1
|
Xie Y, Cheng Q, Xu ML, Xue J, Wu H, Du Y. Itaconate: A Potential Therapeutic Strategy for Autoimmune Disease. Scand J Immunol 2025; 101:e70026. [PMID: 40289463 DOI: 10.1111/sji.70026] [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: 03/04/2025] [Revised: 03/04/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025]
Abstract
Itaconate is a metabolite of the Krebs cycle, and endogenous itaconate is driven by a variety of innate signals that inhibit the production of inflammatory cytokines. The key mechanism of action of itaconate was initially found to be the competitive inhibition of succinate dehydrogenase (SDH), which inhibits the production of inflammatory factors, as well as its antioxidant effects. With increasing research, it was discovered that it modifies cysteine residues of related proteins through the Michael addition, such as modifying the Kelch-like ECH-associated protein 1 (KEAP1) protein and activating the nuclear factor erythroid 2-related factor 2 (NRF2) signalling pathway, as well as glycolytic enzymes and cellular pathway-associated factors that attenuate inflammatory responses and oxidative stress. It also acts on a variety of immune cells, affecting their function and activity, and has been increasingly shown to play a therapeutic role in a variety of inflammatory and autoimmune diseases through a combination of these mechanisms. In conclusion, there has been a great breakthrough in the research of itaconate, from the initial industrial application to the redefinition of the biological functions of itaconate. However, with the deepening of the research, we also found that there are more questions: the mechanism of action of itaconate, more functions of itaconate, clinical application of itaconate, and the use of itaconate still needs to be solved.
Collapse
Affiliation(s)
- Yifan Xie
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Department of Clinic Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Qi Cheng
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Meng Li Xu
- Department of Nephrology, The Third Affiliate Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jing Xue
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Huaxiang Wu
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Du
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
2
|
Yang L, Guan X, Cheng J, Ni L, Yao H, Gao Y, Zhu K, Shi X, Li B, Lin Y. VAMP8 as a biomarker and potential therapeutic target for endothelial cell dysfunction in atherosclerosis. Gene 2025; 942:149231. [PMID: 39800194 DOI: 10.1016/j.gene.2025.149231] [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: 05/30/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Endothelial cell dysfunction has a critical role in the pathophysiology of atherosclerosis. This study aims to uncover pivotal genes and pathways linked to endothelial cell dysfunction in atherosclerosis, as well as to ascertain the assumed causal effects and potential mechanisms. METHODS Datasets relevant to endothelial cell dysfunction in atherosclerosis were collected and divided into training and validation sets. Following differential analysis, we constructed a protein-protein interaction (PPI) network and a molecular interaction map of common-differentially expressed genes (co-DEGs) with proteins known to be involved in atherosclerotic endothelial cell dysfunction. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genome (KEGG), and Gene Set Enrichment Analysis (GSEA) were also conducted. Moreover, human umbilical vein endothelial cells (HUVECs) were cultured in circumstances characterized by elevated glucose levels to establish a cellular injury model simulating atherosclerotic conditions, and quantitative Polymerase Chain Reaction (qPCR) experiments were conducted to validate the differences of co-DEGs. Subsequently, the Summary-data-based Mendelian Randomization (SMR) method was employed. Additionally, we employed the Western Blot (WB) technique to validate the differential expression of VAMP8. Finally, we identified the differential expression of VAMP8 in the validation set and further validated its differential expression by collecting fresh blood samples from 20 patients with atherosclerosis and 20 healthy individuals. RESULTS 14 co-DEGs (FABP5, GULP1, COL4A5, VAMP8, FABP4, PFN2, ANGPT2, TFPI2, NUPR1, SULF1, FGF13, BASP1, EPB41L3, and PBK) were identified. SMR analysis confirmed 10 potential causal effect genes: PSRC1, VAMP8, FES, HNRNPUL1, CFDP1, SAP130, MDN1, OPRL1, UTP11, and HOXC4. The qPCR and WB experiments demonstrated that VAMP8 was significantly upregulated in the injured HUVECs group (p < 0.0001). Compared to the control group, VAMP8 was markedly increased in the blood samples of patients with atherosclerosis (p < 0.0001). CONCLUSIONS VAMP8 may potentially serve as a pathogenic gene in the process of endothelial dysfunction in atherosclerosis.
Collapse
Affiliation(s)
- Luqun Yang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Xin Guan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Jiangwei Cheng
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Lin Ni
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Huijing Yao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yuping Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Kaiyi Zhu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Xiushan Shi
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Bingjie Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yuanyuan Lin
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| |
Collapse
|
3
|
Huang YC, Lai ECC, Liao TC, Weng MY. Evaluating the risk of ischemic stroke at a young age in patients with autoimmune inflammatory rheumatic diseases: a population-based cohort study in Taiwan. Front Immunol 2024; 15:1272557. [PMID: 38404587 PMCID: PMC10884215 DOI: 10.3389/fimmu.2024.1272557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/22/2024] [Indexed: 02/27/2024] Open
Abstract
Background Recent studies have demonstrated an increased incidence of ischemic stroke among patients with certain autoimmune inflammatory rheumatic diseases (AIIRDs). However, the associations between young stroke and AIIRDs have not been fully investigated. This study aimed to evaluate the risk of ischemic stroke among young patients with AIIRDs. Methods The National Health Insurance Research Database in Taiwan was utilized to establish cohorts of patients with AIIRDs diagnosed between 2004 and 2015, who were compared with 1,000,000 control participants. Cox proportional hazards regression models were used to calculate the hazard ratio of ischemic stroke and young ischemic stroke for individual AIIRDs after adjustment for relative risk factors. Results During the study period, a total of 64,120 patients with AIIRDss and 1,000,000 control patients were identified. The overall mean follow-up time was 5.33 years. There were 223 (0.8%) and 1,923 (0.3%) young ischemic stroke-related hospitalizations among patients with AIIRDs and controls, respectively. The incidence rate of young ischemic stroke was 0.08 in patients with rheumatoid arthritis, 0.08 in patients with Sjögren's syndrome, 0.26 in patients with systemic lupus erythematosus, 0.17 in patients with idiopathic inflammatory myositis, 0.24 in patients with systemic sclerosis, 0.05 in patients with Behçet's disease, and 0.44 in patients with systemic vasculitis, versus 0.05 per 100 person-years in the general population. The adjusted hazard ratios for young ischemic stroke were 1.07 (95% CI 0.70-1.43) for rheumatoid arthritis, 1.39 (95% CI 0.94-2.06) for Sjögren's syndrome, 5.79 (95% CI 4.68-7.17) for systemic lupus erythematosus, 2.07 for idiopathic inflammatory myositis (95% CI 0.98-4.38), 2.79 for systemic sclerosis (95% CI 1.38-5.63), 0.82 for Behçet's disease (95% CI 0.26-2.55), and 4.15 (95% CI 1.96-8.82) for systemic vasculitis. Conclusions Patients younger than 50 years with systemic lupus erythematosus, systemic sclerosis, or systemic vasculitis have a significantly elevated risk of developing ischemic stroke. Further research is needed to elucidate the pathogenesis of accelerated atherosclerosis in these AIIRDs.
Collapse
Affiliation(s)
- Ya-Chun Huang
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Edward Chia-Cheng Lai
- School of Pharmacy, Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Chi Liao
- School of Pharmacy, Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Meng-Yu Weng
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
4
|
Akuka A, Ben-Shabat N, Watad A, Tsur AM, Ehrenberg S, McGonagle D, Comaneshter D, Beinart R, Cohen AD, Amital H. Association of anti-Ro seropositivity with cardiac rhythm and conduction disturbances. Eur Heart J 2022; 43:4912-4919. [PMID: 36148481 DOI: 10.1093/eurheartj/ehac516] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 07/09/2022] [Accepted: 09/07/2022] [Indexed: 01/12/2023] Open
Abstract
AIMS Anti-Ro/La autoantibodies are especially prevalent in autoimmune diseases but are also relatively frequent in healthy adults. Their arrhythmogenic effect on the immature cardiac conductive system is well established, with substantial evidence demonstrating an increased risk for congenital atrioventricular block in neonates of seropositive mothers. Despite their wide distribution and their arrhythmogenic potential effect, there are no large population studies conducted in seropositive adults. Thus, this is the first large population-based study to examine the association of anti-Ro/La seropositivity with cardiac rhythm and conduction disturbances. METHODS AND RESULTS This cross-sectional designed study involved the electronic health records of the largest health maintenance organization in Israel. All subjects that were tested positive for anti-Ro/anti-La antibodies between the years 2002 and 2019 were included and were matched by age, gender, and place of residence, with controls. Rates of different cardiac rhythm and conduction disturbances were compared between groups. Sensitivity analyses were performed using propensity score matching. The study population included 17 231 anti-Ro/La seropositive subjects and 84 368 controls. Anti-Ro seropositive patients had higher rates of conduction disturbances (3.0 vs. 1.7%, P < 0.001) and rhythm disturbances (10.5 vs. 7.0%, P < 0.001). Patients who tested positive for anti-La alone did not demonstrate a significant association with arrhythmias. Multivariate logistic regression analysis, controlling for possible confounders, showed an increased risk for cardiac conduction disturbances [odds ratio (OR) 1.44, 95% confidence interval (CI) 1.25-1.66, P < 0.001], as well as for cardiac rhythm disturbances (OR 1.21, 95% CI 1.11-1.31, P < 0.001) among anti-Ro seropositive patients. However, the association with rhythm disturbances was more robust in certain subgroup analyses. CONCLUSIONS Anti-Ro seropositivity is positively associated with adult cardiac conduction disturbances and, to a lesser extent, cardiac rhythm disturbances, regardless of the presence of concurrent autoimmune disease.
Collapse
Affiliation(s)
- Aviram Akuka
- Department of Medicine 'B', Sheba Medical Center, Tel-Hashomer 5262100, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Ramat-Aviv 69978, Israel
| | - Niv Ben-Shabat
- Department of Medicine 'B', Sheba Medical Center, Tel-Hashomer 5262100, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Ramat-Aviv 69978, Israel
| | - Abdulla Watad
- Department of Medicine 'B', Sheba Medical Center, Tel-Hashomer 5262100, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Ramat-Aviv 69978, Israel.,Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds LS2 9JT, UK
| | - Avishai M Tsur
- Department of Medicine 'B', Sheba Medical Center, Tel-Hashomer 5262100, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Ramat-Aviv 69978, Israel.,Israel Defence Forces, Medical Corps, Tel Hashomer, Ramat Gan 52620, Israel.,Faculty of Medicine, Department of Military Medicine, Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Scott Ehrenberg
- Department of Medicine 'B', Sheba Medical Center, Tel-Hashomer 5262100, Israel
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds LS2 9JT, UK
| | - Doron Comaneshter
- Chief Physician's Office, Faculty of Health Sciences, Clalit Health Services Tel Aviv, Ben Gurion University of the Negev, Beer Sheva 8410501, Israel
| | - Roy Beinart
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Ramat-Aviv 69978, Israel.,Heart Institute, Chaim Sheba Medical Center, Tel Hashomer 52620, Israel.,Davidai Arrhythmia Center, Sheba Medical Center, Tel Hashomer 52620, Israel
| | - Arnon D Cohen
- Chief Physician's Office, Faculty of Health Sciences, Clalit Health Services Tel Aviv, Ben Gurion University of the Negev, Beer Sheva 8410501, Israel.,Siaal Research Center for Family Medicine and Primary Care, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva 8410501, Israel
| | - Howard Amital
- Department of Medicine 'B', Sheba Medical Center, Tel-Hashomer 5262100, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Ramat-Aviv 69978, Israel
| |
Collapse
|
5
|
Li Y, Liu Z. Case Report: A Case of Unusual Combination of Hypothyroidism, Myocardial Bridging, and Myocardial Infarction-Induced Left Ventricular Aneurysm. Front Cardiovasc Med 2021; 8:684616. [PMID: 34869621 PMCID: PMC8634395 DOI: 10.3389/fcvm.2021.684616] [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: 03/24/2021] [Accepted: 06/29/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Myocardial bridging (MB) of the coronary artery is a congenital anatomical variation, which has traditionally been considered a benign condition that does not cause cardiovascular events. However, recent studies have shown that MB is associated with major adverse cardiac events, including angina, myocardial infarction, arrhythmia, syncope, and even sudden death. Case: We report a case of a 41-year-old man who had hypothyroidism and MB associated with ventricular aneurysm following myocardial infarction. This patient was admitted to our hospital because of 11 days of sudden discomfort and pain in the chest. An electrocardiogram on admission showed an old myocardial infarction. Coronary angiography showed MB in the distal segment of the left anterior descending artery. Left ventricular angiography, which was performed using a pigtail catheter, showed ventricular aneurysm formation. Thyroid ultrasound demonstrated hypothyroidism and Hashimoto's thyroiditis. Patients with hypothyroidism and MB have a high risk of acute myocardial infarction or even sudden death. Conclusion: Observations in our case suggest that early recognition of hypothyroidism and MB is important for risk stratification and prognosis in patients with myocardial necrosis and acute coronary syndrome. Additionally, this early recognition may have positive effects on cardiovascular outcomes in patients with hypothyroidism.
Collapse
Affiliation(s)
- Yueliang Li
- Department of Cardiology, Qingyuan People's Hospital, the Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | - Zhengjiang Liu
- Department of Cardiology, Qingyuan People's Hospital, the Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| |
Collapse
|
6
|
Psoriasis and Atherosclerosis-Skin, Joints, and Cardiovascular Story of Two Plaques in Relation to the Treatment with Biologics. Int J Mol Sci 2021; 22:ijms221910402. [PMID: 34638740 PMCID: PMC8508744 DOI: 10.3390/ijms221910402] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/21/2021] [Accepted: 09/24/2021] [Indexed: 02/06/2023] Open
Abstract
It is known that both psoriasis (PSO) limited to the skin and psoriatic arthritis (PSA) increase the risk of cardiovascular complications and atherosclerosis progression by inducing systemic inflammatory response. In recent decades, the introduction of biological medications directed initially against TNF-α and, later, different targets in the inflammatory cascade brought a significant breakthrough in the efficacy of PSO/PSA treatment. In this review, we present and discuss the most recent findings related to the interplay between the genetics and immunology mechanisms involved in PSO and PSA, atherosclerosis and the development of cardiac dysfunction, as well as the current PSO/PSA treatment in view of cardiovascular safety and prognosis.
Collapse
|
7
|
Baviera M, Cioffi G, Colacioppo P, Tettamanti M, Fortino I, Roncaglioni MC. Temporal trends from 2005 to 2018 in deaths and cardiovascular events in subjects with newly diagnosed rheumatoid arthritis. Intern Emerg Med 2021; 16:1467-1475. [PMID: 33386606 DOI: 10.1007/s11739-020-02581-z] [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: 07/10/2020] [Accepted: 11/20/2020] [Indexed: 10/22/2022]
Abstract
Although rheumatoid arthritis (RA) is associated with an increased risk of death and cardiovascular (CV) disease, the excess of these risks is expected to have diminished over time, in more recent incident cohorts with RA. We analysed the risk of all-cause death, stroke, and myocardial infarction as primary outcomes and all CV events as secondary outcomes in RA subjects compared to the general population, from 2005 to 2018. The risk outcomes were also evaluated in relation to the time since RA diagnosis. We conducted a cohort study using linkable administrative healthcare databases of the Lombardy Region, Northern Italy. Analyses included subjects newly diagnosed RA subjects and a random sample of No-RA subjects. An adjusted Cox proportional hazard regression model was used to calculate hazard ratios and 95% CIs for all outcomes. The study population comprised 16,047 RA subjects and 500,000 without RA. The risks of dying (HR 1.22, 95% CI 1.15-1.30), stroke (HR 1.39, 95% CI 1.22-1.58), myocardial infarction (HR 2.00, 95% CI 1.78-2.26) were significantly higher in the RA cohort, as were those that for secondary outcomes. Differences between RA and No-RA already emerged during the first five years after diagnosis. Risk patterns remained statistically significant during the next 5 years or more. Subjects with RA still have a higher risk of death and worse CV outcomes than the general population, appearing early and not decreasing with time. Preventive interventions are urgently needed.
Collapse
Affiliation(s)
- Marta Baviera
- Laboratory of Cardiovascular Prevention, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy.
| | - Giovanni Cioffi
- Rheumatology Section, Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
- Division of Cardiac Rehabilitation, S. Pancrazio Hospital, Arco, Trento, Italy
| | - Pierluca Colacioppo
- Laboratory of Cardiovascular Prevention, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Mauro Tettamanti
- Laboratory of Geriatric Neuropsychiatry, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Ida Fortino
- Regional Health Ministry, Lombardy Region, Milan, Italy
| | - Maria Carla Roncaglioni
- Laboratory of Cardiovascular Prevention, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| |
Collapse
|
8
|
Abel D, Ardoin SP, Gorelik M. The potential role of Colchicine in preventing coronary vascular disease in childhood-onset lupus: a new view on an old drug. Pediatr Rheumatol Online J 2021; 19:15. [PMID: 33593369 PMCID: PMC7885423 DOI: 10.1186/s12969-021-00504-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 02/08/2021] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Patients with systemic lupus erythematous have a significantly increased risk of cardiovascular disease, which is not fully explained by traditional cardiovascular disease risk factors. Despite increasing life expectancy in patients with systemic lupus erythematous, mortality due to cardiovascular disease, the major cause of death in these patients, has not changed. Children with lupus suffer from more aggressive disease compared to their adult counterparts, and there is a growing concern for their increased risk of cardiovascular disease as they age. BODY: There is an unmet need for therapies to address the increased risk of cardiovascular disease in childhood-onset lupus. Colchicine has many anti-inflammatory and cardiovascular protective properties, including inhibition of IL-1β and IL-18 activity, key proinflammatory cytokines that are predictive of future adverse cardiovascular events. In the Colchicine Cardiovascular Outcomes Trial (COLCOT), colchicine was recently found to have significant benefit with minimal risk in adults with previous myocardial infarction for prevention of secondary vascular disease. While adult studies are promising, no studies have been conducted in pediatric patients to investigate colchicine's potential for cardiovascular protection in children and adolescents with lupus. CONCLUSIONS Studies investigating colchicine's potential role for cardiovascular protection are needed in pediatric patients with systemic lupus erythematous.
Collapse
Affiliation(s)
- Dori Abel
- Department of Pediatrics, NewYork-Presbyterian Hospital, Columbia University Irving Medical Center, 630 W. 168th Street, New York, NY, 10032-3702, USA.
| | - Stacy P. Ardoin
- grid.261331.40000 0001 2285 7943Department of Medicine, Division of Rheumatology and Immunology, The Ohio State University, 370 W. 9th Ave, Columbus, OH 43210 USA ,grid.240344.50000 0004 0392 3476Department of Rheumatology, Nationwide Children’s Hospital, 700 Children’s Dr, Columbus, OH 43205 USA
| | - Mark Gorelik
- grid.21729.3f0000000419368729Department of Pediatrics, Division of Allergy, Immunology, and Rheumatology, Columbia University Irving Medical Center, 630 W. 168th St, New York, NY 10032-3702 USA
| |
Collapse
|
9
|
Sciarra I, Vasile M, Carboni A, Stefanantoni K, Iannace N, Angelelli C, Scarno AG, Valesini G, Riccieri V. Subclinical atherosclerosis in systemic sclerosis: Different risk profiles among patients according to clinical manifestations. Int J Rheum Dis 2020; 24:502-509. [PMID: 33073534 DOI: 10.1111/1756-185x.14002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 09/27/2020] [Accepted: 09/28/2020] [Indexed: 01/24/2023]
Abstract
INTRODUCTION Like other autoimmune diseases, systemic sclerosis (SSc) has been described to be associated with accelerated atherosclerosis (ATS). Before clinical manifestations of cardiovascular disease (CVD) occur, subclinical ATS can be investigated in different ways. AIM To evaluate the presence of subclinical ATS in a group of patients with SSc, and to identify different risk profiles among patients. METHODS Subclinical ATS was reviewed in 43 SSc patients and 27 healthy controls, using 2 methods: carotid ultrasound and flow mediated dilation (FMD) of the brachial artery. RESULTS Plaques were statistically more frequent in SSc patients than in controls (65% vs 30%, P = .006); intima-media thickness of common carotid artery (CCA-IMT) resulted in statistically higher (median value 0.8 mm vs 0.55 mm; P < .0001) while FMD was significantly lower (median value 9% vs 14%; P = .0086) in patients compared to healthy controls. Among the SSc patients, thickening of CCA-IMT was significantly associated with the presence of diastolic dysfunction of left ventricle (absence of diastolic dysfunction: odds ratio [OR] 0.2, 95% CI 0.04-0.92, P = .038) and with a higher Framingham score (OR 1.3, 95% CI 1.03-1.6], P = .024). The diffuse cutaneous form was slightly protective against pathological FMD (OR 0.12, 95% CI 0.022-0.71, P = .019). CONCLUSIONS This study confirms the involvement of macrocirculation in SSc patients, detecting the presence of subclinical ATS markers more frequently in patients compared to healthy controls. Framingham score, diastolic dysfunction of left ventricle and limited cutaneous form of the disease appeared to be associated with a higher risk of developing ATS.
Collapse
Affiliation(s)
- Iliana Sciarra
- Department of Clinical Internistic Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy.,Department of Biomedical Sciences and Public Health, Marche Polytechnic University, Ancona, Italy
| | - Massimiliano Vasile
- Department of Clinical Internistic Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Alessia Carboni
- Department of Clinical Internistic Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Katia Stefanantoni
- Department of Clinical Internistic Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Nicoletta Iannace
- Department of Clinical Internistic Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Carlotta Angelelli
- Department of Clinical Internistic Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Antongiulio G Scarno
- Department of Clinical Internistic Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Guido Valesini
- Department of Clinical Internistic Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Valeria Riccieri
- Department of Clinical Internistic Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
10
|
Hsu CW, Ng KJ, Lu MC, Koo M. Predicting the Risk of Ischemic Stroke among Patients with Rheumatoid Arthritis Using a Simplified RA-CHADSV Score Based on the CHA 2DS 2-VASc Score. ACTA ACUST UNITED AC 2020; 56:medicina56020073. [PMID: 32059458 PMCID: PMC7073594 DOI: 10.3390/medicina56020073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 11/16/2022]
Abstract
Background and Objectives: The aim of this retrospective cohort study was to develop a new score (RA-CHADSV) (rheumatoid arthritis - congestive heart failure, hypertension, age ≥75 years, diabetes mellitus, stroke/transient ischemic attack/thromboembolism, and vascular disease), modified from the CHA2DS2-VASc score (congestive heart failure, hypertension, age ≥75 years (doubled), diabetes mellitus, stroke/transient ischemic attack (doubled), vascular disease, age 65–74 years, and female), in predicting the risk of ischemic stroke in rheumatoid arthritis (RA) patients without atrial fibrillation (AF). Materials and Methods: Using the Taiwan’s National Health Insurance Research Database, 592 patients with RA diagnosed between 2000 and 2002 were identified and followed until first occurrence of ischemic stroke or the last available date in the database. Incidence rate ratios (IRR) of ischemic stroke for the CHA2DS2-VASc score were calculated using Poisson regression models. A new prediction score RA-CHADSV was developed using multiple logistic regression analysis with bootstrap validation. Results: The area under the receiver operating characteristic curve of the newly developed RA-CHADSV score and the CHA2DS2-VASc score were 0.73 (95% confidence interval (CI) 0.64–0.82) and 0.70 (95% CI 0.61–0.79), respectively. The RA-CHADSV score was significantly associated with a higher ischemic stroke incidence in the patients who scored ≥1 (adjusted IRR 7.39, p < 0.001). Conclusions: A simplified RA-CHADSV score, with comparable efficiency as the CHA2DS2-VASc score, but easier to use clinically was developed for predicting the risk of ischemic stroke among non-AF RA patients.
Collapse
Affiliation(s)
- Chia-Wen Hsu
- Department of Medical Research, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Dalin, 62247 Chiayi, Taiwan;
- School of Nursing, College of Medicine, Chang Gung University, 33302 Taoyuan City, Taiwan
| | - Khai-Jing Ng
- Division of Allergy, Immunology and Rheumatology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Dalin, 62247 Chiayi, Taiwan; (K.-J.N.); (M.-C.L.)
| | - Ming-Chi Lu
- Division of Allergy, Immunology and Rheumatology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Dalin, 62247 Chiayi, Taiwan; (K.-J.N.); (M.-C.L.)
- School of Medicine, Tzu Chi University, Hualien City, 97004 Hualien, Taiwan
| | - Malcolm Koo
- Graduate Institute of Long-term Care, Tzu Chi University of Science and Technology, Hualien City, 97005 Hualien, Taiwan
- Dalla Lana School of Public Health, University of Toronto, Toronto, M5T 3M7 ON, Canada
- Correspondence:
| |
Collapse
|
11
|
The role of proprotein convertase subtilisin-kexin type 9 (PCSK9) in the vascular aging process - is there a link? POLISH JOURNAL OF THORACIC AND CARDIOVASCULAR SURGERY 2019; 16:128-132. [PMID: 31708986 PMCID: PMC6836637 DOI: 10.5114/kitp.2019.88602] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 09/04/2019] [Indexed: 12/11/2022]
Abstract
Lately there are many new, promising low-density lipoprotein cholesterol reducing therapies with PCSK9 inhibitors. We performed selected sampling of the publications in PubMed and made a review according to selected keywords. It summarizes the effect of PCSK9 on vascular aging, directly associated with lipid and glucose metabolism, chronic inflammation, atherosclerosis and hypertension. Serum level of PCSK9 is different in patients affected by certain illnesses (whose risk increases with age) than in healthy individuals. The same could be observed in the case of chronic inflammation. In this review we summarize what is known about the role PCSK9 in human metabolism and how this could affect the vascular aging process. Based on the available sources, we prove that PCSK9 is involved in many biochemical pathways associated with vascular aging. In the future, treatments using PCSK9 inhibition may not only reduce the cardiovascular risk but also slow down this process.
Collapse
|
12
|
Kalinkovich A, Gabdulina G, Livshits G. Autoimmunity, inflammation, and dysbiosis mutually govern the transition from the preclinical to the clinical stage of rheumatoid arthritis. Immunol Res 2018; 66:696-709. [DOI: 10.1007/s12026-018-9048-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
13
|
Nakao M, Nakamura K, Fukasawa T, Shida R, Ito A, Ichimura Y, Takahashi T, Mitsui A, Yoshizaki A, Shibata S, Kamata M, Araki M, Watanabe R, Sato S, Asano Y. Assessment of endothelial function during the loading phase of infliximab in psoriasis: a potential predictor of its drug survival. Int J Dermatol 2018; 58:54-59. [PMID: 30168849 DOI: 10.1111/ijd.14200] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 08/06/2018] [Indexed: 12/01/2022]
Abstract
BACKGROUND Tumor necrosis factor inhibitors decrease the risk of cardiovascular events in moderate to severe psoriasis, but the association between their effects on endothelial function and those on skin lesions has not been well studied. We investigated the association between infliximab effects on endothelial function during the loading phase and those on skin lesions in patients with psoriasis. METHODS We evaluated endothelial function with reactive hyperemia-peripheral arterial tonometry index (RHI) in 15 patients with psoriasis before the first and third infusions of infliximab. Patients were stratified into two groups; those who maintained Psoriasis Area and Severity Index (PASI) 75 response for more than 6 months (defined as responders) and the others (defined as nonresponders). RESULTS Six weeks after the initiation of infliximab (before the third infusion), PASI scores were significantly improved compared with baseline, while RHI values were not altered in the whole patient group. However, when the responders and the nonresponders were analyzed separately, RHI values tended to be decreased before the third infusion compared with baseline in the nonresponders, while being unchanged in the responders. Importantly, the difference in ∆RHI reached a statistical significance between the two groups, and the cutoff value (mean - 2 standard deviation of RHI values in the responders) identified the nonresponders with 67% of sensitivity and 100% of specificity. CONCLUSIONS The decrease in RHI values before the third infusion may serve as a predictor for the long-term unfavorable effect of infliximab on psoriatic skin lesions.
Collapse
Affiliation(s)
- Momoko Nakao
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Kouki Nakamura
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takemichi Fukasawa
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Rino Shida
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Arisa Ito
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Yohei Ichimura
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takehiro Takahashi
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Aya Mitsui
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Ayumi Yoshizaki
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Sayaka Shibata
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Masahiro Kamata
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan.,Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | - Mayuko Araki
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Rei Watanabe
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan.,Department of Dermatology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Shinichi Sato
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Yoshihide Asano
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
14
|
Frismantiene A, Philippova M, Erne P, Resink TJ. Smooth muscle cell-driven vascular diseases and molecular mechanisms of VSMC plasticity. Cell Signal 2018; 52:48-64. [PMID: 30172025 DOI: 10.1016/j.cellsig.2018.08.019] [Citation(s) in RCA: 279] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 08/28/2018] [Accepted: 08/28/2018] [Indexed: 02/06/2023]
Abstract
Vascular smooth muscle cells (VSMCs) are the major cell type in blood vessels. Unlike many other mature cell types in the adult body, VSMC do not terminally differentiate but retain a remarkable plasticity. Fully differentiated medial VSMCs of mature vessels maintain quiescence and express a range of genes and proteins important for contraction/dilation, which allows them to control systemic and local pressure through the regulation of vascular tone. In response to vascular injury or alterations in local environmental cues, differentiated/contractile VSMCs are capable of switching to a dedifferentiated phenotype characterized by increased proliferation, migration and extracellular matrix synthesis in concert with decreased expression of contractile markers. Imbalanced VSMC plasticity results in maladaptive phenotype alterations that ultimately lead to progression of a variety of VSMC-driven vascular diseases. The nature, extent and consequences of dysregulated VSMC phenotype alterations are diverse, reflecting the numerous environmental cues (e.g. biochemical factors, extracellular matrix components, physical) that prompt VSMC phenotype switching. In spite of decades of efforts to understand cues and processes that normally control VSMC differentiation and their disruption in VSMC-driven disease states, the crucial molecular mechanisms and signalling pathways that shape the VSMC phenotype programme have still not yet been precisely elucidated. In this article we introduce the physiological functions of vascular smooth muscle/VSMCs, outline VSMC-driven cardiovascular diseases and the concept of VSMC phenotype switching, and review molecular mechanisms that play crucial roles in the regulation of VSMC phenotypic plasticity.
Collapse
Affiliation(s)
- Agne Frismantiene
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Maria Philippova
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Paul Erne
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Therese J Resink
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland.
| |
Collapse
|
15
|
Byrne CJ, Khurana S, Kumar A, Tai TC. Inflammatory Signaling in Hypertension: Regulation of Adrenal Catecholamine Biosynthesis. Front Endocrinol (Lausanne) 2018; 9:343. [PMID: 30013513 PMCID: PMC6036303 DOI: 10.3389/fendo.2018.00343] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/07/2018] [Indexed: 12/24/2022] Open
Abstract
The immune system is increasingly recognized for its role in the genesis and progression of hypertension. The adrenal gland is a major site that coordinates the stress response via the hypothalamic-pituitary-adrenal axis and the sympathetic-adrenal system. Catecholamines released from the adrenal medulla function in the neuro-hormonal regulation of blood pressure and have a well-established link to hypertension. The immune system has an active role in the progression of hypertension and cytokines are powerful modulators of adrenal cell function. Adrenal medullary cells integrate neural, hormonal, and immune signals. Changes in adrenal cytokines during the progression of hypertension may promote blood pressure elevation by influencing catecholamine biosynthesis. This review highlights the potential interactions of cytokine signaling networks with those of catecholamine biosynthesis within the adrenal, and discusses the role of cytokines in the coordination of blood pressure regulation and the stress response.
Collapse
Affiliation(s)
- Collin J. Byrne
- Department of Biology, Laurentian University, Sudbury, ON, Canada
| | - Sandhya Khurana
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, ON, Canada
| | - Aseem Kumar
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON, Canada
- Biomolecular Sciences Program, Laurentian University, Sudbury, ON, Canada
| | - T. C. Tai
- Department of Biology, Laurentian University, Sudbury, ON, Canada
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, ON, Canada
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON, Canada
- Biomolecular Sciences Program, Laurentian University, Sudbury, ON, Canada
| |
Collapse
|
16
|
Kasselman LJ, Vernice NA, DeLeon J, Reiss AB. The gut microbiome and elevated cardiovascular risk in obesity and autoimmunity. Atherosclerosis 2018. [DOI: 10.1016/j.atherosclerosis.2018.02.036] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
17
|
Hrycek E, Banasiewicz‐Szkróbka I, Żurakowski A, Buszman P, Hrycek A. Accelerated Atherosclerosis in Patients with Systemic Lupus Erythematosus and the Role of Selected Adipocytokines in This Process. Lupus 2017. [DOI: 10.5772/68016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
18
|
Assessment of autonomic function in a cohort of patients with ANCA-associated vasculitis. Clin Auton Res 2016; 26:279-85. [DOI: 10.1007/s10286-016-0364-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 05/26/2016] [Indexed: 10/21/2022]
|
19
|
Anti-citrullinated peptide antibodies and their value for predicting responses to biologic agents: a review. Rheumatol Int 2016; 36:1043-63. [PMID: 27271502 DOI: 10.1007/s00296-016-3506-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 05/25/2016] [Indexed: 12/16/2022]
Abstract
Anti-citrullinated peptide antibodies (ACPAs) play an important pathogenic role both at the onset and during the disease course. These antibodies precede the clinical appearance of rheumatoid arthritis (RA) and are associated with a less favorable prognosis, both clinically and radiologically. The objective of this work was to conduct a comprehensive review of studies published through September 2015 of ACPAs' role as a predictor of the therapeutic response to the biological agents in RA patients. The review also includes summary of the biology and detection of ACPAs as well as ACPAs in relation to joint disease and CV disease and the possible role of seroconversion. The reviews of studies examining TNF inhibitors and tocilizumab yielded negative results. In the case of rituximab, the data indicated a greater probability of clinical benefit in ACPA(+) patients versus ACPA(-) patients, as has been previously described for rheumatoid factor. Nonetheless, the effect is discreet and heterogeneous. Another drug that may have greater effectiveness in ACPA(+) patients is abatacept. Some studies have suggested that the drug is more efficient in ACPA(+) patients and that those patients show greater drug retention. In a subanalysis of the AMPLE trial, patients with very high ACPA titers who were treated with abatacept had a statistically significant response compared to patients with lower titers. In summary, the available studies suggest that the presence of or high titers of ACPA may predict a better response to rituximab and/or abatacept. Evidence regarding TNFi and tocilizumab is lacking. However, there is a lack of studies with appropriate designs to demonstrate that some drugs are superior to others for ACPA(+) patients.
Collapse
|
20
|
Voloshyna I, Teboul I, Littlefield MJ, Siegart NM, Turi GK, Fazzari MJ, Carsons SE, DeLeon J, Reiss AB. Resveratrol counters systemic lupus erythematosus-associated atherogenicity by normalizing cholesterol efflux. Exp Biol Med (Maywood) 2016; 241:1611-9. [PMID: 27190277 DOI: 10.1177/1535370216647181] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 04/01/2016] [Indexed: 11/16/2022] Open
Abstract
Resveratrol is a bioactive molecule used in dietary supplements and herbal medicines and consumed worldwide. Numerous investigations by our group and others have indicated cardioprotective and anti-inflammatory properties of resveratrol. The present study explored potential atheroprotective actions of resveratrol on cholesterol efflux in cultured human macrophages exposed to plasma from systemic lupus erythematosus (SLE) patients. These results were confirmed in ApoE(-/-)Fas(-/-) double knockout mice, displaying a lupus profile with accelerated atherosclerosis. Resveratrol treatment attenuated atherosclerosis in these mice. THP-1 human macrophages were exposed to 10% pooled or individual plasma from patients who met diagnostic criteria for SLE. Expression of multiple proteins involved in reverse cholesterol transport (ABCA1, ABCG1, SR-B1, and cytochrome P450 27-hydroxylase) was assessed using QRT-PCR and Western blotting techniques. Ten-week-old ApoE(-/-)Fas(-/-) double knockout mice (n = 30) were randomly divided into two equal groups of 15, one of which received 0.01% resveratrol for 10 consecutive weeks. Atherosclerosis progression was evaluated in murine aortas. Bone marrow-derived macrophages (BMDM) were cultured and expression of cholesterol efflux proteins was analyzed in each group of mice. Our data indicate that inhibition of cholesterol efflux by lupus plasma in THP-1 human macrophages is rescued by resveratrol. Similarly, administration of resveratrol in a lupus-like murine model reduces plaque formation in vivo and augments cholesterol efflux in BMDM. This study presents evidence for a beneficial role of resveratrol in atherosclerosis in the specific setting of SLE. Therefore, resveratrol may merit investigation as an additional resource available to reduce lipid deposition and atherosclerosis in humans, especially in such vulnerable populations as lupus patients.
Collapse
Affiliation(s)
- Iryna Voloshyna
- Department of Medicine and Winthrop Research Institute, Winthrop University Hospital, Mineola, NY 11501, USA
| | - Isaac Teboul
- Department of Medicine and Winthrop Research Institute, Winthrop University Hospital, Mineola, NY 11501, USA
| | - Michael J Littlefield
- Department of Medicine and Winthrop Research Institute, Winthrop University Hospital, Mineola, NY 11501, USA
| | - Nicolle M Siegart
- Department of Medicine and Winthrop Research Institute, Winthrop University Hospital, Mineola, NY 11501, USA
| | - George K Turi
- Department of Medicine and Winthrop Research Institute, Winthrop University Hospital, Mineola, NY 11501, USA
| | - Melissa J Fazzari
- Department of Medicine and Winthrop Research Institute, Winthrop University Hospital, Mineola, NY 11501, USA
| | - Steven E Carsons
- Department of Medicine and Winthrop Research Institute, Winthrop University Hospital, Mineola, NY 11501, USA
| | - Joshua DeLeon
- Department of Medicine and Winthrop Research Institute, Winthrop University Hospital, Mineola, NY 11501, USA
| | - Allison B Reiss
- Department of Medicine and Winthrop Research Institute, Winthrop University Hospital, Mineola, NY 11501, USA
| |
Collapse
|
21
|
Skin Autofluorescence in Systemic Sclerosis Is Related to the Disease and Vascular Damage: A Cross-Sectional Analytic Study of Comparative Groups. DISEASE MARKERS 2015; 2015:837470. [PMID: 26880854 PMCID: PMC4736188 DOI: 10.1155/2015/837470] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 12/01/2015] [Accepted: 12/03/2015] [Indexed: 11/18/2022]
Abstract
Skin autofluorescence (AF), a relatively simple and time saving procedure, measures the accumulation of advanced glycation end (AGE) products. The importance in autoimmune rheumatic diseases, particularly, systemic sclerosis (SSc), has not been evaluated yet. The aim of our study was to examine the skin AF in the context of SSc patients and to analyse the relations between skin AF and other surrogate measures of atherosclerosis. Forty-seven patients with SSc and 47 healthy volunteers were included in this study as controls. Patients and controls underwent common carotid artery wall assessment, arterial stiffness and wave reflection measurements, laser Doppler measurements of capillary flow, assessment of endothelial function by brachial ultrasound, peripheral arterial tonometry, and AGE measurement by skin AF. Wall properties of the common carotid arteries and wave reflection measurements were not affected in these study patients compared to controls while measures reflecting small capillary flow were altered. The accumulation of AGE products measured by skin AF was more prominent in SSc patients than in healthy controls. AGE products' score was significantly associated with carotid radial pulse wave velocity, intima media/carotid artery diameter ratio, capillary flow percentage change during occlusion, and the disease itself in a multivariate linear analysis model.
Collapse
|
22
|
Andrade D, Bortolotto L, Bonfá E, Borba E. Primary antiphospholipid syndrome: absence of premature atherosclerosis in patients without traditional coronary artery disease risk factors. Lupus 2015; 25:472-8. [PMID: 26585072 DOI: 10.1177/0961203315617841] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 10/23/2015] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To investigate if patients with Primary Antiphospholipid Syndrome (PAPS) with venous and/or arterial thrombosis without traditional coronary artery disease (CAD) risk factors develop early atherosclerotic vascular damage. METHODS 27 female patients with PAPS (Sidney criteria) and 27 age, body mass index (BMI), and sex matched controls were consecutively selected. Exclusion criteria were: black race, age ≥55 years, traditional cardiovascular risk factors, other thrombophilias or connective tissue diseases, corticosteroids use and pregnancy. All subjects underwent Pulse Wave Velocity (PWV) and Echo-Tracking (ET), both in carotidal bed, to analyse vascular functional properties. RESULTS Age (p = 0.92) and BMI (p = 0.91) were comparable in both groups. PAPS patients and controls had similar PWV (9.07 ± 1.08 m/s vs 9.42 ± 1.47 m/s, p = 0.34) as well as echo tracking parameters such as intima-media thickness (683 ± 171 µm vs 636 ± 140 µm, p = 0.52), carotideal diameter (p = 0.26), distensibility (p = 0.92), compliance coefficients (p = 0.36) and elastic modulus (p = 0.78). Patients with exclusively venous thrombosis showed lower PWV than patients with arterial thrombosis (8.55 ± 0.70 m/s vs 9.56 ± 0.94 m/s, p = 0.01), but no difference regarding intima-media thickness (683 ± 171 µm vs 636 ± 140 µm, p = 0.52) was observed. CONCLUSION Patients with PAPS do not seem to be at higher risk of developing premature atherosclerosis. Patients who suffered exclusively venous thrombosis seem to be at lower risk than those with exclusively arterial events. Other studies need to confirm our findings.
Collapse
Affiliation(s)
- D Andrade
- Rheumatology Division, São Paulo University, São Paulo, Brazil
| | - L Bortolotto
- Heart Institute of São Paulo University, São Paulo, Brazil
| | - E Bonfá
- Rheumatology Division, São Paulo University, São Paulo, Brazil
| | - E Borba
- Rheumatology Division, São Paulo University, São Paulo, Brazil
| |
Collapse
|
23
|
Mankad R. Atherosclerotic vascular disease in the autoimmune rheumatologic patient. Curr Atheroscler Rep 2015; 17:497. [PMID: 25721102 DOI: 10.1007/s11883-015-0497-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Autoimmune diseases, such as rheumatoid arthritis and systemic lupus erythematosus, have a strong association with an increased risk of atherosclerotic cardiovascular diseases (ASCVD), particularly ischemic heart disease (IHD). A majority of the autoimmune conditions occur predominantly in women, and as women continue to experience a higher cardiovascular mortality compared to men, this potential added risk factor must be recognized. Inflammation and immune mechanisms have been shown to be an underlying mechanism for the development of atherosclerosis, thus sharing a common mechanism with rheumatologic conditions. There is an under recognition, in both patient and physician, of the increased cardiovascular (CV) risk within the autoimmune population, with present CV risk profile algorithms performing poorly in these patients. Traditional risk factors play a role in the development of IHD in the autoimmune patient, but their overall significance is unclear and does not fully explain the elevated CV risk. The role of inflammation and risk factors in autoimmune conditions, and their link to the elevated CV risk will be explored within this article.
Collapse
Affiliation(s)
- Rekha Mankad
- Division of Cardiovascular Diseases, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA,
| |
Collapse
|
24
|
Baraka E, El Dein M, Farouk H, El Moutaz Y. Hyperhomocysteinemia and metabolic syndrome are risk factors for sub-clinical atherosclerosis in women with systemic lupus erythematosus. EGYPTIAN RHEUMATOLOGIST 2015. [DOI: 10.1016/j.ejr.2014.07.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
25
|
Lerner A, Blank M. Hypercoagulability in celiac disease--an update. Autoimmun Rev 2014; 13:1138-41. [PMID: 25149392 DOI: 10.1016/j.autrev.2014.07.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 07/07/2014] [Indexed: 12/13/2022]
Abstract
Celiac disease is a life-long autoimmune disease affecting multiple organs of genetically susceptible individuals. One of the extra intestinal manifestations of the disease is thromboembolic events like strokes, veins' thrombosis, and pregnancy losses. Hypercoagulable autoimmune diseases like lupus erythematosus and antiphospholipid syndrome, associated with celiac disease just add risk to the patients. Pathogenic predisposing avenues increasing the hypercoagulability in celiac disease are multiple: nutritional deficiencies (B12, folate, and vitamin K), genetic predisposition (MTHFR mutations), thrombophilic autoantibodies, hyperhomocysinemia, endothelial dysfunction and platelet abnormalities. Primary pharmacologic thromboprophylaxis or treating the predisposing factors should be considered on a personal basis.
Collapse
Affiliation(s)
- Aaron Lerner
- Pediatric Gastroenterology and Nutrition Unit, Carmel Medical Center, B. Rappaport School of Medicine, Technion-Israel institute of Technology, Haifa 34362, Israel.
| | - Miri Blank
- The Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Ashomer 52621, Israel
| |
Collapse
|
26
|
Amaya-Amaya J, Montoya-Sánchez L, Rojas-Villarraga A. Cardiovascular involvement in autoimmune diseases. BIOMED RESEARCH INTERNATIONAL 2014; 2014:367359. [PMID: 25177690 PMCID: PMC4142566 DOI: 10.1155/2014/367359] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 05/01/2014] [Indexed: 12/15/2022]
Abstract
Autoimmune diseases (AD) represent a broad spectrum of chronic conditions that may afflict specific target organs or multiple systems with a significant burden on quality of life. These conditions have common mechanisms including genetic and epigenetics factors, gender disparity, environmental triggers, pathophysiological abnormalities, and certain subphenotypes. Atherosclerosis (AT) was once considered to be a degenerative disease that was an inevitable consequence of aging. However, research in the last three decades has shown that AT is not degenerative or inevitable. It is an autoimmune-inflammatory disease associated with infectious and inflammatory factors characterized by lipoprotein metabolism alteration that leads to immune system activation with the consequent proliferation of smooth muscle cells, narrowing arteries, and atheroma formation. Both humoral and cellular immune mechanisms have been proposed to participate in the onset and progression of AT. Several risk factors, known as classic risk factors, have been described. Interestingly, the excessive cardiovascular events observed in patients with ADs are not fully explained by these factors. Several novel risk factors contribute to the development of premature vascular damage. In this review, we discuss our current understanding of how traditional and nontraditional risk factors contribute to pathogenesis of CVD in AD.
Collapse
Affiliation(s)
- Jenny Amaya-Amaya
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 No. 63C-69, 11001000 Bogotá, Colombia
- Mederi, Hospital Universitario Mayor, Calle 24 No. 29-45, 11001000 Bogotá, Colombia
| | - Laura Montoya-Sánchez
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 No. 63C-69, 11001000 Bogotá, Colombia
- Mederi, Hospital Universitario Mayor, Calle 24 No. 29-45, 11001000 Bogotá, Colombia
| | - Adriana Rojas-Villarraga
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 No. 63C-69, 11001000 Bogotá, Colombia
- Mederi, Hospital Universitario Mayor, Calle 24 No. 29-45, 11001000 Bogotá, Colombia
| |
Collapse
|
27
|
Edema and fibrosis imaging by cardiovascular magnetic resonance: how can the experience of Cardiology be best utilized in rheumatological practice? Semin Arthritis Rheum 2014; 44:76-85. [PMID: 24582213 DOI: 10.1016/j.semarthrit.2014.01.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 10/19/2013] [Accepted: 01/17/2014] [Indexed: 12/19/2022]
Abstract
OBJECTIVES CMR, a non-invasive, non-radiating technique can detect myocardial oedema and fibrosis. METHOD CMR imaging, using T2-weighted and T1-weighted gadolinium enhanced images, has been successfully used in Cardiology to detect myocarditis, myocardial infarction and various cardiomyopathies. RESULTS Transmitting this experience from Cardiology into Rheumatology may be of important value because: (a) heart involvement with atypical clinical presentation is common in autoimmune connective tissue diseases (CTDs). (b) CMR can reliably and reproducibly detect early myocardial tissue changes. (c) CMR can identify disease acuity and detect various patterns of heart involvement in CTDs, including myocarditis, myocardial infarction and diffuse vasculitis. (d) CMR can assess heart lesion severity and aid therapeutic decisions in CTDs. CONCLUSION The CMR experience, transferred from Cardiology into Rheumatology, may facilitate early and accurate diagnosis of heart involvement in these diseases and potentially targeted heart treatment.
Collapse
|
28
|
Morris-Rosenfeld S, Lipinski MJ, McNamara CA. Understanding the role of B cells in atherosclerosis: potential clinical implications. Expert Rev Clin Immunol 2013; 10:77-89. [PMID: 24308836 DOI: 10.1586/1744666x.2014.857602] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Atherosclerosis is a progressive inflammatory disease of the medium to large arteries that is the largest contributor to cardiovascular disease. B-cell subsets have been shown in animal models of atherosclerosis to have both atherogenic and atheroprotective properties. In this review, we highlight the research that developed our understanding of the role of B cells in atherosclerosis both in humans and mice. From this we discuss the potential clinical impact B cells could have both as diagnostic biomarkers and as targets for immunotherapy. Finally, we recognize the inherent difficulty in translating findings from animal models into humans given the differences in both cardiovascular disease and the immune system between mice and humans, making the case for greater efforts at addressing the role of B cells in human atherosclerosis.
Collapse
Affiliation(s)
- Samuel Morris-Rosenfeld
- Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA and Department of Medicine, Cardiovascular Division at the University of Virginia, Charlottesville, VA, USA
| | | | | |
Collapse
|
29
|
Actin is a target of T-cell reactivity in patients with advanced carotid atherosclerotic plaques. Mediators Inflamm 2013; 2013:261054. [PMID: 24324294 PMCID: PMC3844233 DOI: 10.1155/2013/261054] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 10/07/2013] [Indexed: 12/04/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall associated with autoimmune reactions. In a previous study, we observed the presence of actin-specific antibodies in sera from patients with carotid atherosclerosis. To extend our previous results we evaluated the possible role of actin as antigenic target of cell-mediated immune reactions in carotid atherosclerosis. Peripheral blood mononuclear cells (PBMC) from 17 patients and 16 healthy subjects were tested by cell proliferation assay and by ELISA for cytokine production. Actin induced a proliferative response in 47% of patients' PBMC samples, with SI ranging from 2.6 to 21.1, and in none of the healthy subjects' samples (patients versus healthy subjects, P = 0.02). The presence of diabetes in patients was significantly associated with proliferative response to actin (P = 0.04). IFN-γ and TNF-α concentrations were higher in PBMC from patients than in those from healthy subjects and in PBMC proliferating to actin than in nonproliferating ones. Our data demonstrate for the first time a role of actin as a target autoantigen of cellular immune reactions in patients with carotid atherosclerosis. The preferential proinflammatory Th1 activation suggests that actin could contribute to endothelial dysfunction, tissue damage, and systemic inflammation in carotid atherosclerosis.
Collapse
|
30
|
Wang PC, Lin HC, Kang JH. Chronic Rhinosinusitis Confers an Increased Risk of Acute Myocardial Infarction. Am J Rhinol Allergy 2013; 27:e178-82. [DOI: 10.2500/ajra.2013.27.3952] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background The link between chronic inflammatory disease and cardiovascular disease (CVD) is recognized. Chronic rhinosinusitis (CRS) is one of the most common chronic inflammatory diseases. However, whether CRS increases the risk for CVD is still unknown. This epidemiology study investigated the risk for acute myocardial infarction (AMI) in patients with CRS using a large-scale population-based cohort study. Methods Data on all study cohorts were retrieved from the Longitudinal Health Insurance Database in Taiwan. In total, data on 7975 CRS subjects from 2001 to 2003 were extracted for the study cohort. We selected 39,875 comparison subjects whose demographic variables matched those of the study cohort. We individually tracked each subject for a 6-year period (from 2001 to 2009) to identify which subjects subsequently received a diagnosis of AMI. A stratified Cox proportional hazards regression was used to compare the 6-year risk of a subsequent AMI after a diagnosis of CRS. Results Among the 47,850 sampled subjects, the incidence rate of AMI during the 6-year follow-up period was 5.66/1000 person-years; rates were 8.49 and 5.09/1000 person-years for the study and comparison cohort, respectively. The hazard ratio (HR) for AMI during the 6-year follow-up period for subjects with CRS was 1.70 (95% confidence interval [CI], 1.52~1.91). In addition, after adjusting for cardiovascular risk factors, the HR of AMI for subjects with CRS was 1.48 (95% CI, 1.32~1.67) compared with subjects without CRS. Conclusion Patients with CRS were at higher risk for AMI occurrence in the 6-year follow-up.
Collapse
Affiliation(s)
- Pi-Chieh Wang
- Department of Family Medicine, PoJen General Hospital, Taipei, Taiwan
| | | | - Jiunn-Horng Kang
- Department of Physical Medicine and Rehabilitation, Taipei, Taiwan
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
31
|
García-Bermúdez M, López-Mejías R, Genre F, Castañeda S, González-Juanatey C, Llorca J, Corrales A, Miranda-Filloy JA, Pina T, Gómez-Vaquero C, Rodríguez-Rodríguez L, Fernández-Gutiérrez B, Pascual-Salcedo D, Balsa A, López-Longo FJ, Carreira P, Blanco R, González-Álvaro I, Martín J, González-Gay MA. Single-nucleotide polymorphisms at the 9p21.3 genomic region not associated with the risk of cardiovascular disease in patients with rheumatoid arthritis. ACTA ACUST UNITED AC 2013; 82:405-9. [DOI: 10.1111/tan.12227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 09/13/2013] [Accepted: 09/22/2013] [Indexed: 02/02/2023]
Affiliation(s)
- M. García-Bermúdez
- Instituto de Parasitología y Biomedicina López-Neyra, IPBLN-CSIC; 18016 Granada Spain
| | - R. López-Mejías
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division; Hospital Universitario Marqués de Valdecilla, IFIMAV; 39008 Santander Spain
| | - F. Genre
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division; Hospital Universitario Marqués de Valdecilla, IFIMAV; 39008 Santander Spain
| | - S. Castañeda
- Rheumatology Department; Hospital Universitario la Princesa, IIS-Princesa; 28006 Madrid Spain
| | | | - J. Llorca
- Department of Epidemiology and Computational Biology, School of Medicine; University of Cantabria, and CIBER Epidemiología y Salud Pública (CIBERESP), IFIMAV; 39011 Santander Spain
| | - A. Corrales
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division; Hospital Universitario Marqués de Valdecilla, IFIMAV; 39008 Santander Spain
| | | | - T. Pina
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division; Hospital Universitario Marqués de Valdecilla, IFIMAV; 39008 Santander Spain
| | - C. Gómez-Vaquero
- Department of Rheumatology; Hospital Universitario Bellvitge; 08908 Barcelona Spain
| | | | | | - D. Pascual-Salcedo
- Rheumatology Department; Hospital Universitario La Paz; 28046 Madrid Spain
| | - A. Balsa
- Rheumatology Department; Hospital Universitario La Paz; 28046 Madrid Spain
| | - F. J. López-Longo
- Rheumatology Department; Hospital General Universitario Gregorio Marañón; 28007 Madrid Spain
| | - P. Carreira
- Rheumatology Department; Hospital Universitario 12 de Octubre; 28041 Madrid Spain
| | - R. Blanco
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division; Hospital Universitario Marqués de Valdecilla, IFIMAV; 39008 Santander Spain
| | - I. González-Álvaro
- Rheumatology Department; Hospital Universitario la Princesa, IIS-Princesa; 28006 Madrid Spain
| | - J. Martín
- Instituto de Parasitología y Biomedicina López-Neyra, IPBLN-CSIC; 18016 Granada Spain
| | - M. A. González-Gay
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division; Hospital Universitario Marqués de Valdecilla, IFIMAV; 39008 Santander Spain
| |
Collapse
|
32
|
Chung WS, Lin CL, Peng CL, Chen YF, Lu CC, Sung FC, Kao CH. Rheumatoid arthritis and risk of acute myocardial infarction--a nationwide retrospective cohort study. Int J Cardiol 2013; 168:4750-4. [PMID: 23938220 DOI: 10.1016/j.ijcard.2013.07.233] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Revised: 07/23/2013] [Accepted: 07/25/2013] [Indexed: 10/26/2022]
Abstract
OBJECTIVES Rheumatoid arthritis (RA) imposes substantial social costs, including an increased risk of work-related disability and accelerated cardiovascular diseases. The aim of the study is to determine the risk of acute myocardial infarction (AMI) associated with RA in a nationwide retrospective cohort study. METHODS Using the catastrophic illness registry of the Taiwan National Health Insurance Research Database (NHIRD), we identified patients with RA from 1998 to 2010. We also randomly selected non-RA controls frequency-matched by age, sex, and index year from the general population free of RA. The risk of AMI was analyzed using Cox proportional hazards regression models including sex, age, and comorbidities. RESULTS From a total of 23.74 million people in the cohort, 29,260 RA patients and 117,040 controls were followed for 193,987 and 792,254 person-years, respectively. The incidence density rate increased in all groups of RA patients than those of the controls. RA patients had a 1.33-fold higher overall incidence of AMI than controls, with an adjusted hazard ration of 1.38. Although the overall adjusted hazard ratio of AMI increased with age, the age-specific RA patients to controls incidence rate ratio was higher for younger RA patients. Subjects with comorbidities of hypertension, diabetes hyperlipidemia, CVA, COPD, or ESRD had increased risk of AMI. Subjects with ESRD had the highest hazard of AMI. CONCLUSION This nationwide retrospective cohort study indicates that AMI risk increased by 38% in RA patients compared to the general population. Comorbidities increase the AMI risk independently.
Collapse
Affiliation(s)
- Wei-Sheng Chung
- Department of Internal Medicine, Taichung Hospital, Department of Health, Executive Yuan, Taichung, Taiwan; Department of Healthcare Administration, Central Taiwan University of Science and Technology, Taichung, Taiwan
| | | | | | | | | | | | | |
Collapse
|
33
|
Cutting-edge issues in coronary disease and the primary antiphospholipid syndrome. Clin Rev Allergy Immunol 2013; 44:51-6. [PMID: 21403998 DOI: 10.1007/s12016-011-8268-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Antiphospholipid syndrome (APS) is the most frequent cause of venous and arterial thrombotic events in young patients. The brain arterial tree is primarily affected, but coronary ischemic manifestations are also relatively frequent. Coronary involvement was suggested to be closely related to the accelerated atherosclerosis linked to the underlying disease in APS associated to systemic autoimmune diseases, in particular, systemic lupus erythematosus. However, arterial ischemic events can occur in primary APS--with no other systemic disorders--even in the absence of traditional cardiovascular risk factors and overt atherosclerosis. From a biological point of view, this finding speaks in favor for a pro-coagulant activity of anti-phospholipid antibodies rather than for their role in atherosclerotic plaque formation. On the other hand, the clinical challenge is to avoid the risk to misdiagnose young patients with potentially life-threatening symptoms, such as myocardial infarction (MI). In fact, the occurrence of nonspecific symptoms related to coronary ischemic events is frequently misdiagnosed because of its rarity in young patients. This issue is well illuminated by two cases of MI in young patients reported in the manuscript together with a systematic review of the associations and implications of coronary ischemic events in APS.
Collapse
|
34
|
Cardiovascular disease in autoimmune rheumatic diseases. Autoimmun Rev 2013; 12:1004-15. [PMID: 23541482 DOI: 10.1016/j.autrev.2013.03.013] [Citation(s) in RCA: 180] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Accepted: 03/07/2013] [Indexed: 12/18/2022]
Abstract
Various autoimmune rheumatic diseases (ARDs), including rheumatoid arthritis, spondyloarthritis, vasculitis and systemic lupus erythematosus, are associated with premature atherosclerosis. However, premature atherosclerosis has not been uniformly observed in systemic sclerosis. Furthermore, although experimental models of atherosclerosis support the role of antiphospholipid antibodies in atherosclerosis, there is no clear evidence of premature atherosclerosis in antiphospholipid syndrome (APA). Ischemic events in APA are more likely to be caused by pro-thrombotic state than by enhanced atherosclerosis. Cardiovascular disease (CVD) in ARDs is caused by traditional and non-traditional risk factors. Besides other factors, inflammation and immunologic abnormalities, the quantity and quality of lipoproteins, hypertension, insulin resistance/hyperglycemia, obesity and underweight, presence of platelets bearing complement protein C4d, reduced number and function of endothelial progenitor cells, apoptosis of endothelial cells, epigenetic mechanisms, renal disease, periodontal disease, depression, hyperuricemia, hypothyroidism, sleep apnea and vitamin D deficiency may contribute to the premature CVD. Although most research has focused on systemic inflammation, vascular inflammation may play a crucial role in the premature CVD in ARDs. It may be involved in the development and destabilization of both atherosclerotic lesions and of aortic aneurysms (a known complication of ARDs). Inflammation in subintimal vascular and perivascular layers appears to frequently occur in CVD, with a higher frequency in ARD than in non-ARD patients. It is possible that this inflammation is caused by infections and/or autoimmunity, which might have consequences for treatment. Importantly, drugs targeting immunologic factors participating in the subintimal inflammation (e.g., T- and B-cells) might have a protective effect on CVD. Interestingly, vasa vasorum and cardiovascular adipose tissue may play an important role in atherogenesis. Inflammation and complement depositions in the vessel wall are likely to contribute to vascular stiffness. Based on biopsy findings, also inflammation in the myocardium and small vessels may contribute to premature CVD in ARDs (cardiac ischemia and heart failure). There is an enormous need for an improved CVD prevention in ARDs. Studies examining the effect of DMARDs/biologics on vascular inflammation and CV risk are warranted.
Collapse
|
35
|
Cardiovascular disease due to accelerated atherosclerosis in systemic vasculitides. Best Pract Res Clin Rheumatol 2013; 27:33-44. [DOI: 10.1016/j.berh.2012.12.004] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 12/22/2012] [Accepted: 12/27/2012] [Indexed: 02/08/2023]
|
36
|
Study of association of CD40-CD154 gene polymorphisms with disease susceptibility and cardiovascular risk in Spanish rheumatoid arthritis patients. PLoS One 2012; 7:e49214. [PMID: 23166616 PMCID: PMC3499567 DOI: 10.1371/journal.pone.0049214] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 10/05/2012] [Indexed: 12/23/2022] Open
Abstract
Objective Rheumatoid arthritis (RA) is a chronic inflammatory disease associated with increased cardiovascular (CV) mortality. Since CD40-CD154 binding has direct consequences on inflammation process initiation, we aimed to replicate previous findings related to disease susceptibility in Spanish RA population. Furthermore, as the major complication in RA disease patients is the development of CV events due to accelerated atherosclerosis, and elevated levels of CD40L/CD154 are present in patients with acute myocardial infarction, we assessed the potential association of CD40 and CD154/CD40L gene variants with CV risk in Spanish RA patients. Methods One thousand five hundred and seventy-five patients fulfilling the 1987 ACR classification criteria for RA and 1600 matched controls were genotyped for the CD40 rs1883832, rs4810485 and rs1535045 and CD154 rs3092952 and rs3092920 gene polymorphisms, using predesigned TaqMan single nucleotide polymorphism genotyping assays. Afterwards, we investigated the influence of CD40-CD154 gene variants in the development of CV events. Also, in a subgroup of 273 patients without history of CV events, we assessed the influence of these polymorphisms in the risk of subclinical atherosclerosis determined by carotid ultrasonography. Results Nominally significant differences in the allele frequencies for the rs1883832 CD40 gene polymorphism between RA patients and controls were found (p = 0.038). Although we did not observe a significant association of CD40-CD154 gene variants with the development of CV events, an ANCOVA model adjusted for sex, age at the time of the ultrasonography assessment, follow-up time, traditional CV risk factors and anti-cyclic citrullinated peptide antibodies disclosed a significant association (p = 0.0047) between CD40 rs1535045 polymorphism and carotid intima media thickness, a surrogate marker of atherosclerosis. Conclusion Data from our pilot study indicate a potential association of rs1883832 CD40 gene polymorphism with susceptibility to RA. Also, the CD40 rs1535045 gene variant may influence development of subclinical atherosclerosis in RA patients.
Collapse
|
37
|
García-Bermúdez M, López-Mejías R, González-Juanatey C, Corrales A, Castañeda S, Ortiz AM, Miranda-Filloy JA, Gómez-Vaquero C, Fernández-Gutiérrez B, Balsa A, Pascual-Salcedo D, Blanco R, Llorca J, Martín J, González-Gay MA. CARD8 rs2043211 (p.C10X) polymorphism is not associated with disease susceptibility or cardiovascular events in Spanish rheumatoid arthritis patients. DNA Cell Biol 2012; 32:28-33. [PMID: 23088220 DOI: 10.1089/dna.2012.1836] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Rheumatoid arthritis (RA) is a complex polygenic inflammatory disease associated with accelerated atherosclerosis, which is the main cause of increased cardiovascular (CV) morbidity and mortality in RA patients. CARD8 is a constituent of inflammasome, which regulates interleukin 1-beta production, and has been associated with a worse disease course in early RA. One thousand six hundred twenty-one patients fulfilling the 1987 ACR classification criteria for RA and 1300 matched controls, were genotyped for the CARD8 rs2043211 (30T>A, p.C10X) single-nucleotide polymorphism (SNP) using predesigned TaqMan SNP genotyping assay. The genotyping success rate in our study was greater than 94%. We assessed CARD8 rs2043211 gene polymorphism results in 1530 Spanish RA patients in whom information on CV disease and CV risk factors was available at the time of the study. Also, a subgroup of patients with no history of CV events (n=276) was assessed for the potential influence of the rs2043211 variant in the development of subclinical atherosclerosis, by measurement of carotid intima-media thickness (IMT) and presence of carotid plaques. No statistically significant differences in allele or genotype frequencies for the rs2043211 CARD8 gene variant between patients with RA and controls were seen. Similarly, CARD8 rs2043211 (30T>A, p.C10X) SNP did not influence the development of CV events or the risk of CV events throughout the time. Likewise, no significant association between this gene variant and carotid IMT or the presence of plaques was found. In summary, our results do not support a role of the CARD8 rs2043211 gene variant in susceptibility to RA or in the development of CV disease in patients with RA.
Collapse
|
38
|
Antiphospholipid antibodies as non-traditional risk factors in atherosclerosis based cardiovascular diseases without overt autoimmunity. A critical updated review. Autoimmun Rev 2012; 11:873-82. [DOI: 10.1016/j.autrev.2012.03.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2012] [Accepted: 03/06/2012] [Indexed: 11/23/2022]
|
39
|
Wang X, Liu X, Kishimoto C, Yuan Z. The role of Fcγ receptors in atherosclerosis. Exp Biol Med (Maywood) 2012; 237:609-16. [PMID: 22688821 DOI: 10.1258/ebm.2012.011373] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Atherosclerosis is widely considered to be an immune-mediated process. Fcγ receptors (Fcγ Rs) contribute to the regulation of a multitude of immune and inflammatory responses and are implicated in human atherosclerotic lesions. Major cell types involved in the pathogenesis of atherosclerosis express Fcγ Rs and their proatherogenic ligands such as immune complexes and C-reactive protein, which act to activate Fcγ R signaling pathways. This review summarizes recent significant progress addressing the multifaceted roles of Fcγ Rs in atherogenesis which comes from the studies of Fcγ R-deficient animal models, clinical investigations and in vitro molecular and cellular studies. These new findings help us appreciate the emerging role of Fcγ Rs in atherosclerosis, and suggest Fcγ Rs as a potential therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Xinhong Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, China
| | | | | | | |
Collapse
|
40
|
Facts and challenges for the autoimmunologist. Lessons from the second Colombian autoimmune symposium. Autoimmun Rev 2012; 11:249-51. [DOI: 10.1016/j.autrev.2011.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
41
|
Profumo E, Buttari B, Riganò R. Oxidative stress in cardiovascular inflammation: its involvement in autoimmune responses. Int J Inflam 2011; 2011:295705. [PMID: 21755027 PMCID: PMC3132615 DOI: 10.4061/2011/295705] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 05/03/2011] [Indexed: 12/19/2022] Open
Abstract
Recently, it has become clear that atherosclerosis is a chronic inflammatory disease in which inflammation and immune responses play a key role. Accelerated atherosclerosis has been reported in patients with autoimmune diseases, suggesting an involvement of autoimmune mechanisms in atherogenesis. Different self-antigens or modified self-molecules have been identified as target of humoral and cellular immune responses in patients with atherosclerotic disease. Oxidative stress, increasingly reported in these patients, is the major event causing structural modification of proteins with consequent appearance of neoepitopes. Self-molecules modified by oxidative events can become targets of autoimmune reactions, thus sustaining the inflammatory mechanisms involved in endothelial dysfunction and plaque development. In this paper, we will summarize the best characterized autoantigens in atherosclerosis and their possible role in cardiovascular inflammation.
Collapse
Affiliation(s)
- Elisabetta Profumo
- Dipartimento di Malattie Infettive, Parassitarie ed Immunomediate, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Roma, Italy
| | | | | |
Collapse
|
42
|
Bartoloni E, Shoenfeld Y, Gerli R. Inflammatory and autoimmune mechanisms in the induction of atherosclerotic damage in systemic rheumatic diseases: Two faces of the same coin. Arthritis Care Res (Hoboken) 2011; 63:178-83. [DOI: 10.1002/acr.20322] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
43
|
Sitia S, Tomasoni L, Atzeni F, Ambrosio G, Cordiano C, Catapano A, Tramontana S, Perticone F, Naccarato P, Camici P, Picano E, Cortigiani L, Bevilacqua M, Milazzo L, Cusi D, Barlassina C, Sarzi-Puttini P, Turiel M. From endothelial dysfunction to atherosclerosis. Autoimmun Rev 2010; 9:830-4. [DOI: 10.1016/j.autrev.2010.07.016] [Citation(s) in RCA: 384] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2010] [Indexed: 12/22/2022]
|
44
|
Abstract
Atherosclerosis is now recognized as a chronic inflammatory disease and is characterized by features of inflammation at all stages of its development. It also appears to display elements of autoimmunity, and several autoantibodies including those directed against oxidized low-density lipoprotein (ox-LDL) and heat shock proteins (Hsps) have been identified in atherosclerosis. Immune complexes (ICs) may form between these antigens and autoantibodies and via Fc receptor signaling and complement activation may modulate the inflammation in atherosclerosis. Antibody isotype may direct the role that ICs play in atherogenesis, immunoglobulin G (IgG) being potentially pro-atherogenic and immunoglobulin M (IgM) playing a protective role. Therapeutic options targeting complement activation and those which are potentially Fc-receptor mediated have been investigated in animal models, though targeting Fc receptor signaling is an area that needs further investigation.
Collapse
|
45
|
Abstract
Systemic sclerosis (SSc) is a chronic disease of unknown etiology, characterized by enhanced fibrosis, and microvascular abnormalities. During the past several decades, the death rates due to cardiovascular disease or cerebrovascular disease in SSc patients substantially increased and are currently responsible for 20-30% of mortality. Various autoimmune rheumatic diseases such as rheumatoid arthritis and systemic lupus erythematosus accelerate atherosclerosis. Although microvascular disease is a hallmark of SSc, an ongoing debate exists regarding the presence and extent of macrovascular diseases and the presence of accelerated atherosclerosis in SSc patients. Despite conflicting results as to intima-media thickness (IMT) in SSc patients, the most recent and largest study has found no difference in either plaque occurrence or IMT. Additionally, abnormal coronary flow reserve in SSc patients appears to be due to microvascular involvement rather than atherosclerosis of the epicardial coronary arteries. Angiographic findings as well as computed tomography studies have generated conflicting reports as to coronary atherosclerosis in SSc. Herein, we review the current knowledge of macrovascular involvement and atherosclerosis in SSc. The differences between SSc and other autoimmune rheumatic diseases in the presence and extent of atherosclerosis need to be further investigated.
Collapse
|
46
|
Silvestrelli G, Corea F, Micheli S, Lanari A. Clinical pharmacology and vascular risk. Open Neurol J 2010; 4:64-72. [PMID: 20721317 PMCID: PMC2923338 DOI: 10.2174/1874205x01004020064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Revised: 12/21/2009] [Accepted: 12/22/2009] [Indexed: 12/23/2022] Open
Abstract
Pharmacological treatment and several drugs of abuse have been associated with ischemic heart disease (IHD) and cerebrovascular diseases (CVD). However, there is a paucity of data on the independent risk of vascular disease (VD) associated with pharmacological treatment and no controlled trials demonstrating a reduction in risk with abstinence. Information about IHD and CVD-related drug abuse is mainly limited to epidemiological studies focused on urban populations. The potential link between some pharmacological treatments (estrogen, some oncologic drugs and some atypical antipsychotics) and cerebrovascular adverse events was analyzed, but disagreement about an association persists. Drugs of abuse, including cocaine, amphetamines and heroin, have been associated with an increased vascular risk. These drugs can cause abrupt changes in blood pressure, vasculitic-type changes, lead to embolization caused by infective endocarditis, and hemostatic and hematologic abnormalities that can result in increased blood viscosity and platelet aggregation. Long-term treatment strategies based on medication, psychological support, and outreach programs play an important role in treatment of drug dependency. In these last years public interest in risk factors for VD has been constantly increasing and the successful identification and management of pharmacological treatment and drug abuse can be challenging. One of the major public health issues for the future will be to focus more on new vascular risk factor recognition and management. The objective of this chapter is to review the relevance of IHD and CVD associated with various pharmacological treatments and drug abuse with focusing on ischemic disease. This chapter reports the clinical evidence of this association and analyzes the experimental role of new drugs as a growing risk factor of VD with the hypothetical new association. In conclusion, in this chapter great attention is paid to evaluating the scientific and real evidence of cerebrovascular effect and drug use and abuse so as to identify a new groups of "modifiable" risk factors.
Collapse
Affiliation(s)
- G. Silvestrelli
- Stroke Unit, Section of Neurology, C. Poma Hospital, Mantova, Italy
| | - F. Corea
- UO Gravi Cerebrolesioni, Odpedale San Giovanni Battista, Foligno, Italy
| | - S. Micheli
- Stroke Unit, Division of Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - A. Lanari
- Stroke Unit, Section of Neurology, C. Poma Hospital, Mantova, Italy
| |
Collapse
|
47
|
|
48
|
Lack of antilipoprotein lipase antibodies in Takayasu's arteritis. Clin Dev Immunol 2009; 2009:803409. [PMID: 19606253 PMCID: PMC2709718 DOI: 10.1155/2009/803409] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Accepted: 05/23/2009] [Indexed: 11/17/2022]
Abstract
Background. Antilipoprotein lipase (anti-LPL) antibodies were described in rheumatic diseases. In systemic lupus erythematosus they were highly associated with inflammatory markers and dyslipidemia, and may ultimately contribute to vascular damage. The relevance of this association in Takayasu's arteritis, which is characterized by major inflammatory process affecting vessels, has not been determined. Objectives. To analyze the presence of anti-LPL antibodies in patients with Takayasu's arteritis and its association with inflammatory markers and lipoprotein risk levels.
Methods. Thirty sera from patients with Takayasu's arteritis, according to ACR criteria, were consecutively included. IgG anti-LPL was detected by a standard ELISA. Lipoprotein risk levels were evaluated according to NCEP/ATPIII. Inflammatory markers included ESR and CRP values. Results. Takayasu's arteritis patients had a mean age of 34 years old and all were females. Half of the patients presented high ESR and 60% elevated CRP. Lipoprotein NCEP risk levels were observed in approximately half of the patients: 53% for total cholesterol, 43% for triglycerides, 16% for HDL-c and 47% for LDL-c. In spite of the high frequency of dyslipidemia and inflammatory markers in these patients no anti-LPL were detected. Conclusions. The lack of anti-LPL antibodies in Takayasu's disease implies distinct mechanisms underlying dyslipidemia compared to systemic lupus erythematosus.
Collapse
|