan Demirkaya
- Division of Paediatric Rheumatology, Department of Paediatrics, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (M.R.); (E.D.)
- Canadian Behcet and Autoinflammatory Disease Center (CAN-BE-AID), University of Western Ontario, London, ON N6A 5C1, Canada;
- Lawson Health Research Institute, London Health Sciences Center, London, ON N6C 2R5, Canada;
| |
Collapse
|
42
|
Bellocchi C, Carandina A, Montinaro B, Targetti E, Furlan L, Rodrigues GD, Tobaldini E, Montano N. The Interplay between Autonomic Nervous System and Inflammation across Systemic Autoimmune Diseases. Int J Mol Sci 2022; 23:ijms23052449. [PMID: 35269591 PMCID: PMC8910153 DOI: 10.3390/ijms23052449] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 12/13/2022] Open
Abstract
The autonomic nervous system (ANS) and the immune system are deeply interrelated. The ANS regulates both innate and adaptive immunity through the sympathetic and parasympathetic branches, and an imbalance in this system can determine an altered inflammatory response as typically observed in chronic conditions such as systemic autoimmune diseases. Rheumatoid arthritis, systemic lupus erythematosus, and systemic sclerosis all show a dysfunction of the ANS that is mutually related to the increase in inflammation and cardiovascular risk. Moreover, an interaction between ANS and the gut microbiota has direct effects on inflammation homeostasis. Recently vagal stimulation techniques have emerged as an unprecedented possibility to reduce ANS dysfunction, especially in chronic diseases characterized by pain and a decreased quality of life as well as in chronic inflammation.
Collapse
Affiliation(s)
- Chiara Bellocchi
- Department of Internal Medicine, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.C.); (B.M.); (E.T.); (L.F.); (E.T.)
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy;
- Correspondence: (C.B.); (N.M.)
| | - Angelica Carandina
- Department of Internal Medicine, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.C.); (B.M.); (E.T.); (L.F.); (E.T.)
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy;
| | - Beatrice Montinaro
- Department of Internal Medicine, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.C.); (B.M.); (E.T.); (L.F.); (E.T.)
| | - Elena Targetti
- Department of Internal Medicine, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.C.); (B.M.); (E.T.); (L.F.); (E.T.)
| | - Ludovico Furlan
- Department of Internal Medicine, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.C.); (B.M.); (E.T.); (L.F.); (E.T.)
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy;
| | - Gabriel Dias Rodrigues
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy;
- Laboratory of Experimental and Applied Exercise Physiology, Department of Physiology and Pharmacology, Fluminense Federal University, Niterói 24210-130, Brazil
| | - Eleonora Tobaldini
- Department of Internal Medicine, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.C.); (B.M.); (E.T.); (L.F.); (E.T.)
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy;
| | - Nicola Montano
- Department of Internal Medicine, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.C.); (B.M.); (E.T.); (L.F.); (E.T.)
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy;
- Correspondence: (C.B.); (N.M.)
| |
Collapse
|