1
|
Bartoloni E, Cacciapaglia F, Erre GL, Gremese E, Manfredi A, Piga M, Sakellariou G, Spinelli FR, Viapiana O, Atzeni F. Immunomodulation for accelerated atherosclerosis in rheumatoid arthritis and systemic lupus erythematosus. Autoimmun Rev 2025; 24:103760. [PMID: 39894242 DOI: 10.1016/j.autrev.2025.103760] [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: 05/18/2024] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
In the last decades, consisting evidence supported a close relationship between both innate and adaptive immune systems and the accelerated cardiovascular (CV) disease characterizing autoimmune diseases, such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Indeed, several cell lines involved in the pathogenesis of these autoimmune diseases, such as macrophages and dendritic cells, as well as different T and B lymphocyte subsets, and inflammatory cytokines, have been demonstrated to be directly involved in the mechanisms underlying early atherosclerotic arterial wall damage. Traditional CV risk factors play a concomitant role but do not sufficiently account for the increased prevalence of CV disease in these patients. Indeed, the pathophysiological link between RA and SLE and atherosclerosis is based on complex inflammatory pathways that interconnect these conditions and may explain the significant morbidity and mortality rates demonstrated in these patients, with consequent significant negative effects on quality of life and long-term survival. Consequently, it is intriguing to hypothesize that immunosuppressive drugs commonly used in the treatment of these pathologies may also exert an immunomodulatory and anti-inflammatory effect in mitigating the atherosclerotic damage that has been demonstrated to occur early in the initial stages of the disease. Recognizing risk factors, predicting occurrences and early intervention to prevent CV disease development have emerged as critical objectives in RA and SLE treatment. In this review, we aimed to provide an updated overview of the atherogenic effects exerted by the immune and inflammatory pathways involved in the pathogenesis of RA and SLE. Moreover, we examined the available evidence which may support the potential effects of immunosuppressive therapies in reducing CV damage and, consequently, CV disease risk in these patients.
Collapse
Affiliation(s)
- Elena Bartoloni
- Rheumatology Unit, Department of Medicine and Surgery, University of Perugia, Italy
| | - Fabio Cacciapaglia
- Rheumatology Unit, Department of Precision and Regenerative Medicine and Ionian Area (DePReMeI), University of Bari, Bari, Italy; Department of Medicine and Surgery, LUM University "Giuseppe De Gennaro" Casamassima & Rheumatology Service "Miulli" General Hospital Acquaviva delle Fonti, Bari, Italy
| | - Gian Luca Erre
- Dipartimento di Scienze Mediche, Chirurgiche e Sperimentali, Università degli Studi di Sassari, Italy
| | - Elisa Gremese
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Andreina Manfredi
- University of Modena and Reggio Emilia,AUSL-IRCCS Reggio Emilia, Reggio Emilia, Italy
| | - Matteo Piga
- Rheumatology Unit, AOU Cagliari, Department of Medical Sciences and Public Health, University of Cagliari, Italy
| | - Garifallia Sakellariou
- Department of Internal Medicine and Therapeutics, University of Pavia, Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Francesca Romana Spinelli
- Reumatology Unit, Department of Internal, Anesthesiological, and Cardiovascular Clinical Sciences, Sapienza University of Rome, Rome, Italy
| | - Ombretta Viapiana
- Rheumatology Unit, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Italy
| | - Fabiola Atzeni
- Rheumatology Unit, Department of Experimental and Internal Medicine, University of Messina, Italy.
| |
Collapse
|
2
|
Chee YJ, Dalan R, Cheung C. The Interplay Between Immunity, Inflammation and Endothelial Dysfunction. Int J Mol Sci 2025; 26:1708. [PMID: 40004172 PMCID: PMC11855323 DOI: 10.3390/ijms26041708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/04/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
The endothelium is pivotal in multiple physiological processes, such as maintaining vascular homeostasis, metabolism, platelet function, and oxidative stress. Emerging evidence in the past decade highlighted the immunomodulatory function of endothelium, serving as a link between innate, adaptive immunity and inflammation. This review examines the regulation of the immune-inflammatory axis by the endothelium, discusses physiological immune functions, and explores pathophysiological processes leading to endothelial dysfunction in various metabolic disturbances, including hyperglycemia, obesity, hypertension, and dyslipidaemia. The final section focuses on the novel, repurposed, and emerging therapeutic targets that address the immune-inflammatory axis in endothelial dysfunction.
Collapse
Affiliation(s)
- Ying Jie Chee
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore 308433, Singapore;
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore;
| | - Rinkoo Dalan
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore 308433, Singapore;
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore;
| | - Christine Cheung
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore;
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138632, Singapore
| |
Collapse
|
3
|
Huang C, Ding Y, Chen Z, Wu L, Wei W, Zhao C, Yang M, Lin S, Wang Q, Tian X, Zhao J, Li M, Zeng X. Future atherosclerotic cardiovascular disease in systemic lupus erythematosus based on CSTAR (XXVIII): the effect of different antiphospholipid antibodies isotypes. BMC Med 2025; 23:8. [PMID: 39757171 PMCID: PMC11702278 DOI: 10.1186/s12916-024-03843-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 12/20/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Patients with systemic lupus erythematosus (SLE) suffered from an increasing risk of cardiovascular diseases. In this multi-center prospective study, we aimed to determine the association between antiphospholipid antibodies (aPLs) and future atherosclerotic cardiovascular disease (ASCVD) in SLE. METHODS In total, 1573 SLE patients were recruited based on the Chinese SLE Treatment and Research group (CSTAR) registry. aPLs profile, including anticardiolipin antibodies (aCL) IgG/IgM, anti-β2 glycoprotein I antibodies (aβ2GPI) IgG/IgM, and lupus anticoagulant (LA), were measured in each center. Future ASCVD events were defined as new-onset myocardial infarction, stroke, artery revascularization, or cardiovascular death. RESULTS Among the 1573 SLE patients, 525 (33.4%) had positive aPLs. LA had the highest prevalence (324 [20.6%]), followed by aCL IgG (249 [15.8%]), aβ2GPI IgG (199 [12.7%]). 116 (7.37%) patients developed ASCVD during the mean follow-up of 4.51 ± 2.32 years and 92 patients were aPLs positive. In univariate Cox regression analysis, both aPLs (HR = 7.81, 95% CI 5.00-12.24, p < 0.001) and traditional risk factors of cardiovascular disease were associated with future ASCVD events. In multiple Cox regression analysis, aCL IgG (HR = 1.95, 95% CI 1.25-3.00, p = 0.003), aCL IgM (HR = 1.83, 95% CI 1.03-3.20, p = 0.039), and LA (HR = 5.13, 95% CI 3.23-8.20, p < 0.001) positivity remained associated with ASCVD; traditional risk factors for ASCVD, including smoking, gender, age and hypertension, also play an independent role in SLE patients. More importantly, Aspirin can reduce ASCVD risk in SLE patients with positive aPLs (HR = 0.57 95% CI, 0.25-0.93, P = 0.026). CONCLUSIONS SLE patients with positive aPLs, especially positive aCL IgG/IgM and LA, warrant more care and surveillance of future ASCVD events during follow-up. Aspirin may have a protective effect on future ASCVD.
Collapse
Grants
- 2022-PUMCH-A-008, PUMCH-A-A038 National High Level Hospital Clinical Research Funding
- 2022-PUMCH-A-008, PUMCH-A-A038 National High Level Hospital Clinical Research Funding
- 2022-PUMCH-A-008, PUMCH-A-A038 National High Level Hospital Clinical Research Funding
- 2022-PUMCH-A-008, PUMCH-A-A038 National High Level Hospital Clinical Research Funding
- 2022-PUMCH-A-008, PUMCH-A-A038 National High Level Hospital Clinical Research Funding
- No. Z 201100005520022,23, 25-27 Beijing Municipal Natural Science Foundation
- No. Z 201100005520022,23, 25-27 Beijing Municipal Natural Science Foundation
- No. Z 201100005520022,23, 25-27 Beijing Municipal Natural Science Foundation
- No. Z 201100005520022,23, 25-27 Beijing Municipal Natural Science Foundation
- No. Z 201100005520022,23, 25-27 Beijing Municipal Natural Science Foundation
- 2021-I2M-1-005 CAMS Innovation Fund for Medical Sciences (CIFMS)
- 2021-I2M-1-005 CAMS Innovation Fund for Medical Sciences (CIFMS)
- 2021-I2M-1-005 CAMS Innovation Fund for Medical Sciences (CIFMS)
- 2021-I2M-1-005 CAMS Innovation Fund for Medical Sciences (CIFMS)
- 2021-I2M-1-005 CAMS Innovation Fund for Medical Sciences (CIFMS)
- 2021YFC2501300 Chinese National Key Technology R&D Program, Ministry of Science and Technology
- Chinese National Key Technology R&D Program, Ministry of Science and Technology
Collapse
Affiliation(s)
- Can Huang
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College and Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, No. 1 Shuaifuyuan, Wangfujing Ave, Beijing, 100730, China
| | - Yufang Ding
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College and Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, No. 1 Shuaifuyuan, Wangfujing Ave, Beijing, 100730, China
| | - Zhen Chen
- Department of Rheumatology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Lijun Wu
- Department of Rheumatology and Immunology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China
| | - Wei Wei
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Cheng Zhao
- Department of Rheumatology and Immunology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Min Yang
- Department of Rheumatic and TCM Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shudian Lin
- Department of Rheumatology and Immunology, Hainan General Hospital, Haikou, 570311, China
| | - Qian Wang
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College and Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, No. 1 Shuaifuyuan, Wangfujing Ave, Beijing, 100730, China
| | - Xinping Tian
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College and Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, No. 1 Shuaifuyuan, Wangfujing Ave, Beijing, 100730, China
| | - Jiuliang Zhao
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College and Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, No. 1 Shuaifuyuan, Wangfujing Ave, Beijing, 100730, China.
| | - Mengtao Li
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College and Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, No. 1 Shuaifuyuan, Wangfujing Ave, Beijing, 100730, China.
| | - Xiaofeng Zeng
- Department of Rheumatology, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College and Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, No. 1 Shuaifuyuan, Wangfujing Ave, Beijing, 100730, China
| |
Collapse
|
4
|
Sener S, Sener YZ, Batu ED, Ertugrul I, Basaran O, Bilginer Y, Karagoz T, Ozen S. Evaluation of cardiac function using echocardiography in childhood-onset systemic lupus erythematosus patients treated with hydroxychloroquine. Expert Rev Cardiovasc Ther 2024:1-5. [PMID: 39699079 DOI: 10.1080/14779072.2024.2445800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/05/2024] [Accepted: 12/18/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND This study aimed to evaluate the effects of hydroxychloroquine on cardiac functions and left ventricular mass in patients with childhood-onset systemic lupus erythematosus (cSLE). RESEARCH DESIGN AND METHODS Fifty patients with cSLE undergoing treatment with hydroxychloroquine underwent echocardiographic evaluation. All patients exhibited negative disease activity markers and were clinically in remission. RESULTS The median duration of hydroxychloroquine exposure was 7.1 (5.2-9.5) years, with a median cumulative dose of 784.8 (509.5-3437.6) grams. No correlation was identified between the parameters of left ventricular ejection fraction, left ventricular mass index and geometry, and cumulative hydroxychloroquine dose (p = 0.245, p = 0.094, p = 0.146, respectively). Furthermore, no significant correlation was identified between the cumulative dose of hydroxychloroquine and diastolic cardiac parameters (all p > 0.05). A comparison of the patients who received a cumulative dose of hydroxychloroquine below the median dose (the low-dose group) with those who received a higher dose (the high-dose group) revealed no significant differences in the echocardiographic parameters (all p > 0.05). CONCLUSIONS The findings of this study indicate that chronic hydroxychloroquine use in patients with cSLE does not result in adverse changes in left ventricular mass or impairment of cardiac functions. However, these patients should undergo regular evaluation to monitor for the potential development of cardiotoxicity.
Collapse
Affiliation(s)
- Seher Sener
- Division of Pediatric Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Yusuf Ziya Sener
- Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Ezgi Deniz Batu
- Division of Pediatric Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Ilker Ertugrul
- Division of Pediatric Cardiology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Ozge Basaran
- Division of Pediatric Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Yelda Bilginer
- Division of Pediatric Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Tevfik Karagoz
- Division of Pediatric Cardiology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Seza Ozen
- Division of Pediatric Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
5
|
Corona-Meraz FI, Vázquez-Del Mercado M, Sandoval-García F, Robles-De Anda JA, Tovar-Cuevas AJ, Rosales-Gómez RC, Guzmán-Ornelas MO, González-Inostroz D, Peña-Nava M, Martín-Márquez BT. Biomarkers in Systemic Lupus Erythematosus along with Metabolic Syndrome. J Clin Med 2024; 13:1988. [PMID: 38610754 PMCID: PMC11012563 DOI: 10.3390/jcm13071988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Metabolic syndrome (MetS) is a group of physiological abnormalities characterized by obesity, insulin resistance (IR), and hypertriglyceridemia, which carry the risk of developing cardiovascular disease (CVD) and type 2 diabetes (T2D). Immune and metabolic alterations have been observed in MetS and are associated with autoimmune development. Systemic lupus erythematosus (SLE) is an autoimmune disease caused by a complex interaction of environmental, hormonal, and genetic factors and hyperactivation of immune cells. Patients with SLE have a high prevalence of MetS, in which elevated CVD is observed. Among the efforts of multidisciplinary healthcare teams to make an early diagnosis, a wide variety of factors have been considered and associated with the generation of biomarkers. This review aimed to elucidate some primary biomarkers and propose a set of assessments to improve the projection of the diagnosis and evolution of patients. These biomarkers include metabolic profiles, cytokines, cardiovascular tests, and microRNAs (miRs), which have been observed to be dysregulated in these patients and associated with outcomes.
Collapse
Affiliation(s)
- Fernanda Isadora Corona-Meraz
- Multidisciplinary Health Research Center, Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara 45425, Jalisco, Mexico; (A.-J.T.-C.); (R.-C.R.-G.); (M.-O.G.-O.)
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
| | - Mónica Vázquez-Del Mercado
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
- Rheumatology Service, Internal Medicine Division, Civil Hospital of Guadalajara “Dr. Juan I. Menchaca”, Guadalajara 44340, Jalisco, Mexico
- Academic Group UDG-CA-703, “Immunology and Rheumatology”, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Flavio Sandoval-García
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
- Academic Group UDG-CA-703, “Immunology and Rheumatology”, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Jesus-Aureliano Robles-De Anda
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
| | - Alvaro-Jovanny Tovar-Cuevas
- Multidisciplinary Health Research Center, Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara 45425, Jalisco, Mexico; (A.-J.T.-C.); (R.-C.R.-G.); (M.-O.G.-O.)
| | - Roberto-Carlos Rosales-Gómez
- Multidisciplinary Health Research Center, Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara 45425, Jalisco, Mexico; (A.-J.T.-C.); (R.-C.R.-G.); (M.-O.G.-O.)
| | - Milton-Omar Guzmán-Ornelas
- Multidisciplinary Health Research Center, Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara 45425, Jalisco, Mexico; (A.-J.T.-C.); (R.-C.R.-G.); (M.-O.G.-O.)
| | - Daniel González-Inostroz
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
| | - Miguel Peña-Nava
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
| | - Beatriz-Teresita Martín-Márquez
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
- Academic Group UDG-CA-703, “Immunology and Rheumatology”, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
6
|
Garg S, Chewning B, Hutson P, Astor BC, Bartels CM. Reference Range of Hydroxychloroquine Blood Levels That Can Reduce Odds of Active Lupus and Prevent Flares. Arthritis Care Res (Hoboken) 2024; 76:241-250. [PMID: 37667434 PMCID: PMC11078155 DOI: 10.1002/acr.25228] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/04/2023] [Accepted: 08/31/2023] [Indexed: 09/06/2023]
Abstract
OBJECTIVE Recent data show that lower hydroxychloroquine (HCQ) doses are associated with a two- to six-fold higher risk of lupus flares. Thus, establishing an effective reference range of HCQ blood levels with upper and lower bounds for efficacy may support individualizing HCQ dosing to prevent flares. METHODS HCQ levels in whole blood and Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) were measured during the baseline visit and again during a standard of care routine follow-up visit. Active cross-sectional lupus at baseline was defined as SLEDAI ≥6; a within subject flare was defined as a subsequent three-point increase in SLEDAI with clinical symptoms requiring therapy change. We examined associations between active lupus and HCQ blood levels at baseline and flares and HCQ levels during 6 to 12-month routine lupus follow-up visits using mixed regression analysis. RESULTS Among 158 baseline patient visits, 19% had active lupus. Odds of active lupus were 71% lower in patients with levels within a 750 to 1,200 ng/mL range (adjusted odds ratio 0.29, 95% confidence interval 0.08-0.96). Using convenience sampling strategy during a pandemic, we longitudinally followed 42 patients. Among those patients, 17% flared during their follow-up visit. Maintaining HCQ levels within 750 to 1,200 ng/mL reduced the odds of a flare by 26% over a nine-month median follow-up. CONCLUSION An effective reference range of HCQ blood levels, 750 to 1,200 ng/mL, was associated with 71% lower odds of active lupus, and maintaining levels within this range reduced odds of flares by 26%. These findings could guide clinicians to individualize HCQ doses to maintain HCQ levels within this range to maximize efficacy.
Collapse
Affiliation(s)
- Shivani Garg
- University of Wisconsin School of Medicine and Public Health, Department of Medicine, Division of Rheumatology, Madison, WI, USA
| | - Betty Chewning
- University of Wisconsin School of Pharmacy, Madison, WI, USA
| | - Paul Hutson
- University of Wisconsin School of Pharmacy, Madison, WI, USA
| | - Brad C. Astor
- University of Wisconsin School of Medicine and Public Health, Department of Medicine, Division of Nephrology, Madison, WI, USA
- University of Wisconsin School of Medicine and Public Health, Department of Population Health Sciences, Madison, WI, USA
| | - Christie M. Bartels
- University of Wisconsin School of Medicine and Public Health, Department of Medicine, Division of Rheumatology, Madison, WI, USA
| |
Collapse
|
7
|
Waksman R, Merdler I, Case BC, Waksman O, Porto I. Targeting inflammation in atherosclerosis: overview, strategy and directions. EUROINTERVENTION 2024; 20:32-44. [PMID: 38165117 PMCID: PMC10756224 DOI: 10.4244/eij-d-23-00606] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/06/2023] [Indexed: 01/03/2024]
Abstract
Atherosclerosis is a chronic condition characterised by the build-up of plaque in the inner lining of the blood vessels and it is the main underlying cause of cardiovascular disease. The development of atherosclerosis is associated with the accumulation of cholesterol and inflammation. Although effective therapies exist to lower low-density lipoprotein cholesterol (LDL-C) levels, some patients still experience cardiovascular events due to persistent inflammation, known as residual inflammatory risk (RIR). Researchers have conducted laboratory and animal studies to investigate the measurement and targeting of the inflammatory cascade associated with atherosclerosis, which have yielded promising results. In addition to guideline-directed lifestyle modifications and optimal medical therapy focusing on reducing LDL-C levels, pharmacological interventions targeting inflammation may provide further assistance in preventing future cardiac events. This review aims to explain the mechanisms of inflammation in atherosclerosis, identifies potential biomarkers, discusses available therapeutic options and their strengths and limitations, highlights future advancements, and summarises notable clinical studies. Finally, an evaluation and management algorithm for addressing RIR is presented.
Collapse
Affiliation(s)
- Ron Waksman
- MedStar Heart & Vascular Institute, MedStar Washington Hospital Center, MedStar Georgetown University Hospital, Washington, D.C., USA
| | - Ilan Merdler
- MedStar Heart & Vascular Institute, MedStar Washington Hospital Center, MedStar Georgetown University Hospital, Washington, D.C., USA
| | - Brian C Case
- MedStar Heart & Vascular Institute, MedStar Washington Hospital Center, MedStar Georgetown University Hospital, Washington, D.C., USA
| | - Ori Waksman
- MedStar Heart & Vascular Institute, MedStar Washington Hospital Center, MedStar Georgetown University Hospital, Washington, D.C., USA
| | - Italo Porto
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino - Italian IRCCS Cardiology Network, Genoa, Italy
| |
Collapse
|
8
|
Tobin R, Patel N, Tobb K, Weber B, Mehta PK, Isiadinso I. Atherosclerosis in Systemic Lupus Erythematosus. Curr Atheroscler Rep 2023; 25:819-827. [PMID: 37768411 DOI: 10.1007/s11883-023-01149-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2023] [Indexed: 09/29/2023]
Abstract
PURPOSE OF THE REVIEW Systemic lupus erythematosus (SLE) patients are at increased risk of cardiovascular disease (CVD) compared to the general population, despite most patients being young females, who are not classically considered to be at high risk for cardiovascular disease using traditional risk assessment tools. The purpose of this review is to discuss the pathophysiology of atherosclerosis in SLE and raise awareness of the relationship between SLE and CVD. RECENT FINDINGS The increased risk of CVD in SLE patients is multifactorial, due to proatherogenic lipid profiles, immune dysregulation and inflammation, side effects of lupus treatment, and microvascular dysfunction. Conventional CV risk models often underperform in the identification of SLE patients at high risk of atherosclerosis. The use of non-invasive imaging serves as a strategy to identify patients with evidence of subclinical CVD and in the evaluation of symptomatic patients. Identification of subclinical atherosclerosis allows for aggressive management of CV risk factors. SLE patients experience an increased risk of atherosclerotic CVD, which is not solely explained by traditional CV risk factors. It is imperative that clinicians are aware of this association to implement prompt detection and treatment of atherosclerotic CVD in SLE patients.
Collapse
Affiliation(s)
- Rachel Tobin
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| | - Nidhi Patel
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Kardie Tobb
- Cone Health Medical Group, Greensboro, NC, USA
| | - Brittany Weber
- Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Puja K Mehta
- Division of Cardiology, Department of Medicine, Center for Heart Disease Prevention, Emory University School of Medicine, Atlanta, GA, USA
| | - Ijeoma Isiadinso
- Division of Cardiology, Department of Medicine, Center for Heart Disease Prevention, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
9
|
Jimenez AL, Valle A, Mustehsan MH, Wang S, Law J, Guerrero MS, Mowrey WB, Horton DB, Briceno D, Broder A. Association of Hydroxychloroquine Dose With Adverse Cardiac Events in Patients With Systemic Lupus Erythematosus. Arthritis Care Res (Hoboken) 2023; 75:1673-1680. [PMID: 36331104 PMCID: PMC10156898 DOI: 10.1002/acr.25052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 09/27/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE To determine whether hydroxychloroquine (HCQ) dose is associated with adverse cardiac outcomes in patients with systemic lupus erythematosus (SLE). METHODS Patients with SLE taking HCQ and with ≥1 echocardiogram followed at a tertiary care center in the Bronx, New York between 2005 and 2021 were included. The HCQ weight-based dose at the HCQ start date was the main exposure of interest. The outcome was incident all-cause heart failure with reduced ejection fraction (HFrEF), life-threatening arrhythmia, or cardiac death. We used Fine-Gray regression models with death as a competing event to study the association of HCQ dose with the outcome. Due to a significant interaction between smoking and HCQ exposure, models were stratified by smoking status. Propensity score analysis was performed as a secondary analysis. RESULTS Of 294 patients, 37 (13%) developed the outcome over a median follow-up time of 7.9 years (interquartile range [IQR] 4.2-12.3 years). In nonsmokers (n = 226), multivariable analysis adjusted for age, body mass index, hypertension, chronic kidney disease, diabetes mellitus, and thromboembolism showed that higher HCQ weight-based doses were not associated with an increased risk of the outcome (subdistribution hazard ratio [HR] 0.62 [IQR 0.41-0.92], P = 0.02). Similarly, higher baseline HCQ doses were not associated with a higher risk of the outcome among smokers (n = 68) (subdistribution HR 0.85 [IQR 0.53-1.34] per mg/kg, P = 0.48). Propensity score analysis showed comparable results. CONCLUSION Higher HCQ doses were not associated with an increased risk of HFrEF, life-threatening arrhythmia, or cardiac death among patients with SLE and may decrease the risk among nonsmokers.
Collapse
Affiliation(s)
| | - Ana Valle
- Albert Einstein College of Medicine/Montefiore Medical Center, the Bronx, New York
| | | | - Shudan Wang
- Albert Einstein College of Medicine/Montefiore Medical Center, the Bronx, New York
| | - Jammie Law
- Albert Einstein College of Medicine/Montefiore Medical Center, the Bronx, New York
| | | | - Wenzhu B Mowrey
- Albert Einstein College of Medicine/Montefiore Medical Center, the Bronx, New York
| | - Daniel B Horton
- Rutgers Center for Pharmacoepidemiology and Treatment Science and Institute for Health, Health Care Policy, and Aging Research, New Brunswick, New Jersey
| | | | - Anna Broder
- Hackensack University Hospital, Hackensack, New Jersey
| |
Collapse
|
10
|
Athanassiou P, Athanassiou L. Current Treatment Approach, Emerging Therapies and New Horizons in Systemic Lupus Erythematosus. Life (Basel) 2023; 13:1496. [PMID: 37511872 PMCID: PMC10381582 DOI: 10.3390/life13071496] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 06/18/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Systemic lupus erythematosus (SLE), the prototype of systemic autoimmune diseases is characterized by extreme heterogeneity with a variable clinical course. Renal involvement may be observed and affects the outcome. Hydroxychloroquine should be administered to every lupus patient irrespective of organ involvement. Conventional immunosuppressive therapy includes corticosteroids, methotrexate, cyclophosphamide, mycophenolate mofetil, azathioprine, cyclosporine and tacrolimus. However, despite conventional immunosuppressive treatment, flares occur and broad immunosuppression is accompanied by multiple side effects. Flare occurrence, target organ involvement, side effects of broad immunosuppression and increased knowledge of the pathogenetic mechanisms involved in SLE pathogenesis as well as the availability of biologic agents has led to the application of biologic agents in SLE management. Biologic agents targeting various pathogenetic paths have been applied. B cell targeting agents have been used successfully. Belimumab, a B cell targeting agent, has been approved for the treatment of SLE. Rituximab, an anti-CD20 targeting agent is also used in SLE. Anifrolumab, an interferon I receptor-targeting agent has beneficial effects on SLE. In conclusion, biologic treatment is applied in SLE and should be further evaluated with the aim of a good treatment response and a significant improvement in quality of life.
Collapse
Affiliation(s)
| | - Lambros Athanassiou
- Department of Rheumatology, Asclepeion Hospital, Voula, GR16673 Athens, Greece
| |
Collapse
|
11
|
Garg S, Ferguson S, Chewning B, Gomez S, Keevil J, Bartels C. Clarifying misbeliefs about hydroxychloroquine (HCQ): developing the HCQ benefits versus harm decision aid (HCQ-SAFE) per low health literacy standards. Lupus Sci Med 2023; 10:e000935. [PMID: 37500292 PMCID: PMC10387621 DOI: 10.1136/lupus-2023-000935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 07/17/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND Up to 83% of patients with SLE stop taking hydroxychloroquine (HCQ) within the first year due to knowledge gaps regarding the survival benefits of HCQ versus inflated fears of rare toxicity. Thus, there is a need for a shared decision-making tool that highlights HCQ's significant benefits versus rare harms to improve patients' understanding and align treatments with their values. The objective of this study was to describe development and piloting of a decision aid (HCQ-SAFE) to facilitate HCQ adherence, and safe, effective use by engaging patients in therapeutic decision-making. METHODS HCQ-SAFE was developed via a collaborative process involving patients, clinicians, implementation scientists and health literacy experts. The initial prototype was informed by Agency for Healthcare Research and Quality (AHRQ) low literacy principles and key themes about HCQ use from six prior patient and clinician focus groups, with iterative expert and stakeholder feedback to deliver a final prototype. We implemented HCQ-SAFE in four clinics to examine usability and feasibility on Likert scales (0-7) and net promoter score (0%-100%). RESULTS The final HCQ-SAFE shared decision-making laminated tool organises data using pictograms showing how HCQ use reduces risk of organ damage, early death and blood clots versus low risk of eye toxicity.HCQ-SAFE was reviewed in all eligible patient visits (n=40) across four clinics on an average of ~8 min, including 25% non-English-speaking patients. All patients scored 100% on the knowledge post-test; no decisional conflicts were noted after using HCQ-SAFE. HCQ-SAFE garnered high clinician and patient satisfaction with 100% likelihood to recommend to peers. CONCLUSIONS HCQ-SAFE is a stakeholder-informed feasible shared decision-making tool that enhances communication and can potentially improve knowledge, clarify misbeliefs and engage patients in treatment decisions, including those with limited English proficiency.
Collapse
Affiliation(s)
- Shivani Garg
- Department of Medicine, Division of Rheumatology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Sancia Ferguson
- Department of Medicine, Division of Rheumatology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Betty Chewning
- Department of Pharmacy, School of Pharmacy, Madison, Wisconsin, USA
| | - Shelby Gomez
- Department of Pharmacy, School of Pharmacy, Madison, Wisconsin, USA
| | | | - Christie Bartels
- Department of Medicine, Division of Rheumatology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
12
|
Nagy N, Papp G, Gáspár-Kiss E, Diószegi Á, Tarr T. Changes in Clinical Manifestations and Course of Systemic Lupus Erythematosus and Secondary Antiphospholipid Syndrome over Three Decades. Biomedicines 2023; 11:biomedicines11041218. [PMID: 37189836 DOI: 10.3390/biomedicines11041218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/30/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is often associated with antiphospholipid syndrome (APS), which potentially results in a more severe disease course and reduced life expectancy. Since the therapeutic guidelines have been refined in the last 15 years, we assumed that the diseases course has become more favorable. In order to shed light on these achievements, we compared the data of SLE patients diagnosed before and since 2004. In our retrospective study, we assessed a wide spectrum of clinical and laboratory data of 554 SLE patients who received regular follow-up care and therapy at our autoimmune center. Among these patients, 247 had antiphospholipid antibodies (APAs) without clinical signs of APS, and 113 had definitive APS. In the APS group, among patients diagnosed since 2004, deep vein thrombosis (p = 0.049) and lupus anticoagulant positivity (p = 0.045) were more frequent, while acute myocardial infarction was less frequent (p = 0.021) compared with patients diagnosed before 2004. Among the APA positive patients without definitive APS, anti-cardiolipin antibody positivity (p = 0.024) and development of chronic renal failure (p = 0.005) decreased in patients diagnosed since 2004. Our study demonstrates that the disease course has changed in recent years; however, in the presence of APS, we have to expect repeated thrombotic events despite adequate anticoagulant therapy.
Collapse
Affiliation(s)
- Nikolett Nagy
- Division of Clinical Immunology, Institute of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Gábor Papp
- Division of Clinical Immunology, Institute of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Eszter Gáspár-Kiss
- Division of Clinical Immunology, Institute of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Ágnes Diószegi
- Division of Metabolic Disorders, Institute of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Tünde Tarr
- Division of Clinical Immunology, Institute of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| |
Collapse
|
13
|
Ivey AD, Matthew Fagan B, Murthy P, Lotze MT, Zeh HJ, Hazlehurst LA, Geldenhuys WJ, Boone BA. Chloroquine reduces neutrophil extracellular trap (NET) formation through inhibition of peptidyl arginine deiminase 4 (PAD4). Clin Exp Immunol 2023; 211:239-247. [PMID: 36655514 PMCID: PMC10038322 DOI: 10.1093/cei/uxad005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 12/03/2022] [Accepted: 01/18/2023] [Indexed: 01/20/2023] Open
Abstract
Neutrophil extracellular traps (NETs) occur when chromatin is decondensed and extruded from the cell, generating a web-like structure. NETs have been implicated in the pathogenesis of several sterile disease states and thus are a potential therapeutic target. Various pathways have been shown to induce NETs, including autophagy, with several key enzymes being activated like peptidyl arginine deiminase 4 (PAD4), an enzyme responsible for citrullination of histones, allowing for DNA unwinding and subsequent release from the cell. Pre-clinical studies have already demonstrated that chloroquine (CQ) and hydroxychloroquine (HCQ) are able to reduce NETs and slow disease progression. The exact mechanism as to how these drugs reduce NETs has yet to be elucidated. CQ and HCQ decrease NET formation from various NET activators, independent of their autophagy inhibitory function. CQ and HCQ were found to inhibit PAD4 exclusively, in a dose-dependent manner, confirmed with reduced CitH3+ NETs after CQ or HCQ treatment. Circulating CitH3 levels were reduced in pancreatic cancer patients after HCQ treatment. In silico screening of PAD4 protein structure identified a likely binding site interaction at Arg639 for CQ and Trp347, Ser468, and Glu580 for HCQ. SPR analysis confirmed the binding of HCQ and CQ with PAD4 with KD values of 54.1 µM (CQ) and 88.1 µM (HCQ). This data provide evidence of direct PAD4 inhibition as a mechanism for CQ/HCQ inhibition of NETs. We propose that these drugs likely reduce NET formation through multiple mechanisms; the previously established TLR9 and autophagy inhibitory mechanism and the novel PAD4 inhibitory mechanism.
Collapse
Affiliation(s)
- Abby D Ivey
- Cancer Cell Biology, West Virginia University, Morgantown, WV, USA
| | - B Matthew Fagan
- Department of Surgery, West Virginia University, Morgantown, WV, USA
| | - Pranav Murthy
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Herbert J Zeh
- Department of Surgery, UT Southwestern, Dallas, TX, USA
| | - Lori A Hazlehurst
- Cancer Cell Biology, West Virginia University, Morgantown, WV, USA
- Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USA
| | - Werner J Geldenhuys
- Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USA
- Neuroscience, West Virginia University, Morgantown, WV, USA
| | - Brian A Boone
- Cancer Cell Biology, West Virginia University, Morgantown, WV, USA
- Department of Surgery, West Virginia University, Morgantown, WV, USA
- Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
14
|
Nutritional Approaches to Modulate Cardiovascular Disease Risk in Systemic Lupus Erythematosus: A Literature Review. Nutrients 2023; 15:nu15041036. [PMID: 36839394 PMCID: PMC9958972 DOI: 10.3390/nu15041036] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic pathology characterized by a bimodal mortality pattern attributed to clinical disease activity and cardiovascular disease (CVD). A complex interaction between traditional CVD risk factors such as obesity, dyslipidemia, smoking, insulin resistance, metabolic syndrome, and hypertension, as well as the presence of non-traditional CVD risk factors such as hyperhomocysteinemia, pro-inflammatory cytokines, and C-reactive protein levels, has been suggested as a cause of the high prevalence of CVD in SLE patients. On the other hand, environmental factors, such as nutritional status, could influence the disease's prognosis; several nutrients have immunomodulators, antioxidants, and anti-cardiometabolic risk properties which could reduce SLE severity and organ damage by decreasing the development of traditional and non-traditional CVD risk factors. Therefore, this critical literature review discusses the therapeutic potential of nutritional approaches that could modulate the development of the main comorbidities related to CVD risk in SLE patients.
Collapse
|
15
|
Cen Y, Xiong Y, Qin R, Tao H, Yang Q, Pan X. Anti-malarial artesunate ameliorates atherosclerosis by modulating arterial inflammatory responses via inhibiting the NF-κB-NLRP3 inflammasome pathway. Front Pharmacol 2023; 14:1123700. [PMID: 36817159 PMCID: PMC9931906 DOI: 10.3389/fphar.2023.1123700] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction: Chronic inflammation plays a critical role in the pathogenesis of atherosclerosis (AS), and involves a complex interplay between blood components, macrophages, and arterial wall. Therefore, it is valuable in the development of targeted therapies to treat AS. Methods: AS rat model was induced by atherogenic diet plus with lipopolysaccharide (LPS) and then treated by anti-malarial artesunate (Art), a succinate derivative of artemisinin. The arterial morphology was observed after Oil red O, hematoxylin-eosin, and Masson's staining. The arterial protein level was detected by immunohistochemistry or immunofluorescence. The expression level of mRNA was determined by PCR array or real-time PCR. Results: Herein, we showed that Art possessed a dose-dependently protective effect on AS rats. In detail, Art showed a comparable inhibitory effect on arterial plaque and serum lipids compared to those of rosuvastatin (RS), and further showed a better inhibition on arterial lipid deposition and arterial remodeling comprised of arterial wall thicken and vascular collagen deposition, than those of RS. The improvement of Art on AS rats was related to inhibit arterial macrophage recruitment, and inhibit nuclear factor κB (NF-κB)-related excessive arterial inflammatory responses. Critically, Art showed significant inhibition on the NLRP3 inflammasome activation in both arterial wall and arterial macrophages, by down-regulating the expression of NOD-like receptor thermal protein domain associated protein 3 (NLRP3) and apoptosis associated speckle-like protein containing CARD (ASC), leading to less production of the NLRP3 inflammasome-derived caspase-1, interleukin-1β (IL-1β), IL-18, and subsequent transforming growth factor β1 (TGF-β1) in AS rats. Conclusion: We propose that Art is an anti-AS agent acts through modulating the arterial inflammatory responses via inhibiting the NF-κB - NLRP3 inflammasome pathway.
Collapse
|
16
|
Mangoni AA, Sotgia S, Zinellu A, Carru C, Pintus G, Damiani G, Erre GL, Tommasi S. Methotrexate and cardiovascular prevention: an appraisal of the current evidence. Ther Adv Cardiovasc Dis 2023; 17:17539447231215213. [PMID: 38115784 PMCID: PMC10732001 DOI: 10.1177/17539447231215213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/02/2023] [Indexed: 12/21/2023] Open
Abstract
New evidence continues to accumulate regarding a significant association between excessive inflammation and dysregulated immunity (local and systemic) and the risk of cardiovascular events in different patient cohorts. Whilst research has sought to identify novel atheroprotective therapies targeting inflammation and immunity, several marketed drugs for rheumatological conditions may serve a similar purpose. One such drug, methotrexate, has been used since 1948 for treating cancer and, more recently, for a wide range of dysimmune conditions. Over the last 30 years, epidemiological and experimental studies have shown that methotrexate is independently associated with a reduced risk of cardiovascular disease, particularly in rheumatological patients, and exerts several beneficial effects on vascular homeostasis and blood pressure control. This review article discusses the current challenges with managing cardiovascular risk and the new frontiers offered by drug discovery and drug repurposing targeting inflammation and immunity with a focus on methotrexate. Specifically, the article critically appraises the results of observational, cross-sectional and intervention studies investigating the effects of methotrexate on overall cardiovascular risk and individual risk factors. It also discusses the putative molecular mechanisms underpinning the atheroprotective effects of methotrexate and the practical advantages of using methotrexate in cardiovascular prevention, and highlights future research directions in this area.
Collapse
Affiliation(s)
- Arduino A. Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
- Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Bedford Park, SA 5042, Australia
| | - Salvatore Sotgia
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy; Quality Control Unit, University Hospital (AOUSS), Sassari, Italy
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy; Quality Control Unit, University Hospital (AOUSS), Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Quality Control Unit, University Hospital (AOUSS), Sassari, Italy
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy; Quality Control Unit, University Hospital (AOUSS), Sassari, Italy
| | - Giovanni Damiani
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
- Italian Centre of Precision Medicine and Chronic Inflammation, Milan, Italy
| | - Gian Luca Erre
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University Hospital (AOUSS) and University of Sassari, Sassari, Italy
| | - Sara Tommasi
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, SA, Australia
| |
Collapse
|
17
|
Cai T, Zhao J, Yang Y, Jiang Y, Zhang JA. Hydroxychloroquine use reduces mortality risk in systemic lupus erythematosus: A systematic review and meta-analysis of cohort studies. Lupus 2022; 31:1714-1725. [DOI: 10.1177/09612033221129774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Objective Hydroxychloroquine (HCQ) is widely used in patients with systemic lupus erythematosus (SLE), but its effects on the mortality have not reached a definite conclusion. In this systematic review and meta-analysis, we aimed to assess whether HCQ use could reduce the risk of mortality in SLE patients. Methods PubMed, Embase, Web of Science, and Cochrane database were searched from inception to April 17, 2022 without language restrictions to explore the relationship between HCQ use and SLE mortality. The relative risk (HR) was pooled using the STATA software. Results A total of 21 studies with a pooled patient population of 26,037 were included in the study, including 14 studies on the association between HCQ alone and mortality risk and seven studies on the association between HCQ/chloroquine (CQ) and mortality risk. The pooled findings suggested that HCQ significantly reduced the overall mortality risk of SLE (pooled HR 0.46, 95% CI 0.38–0.57, p < 0.001). In subgroup analysis of SLE complications, HCQ use also decreased the risk of death in SLE patients with renal (HR=0.43, 95% CI 0.26–0.70, p = 0.001) and cardiopulmonary involvement (HR=0.37, 95% CI= 0.25–0.54, p < 0.001). In addition, HCQ use was also protective against the risk of mortality in SLE patients in different regions, such as Asia (HR=0.46, 95% CI=0.33–0.64, p < 0.001), Europe (HR= 0.40, 95% CI = 0.22–0.71, p = 0.002), and America (HR=0.52, 95% CI= 0.42–0.64, p < 0.001). Conclusion Our data suggested that HCQ use was associated with a reduced risk of mortality in patients with SLE.
Collapse
Affiliation(s)
- Tiantian Cai
- Department of Endocrinology & Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, People’s Republic of China
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jing Zhao
- Department of Endocrinology & Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, People’s Republic of China
| | - Yanping Yang
- Department of Endocrinology & Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, People’s Republic of China
| | - Yanfei Jiang
- Department of Endocrinology & Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, People’s Republic of China
| | - Jin-an Zhang
- Department of Endocrinology & Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, People’s Republic of China
| |
Collapse
|
18
|
Huang H, Ma X, Xu L, Wang X, Shi D, Zhao F, Zhang Y. Spontaneous coronary artery dissection and atherosclerosis in a young man with systemic lupus erythematosus: A case report and literature review. Front Cardiovasc Med 2022; 9:951188. [PMID: 36035908 PMCID: PMC9402264 DOI: 10.3389/fcvm.2022.951188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/20/2022] [Indexed: 11/26/2022] Open
Abstract
Background Spontaneous coronary artery dissection (SCAD) is a rare coronary artery disease that frequently occurs in young, female patients without risk factors, and conservative treatment is often recommended for its management. The patient reported here is a male patient with systemic lupus erythematosus (SLE). Case summary We described a 28-year-old man with SLE who presented with acute ST-segment elevation myocardial infarction (STEMI), and was diagnosed with SCAD through a long dissection of the left anterior descending branch (LAD) by coronary angiography. The patient was treated with percutaneous coronary intervention (PCI) with stent implantation. Ten years later, he developed in-stent stenosis and other coronary atherosclerosis and was retreated with PCIs. Based on this case and according to the literature review, the existing treatment and prognosis of SLE with spontaneous coronary artery dissection and atherosclerosis are discussed. Conclusion Cardiovascular complications should be considered in patients with systemic lupus erythematosus, although they may not initially be atherosclerotic diseases. Attention should be paid to distinguish spontaneous coronary dissection in order to minimize missed or delayed diagnoses and take appropriate managements, as well as the development of atherosclerosis in SLE patients, and timely intervention has a better prognosis.
Collapse
Affiliation(s)
- Hongbo Huang
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaojuan Ma
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Linjie Xu
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xin Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dazhuo Shi
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Dazhuo Shi
| | - Fuhai Zhao
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Fuhai Zhao
| | - Ying Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Ying Zhang
| |
Collapse
|
19
|
Rua-Figueroa Fernández de Larrinoa Í, Lozano MJC, Fernández-Cid CM, Cobo T, Salman Monte TC, Freire González M, Hidalgo Bermejo FJ, Román Gutiérrez CS, Cortés-Hernández J. Preventing organ damage in systemic lupus erythematosus: the impact of early biological treatment. Expert Opin Biol Ther 2022; 22:821-829. [PMID: 35815355 DOI: 10.1080/14712598.2022.2096406] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION One of the most important aims in the management of systemic lupus erythematosus (SLE) is to avoid or delay the accumulation of organ damage. The first five years after diagnosis are crucial for prognosis. AREAS COVERED This manuscript reviews available data on organ damage accrual in SLE and early therapeutic intervention as a possible strategy to prevent its long-term accrual. EXPERT OPINION Organ damage can be minimized by controlling disease activity and risk of flares, reducing the dose of glucocorticoids, and ensuring a proper therapeutic intervention with an early introduction of the right therapies. The current standard treatment cannot provide clinical remission in all patients with SLE. Therefore, there is a clinical need for introducing new therapeutic strategies able to achieve the main therapeutic objectives. The addition of biologic and other therapeutic agents to the standard of care is effective for controlling disease activity and for preventing severe flares, enabling a reduced use of glucocorticoids, and presumably reducing organ damage progression. Considering its efficacy and safety, early inclusion of biologic agents in the first lines of the treatment algorithm, at least in certain patients, could be considered as an innovative treatment approach to decrease disease burden in SLE patients.
Collapse
Affiliation(s)
| | | | | | - Tatiana Cobo
- Rheumatology Department, Hospital Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Tarek C Salman Monte
- Rheumatology Department, Hospital Universitario Infanta Sofía, San Sebastián de los Reyes, Spain
| | | | | | | | | |
Collapse
|
20
|
Infante M, Padilla N, Alejandro R, Caprio M, Della-Morte D, Fabbri A, Ricordi C. Diabetes-Modifying Antirheumatic Drugs: The Roles of DMARDs as Glucose-Lowering Agents. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:571. [PMID: 35629988 PMCID: PMC9143119 DOI: 10.3390/medicina58050571] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/14/2022] [Accepted: 04/18/2022] [Indexed: 02/06/2023]
Abstract
Systemic inflammation represents a shared pathophysiological mechanism which underlies the frequent clinical associations among chronic inflammatory rheumatic diseases (CIRDs), insulin resistance, type 2 diabetes (T2D), and chronic diabetes complications, including cardiovascular disease. Therefore, targeted anti-inflammatory therapies are attractive and highly desirable interventions to concomitantly reduce rheumatic disease activity and to improve glucose control in patients with CIRDs and comorbid T2D. Therapeutic approaches targeting inflammation may also play a role in the prevention of prediabetes and diabetes in patients with CIRDs, particularly in those with traditional risk factors and/or on high-dose corticosteroid therapy. Recently, several studies have shown that different disease-modifying antirheumatic drugs (DMARDs) used for the treatment of CIRDs exert antihyperglycemic properties by virtue of their anti-inflammatory, insulin-sensitizing, and/or insulinotropic effects. In this view, DMARDs are promising drug candidates that may potentially reduce rheumatic disease activity, ameliorate glucose control, and at the same time, prevent the development of diabetes-associated cardiovascular complications and metabolic dysfunctions. In light of their substantial antidiabetic actions, some DMARDs (such as hydroxychloroquine and anakinra) could be alternatively termed "diabetes-modifying antirheumatic drugs", since they may be repurposed for co-treatment of rheumatic diseases and comorbid T2D. However, there is a need for future randomized controlled trials to confirm the beneficial metabolic and cardiovascular effects as well as the safety profile of distinct DMARDs in the long term. This narrative review aims to discuss the current knowledge about the mechanisms behind the antihyperglycemic properties exerted by a variety of DMARDs (including synthetic and biologic DMARDs) and the potential use of these agents as antidiabetic medications in clinical settings.
Collapse
Affiliation(s)
- Marco Infante
- Clinical Cell Transplant Program (CCTP), Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL 33136, USA; (R.A.); (C.R.)
- Department of Systems Medicine, Diabetes Research Institute Federation (DRIF), University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy;
- Section of Endocrinology, UniCamillus, Saint Camillus International University of Health Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Via Cola di Rienzo 28, 00192 Rome, Italy
| | - Nathalia Padilla
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Colonia Centroamérica L-823, Managua 14048, Nicaragua;
| | - Rodolfo Alejandro
- Clinical Cell Transplant Program (CCTP), Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL 33136, USA; (R.A.); (C.R.)
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, Via di Val Cannuta 247, 00166 Rome, Italy;
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Via di Val Cannuta 247, 00166 Rome, Italy;
| | - David Della-Morte
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Via di Val Cannuta 247, 00166 Rome, Italy;
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
- Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami Miller School of Medicine, 1120 NW 14th St., Miami, FL 33136, USA
| | - Andrea Fabbri
- Department of Systems Medicine, Diabetes Research Institute Federation (DRIF), University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy;
| | - Camillo Ricordi
- Clinical Cell Transplant Program (CCTP), Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL 33136, USA; (R.A.); (C.R.)
| |
Collapse
|
21
|
Boujelbane F, Nasr K, Sadaoui H, Bui HM, Gantri F, Mzoughi N. Decomposition mechanism of hydroxychloroquine in aqueous solution by gamma irradiation. CHEMICAL PAPERS 2022; 76:1777-1787. [PMID: 35106020 PMCID: PMC8794614 DOI: 10.1007/s11696-021-01969-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/02/2021] [Indexed: 01/18/2023]
Abstract
One of the advanced oxidative processes is gamma irradiation, an efficient technique for removing pesticides and pharmaceutical products. Radiolytic degradation leads to free radical’s formation, which facilitates molecular lesion and breaks the chemical bonds. The use of pharmaceutical compounds, such as hydroxychloroquine (HCQ), is increasing nowadays due to the Covid 19 pandemic situation. This study focused on gamma radiation-induced degradation of HCQ in aqueous solution. The degradation was monitored by High-Performance Liquid Chromatography (HPLC) using an Eclipse XDB-C18 column (150 × 3.0 mm, 3.5 µm) and a mobile phase composed of 94% water (phosphate buffer at pH = 3.6) and 6% acetonitrile, with a DAD detection at λ = 343 nm. The effect of different gamma radiation doses (from 0.05 to 3 kGy) was investigated. Chromatographic analysis shows that 1 kGy dose is effective to degrade completely HCQ at 20 ppm and following a first-pseudo-kinetic order with a dose constant corresponding to 4.2 kGy−1. A comparison was done between gamma degradation and other methods. LC-QToF-MS/MS identified the intermediate products, and their kinetic constants were determined. A mechanism pathway was proposed for HCQ degradation under gamma irradiation.
Collapse
Affiliation(s)
- F. Boujelbane
- Research Laboratory on Matter and Energy for Nuclear Science Development, LR16CNSTN02, CNSTN, Sidi Thabet Technopark, 2020 Tunis, Tunisia
- Radiochemistry Laboratory, CNSTN, Sidi Thabet Technopark, 2020 Tunis, Tunisia
| | - K. Nasr
- Research Laboratory on Matter and Energy for Nuclear Science Development, LR16CNSTN02, CNSTN, Sidi Thabet Technopark, 2020 Tunis, Tunisia
- Radiochemistry Laboratory, CNSTN, Sidi Thabet Technopark, 2020 Tunis, Tunisia
| | - H. Sadaoui
- Sciences and Environmental Technologies Laboratory, High Institute of Environmental Sciences and Technologies of Borj Cedria, University of Carthage, Tunis, Tunisia
- Faculty of Science of Bizerte, University of Carthage, Jarzouna, 7000 Bizerte, Tunisia
| | - H. M. Bui
- Department of Environmental Sciences, Saigon University, Ho Chi Minh City, 70000 Vietnam
| | - F. Gantri
- Research Laboratory on Matter and Energy for Nuclear Science Development, LR16CNSTN02, CNSTN, Sidi Thabet Technopark, 2020 Tunis, Tunisia
| | - N. Mzoughi
- Sciences and Environmental Technologies Laboratory, High Institute of Environmental Sciences and Technologies of Borj Cedria, University of Carthage, Tunis, Tunisia
| |
Collapse
|
22
|
Alamilla-Sanchez ME, Alcala-Salgado MA, Alonso-Bello CD, Fonseca-Gonzalez GT. Mechanism of Action and Efficacy of Immunosupressors in Lupus Nephritis. Int J Nephrol Renovasc Dis 2021; 14:441-458. [PMID: 34924767 PMCID: PMC8675090 DOI: 10.2147/ijnrd.s335371] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 11/16/2021] [Indexed: 11/23/2022] Open
Abstract
Approximately 70% of the patients with systemic lupus erythematosus will have clinical evidence of kidney damage during their evolution. Patients with impaired renal function at onset and those with recurrent flares have a poor prognosis. Understanding the mechanism of action of immunosuppressants is essential for proper prescription. Steroids inhibit the DNA sequence that promotes the release of inflammatory cytokines. Phosphoramide mustard, metabolite of cyclophosphamide, cross-link with the DNA, causing the aggregation of an alkyl group, causing cell death. Mycophenolate inhibits inosine monophosphate dehydrogenase, prevents de novo synthesis of guanine, inducing cell arrest in S phase. Azathioprine blocks the synthesis of purines and induces apoptosis. Calcineurin inhibitors prevent the dephosphorylation of NFAT and reduce the production of interleukin 2. Antimalarials alter the enzymatic release of lysosomes by increasing intravesicular pH. The mechanism of action of rituximab is related to complement-dependent cytotoxicity and the elimination of anti-CD20-labeled B cells. Progress in the knowledge and management of low doses of steroids may change the current paradigm and reduce the frequency of related adverse events. Mycophenolate seems to be a better choice than cyclophosphamide for induction, it is also preferred over azathioprine as a maintenance immunosuppressive agent, although azathioprine is preferred in women with a desire for conception, those pregnant, or with low resources. For treatment-resistant cases, tacrolimus, rituximab or belimumab may be effective. Ongoing clinical trials with new drugs offer promising results.
Collapse
Affiliation(s)
| | | | - Cesar D Alonso-Bello
- Department of Immunology, Centro Medico Nacional “20 de Noviembre”, Mexico City, Mexico
| | | |
Collapse
|
23
|
Lertratanakul A, Sun J, Wu PW, Lee J, Dyer A, Pearce W, McPherson D, Sutton-Tyrrell K, Thompson T, Barinas-Mitchell E, Ramsey-Goldman R. Risk factors for changes in carotid intima media thickness and plaque over 5 years in women with systemic lupus erythematosus. Lupus Sci Med 2021; 8:e000548. [PMID: 34876504 PMCID: PMC8655565 DOI: 10.1136/lupus-2021-000548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/10/2021] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To investigate the occurrence of and risk factors for progression of carotid intima media thickness (IMT) and plaque in women with and without SLE. METHODS A cohort of 149 women with SLE and 126 controls participated in SOLVABLE (Study of Lupus Vascular and Bone Long-term Endpoints). Demographics, cardiovascular and SLE factors, and laboratory assessments were collected at baseline. Carotid IMT and plaque were measured using B-mode ultrasound at baseline and at 5-year follow-up. Regression models were used to identify predictors of progression in carotid IMT and plaque; multivariate models were adjusted for age, hypertension and total cholesterol to high-density lipoprotein ratio. RESULTS The mean±SD follow-up time was 5.35±0.60 years in cases and 5.62±0.66 years in controls. The mean IMT change per year was 0.008±0.015 mm in cases and 0.005±0.019 mm in controls (p=0.24). At follow-up, 31.5% of cases and 15% of controls had plaque progression, with a relative risk for plaque progression of 2.09 (95% CI 1.30 to 3.37). In SLE cases, higher fasting glucose and lower fibrinogen were associated with IMT progression after adjustment. Larger waist circumference and non-use of hydroxychloroquine were associated with plaque progression after adjustment. CONCLUSION Potential modifiable risk factors for carotid IMT and plaque progression in women with SLE were identified, suggesting that monitoring of glucose and waist circumference and use of hydroxychloroquine may be beneficial.
Collapse
Affiliation(s)
- Apinya Lertratanakul
- Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Julia Sun
- Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Peggy W Wu
- Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jungwha Lee
- Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Alan Dyer
- Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - William Pearce
- Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - David McPherson
- Cardiology, University of Texas McGovern Medical School, Houston, Texas, USA
| | | | - Trina Thompson
- Ultrasound Research Laboratory, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Emma Barinas-Mitchell
- Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
24
|
Cardiovascular Disease in Patients With Systemic Lupus Erythematosus: Potential for Improved Primary Prevention With Statins. Cardiol Rev 2021; 29:323-327. [PMID: 34609986 DOI: 10.1097/crd.0000000000000383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cardiovascular disease (CVD) is a significant cause of morbidity and mortality in patients with systemic lupus erythematosus (SLE). This is especially true in SLE patients with traditional CVD risk factors (eg, hypertension, hyperlipidemia, obesity) and disease-related risk factors (eg, increased SLE disease activity, elevated C-reactive protein levels, and antiphospholipid antibodies). The only guidelines in the primary prevention of CVD in SLE patients involve reducing traditional risk factors, but there are additional therapies that may be beneficial, including statin use. Current data on statin use for prevention of CVD in SLE patients are limited, but there have been some promising results. Statin use has been shown to be especially important in SLE patients for decreasing low-density lipoprotein levels and preventing CVD in hyperlipidemic patients. In addition, there is evidence suggesting that it may be beneficial to use statins in SLE patients with chronically elevated high-sensitivity C-reactive protein levels and antiphospholipid antibodies. It is important to continue to investigate the impact of statins on CVD in SLE patients, as they could significantly improve outcomes in patients with this disease.
Collapse
|
25
|
Haberland A, Müller J. Lack of efficacy of mono-mode of action therapeutics in COVID-19 therapy - How the lack of predictive power of preclinical cell and animal studies leads developments astray. Chem Biol Drug Des 2021; 99:32-45. [PMID: 34549885 PMCID: PMC8653042 DOI: 10.1111/cbdd.13954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/17/2021] [Accepted: 09/15/2021] [Indexed: 11/26/2022]
Abstract
The diverse experiences regarding the failure of tested drugs in the fight against COVID‐19 made it clear that one should at least question the requirement to apply classical preclinical development strategies that demand cell and animal efficacy models to be tested before going into clinical trials. Most animals are not susceptible to infection with SARS‐CoV‐2, and so this led to one‐sided virus replication experiments in cells and the use of animal models that have little in common with the complex pathogenesis of COVID‐19 in humans. Therefore, non‐clinical development strategies were designed to meet regulatory requirements, but they did not truly reflect the situation in the clinic. This has led the search for effective agents astray in many cases. As proof of this statement, we now bring together the results of such required preclinical experiments and compare with the results in clinical trials. Two clear conclusions that can be drawn from the experience to date: The required preclinical models are unsuitable for the development of innovative treatments medical devices in the case of COVID‐19 and mono‐action strategies (e.g. direct antivirals) are of very little or no benefit to patients under randomized,blinded conditions. Our hypothesis is that the complex situation of COVID‐19 may benefit from multi‐mode drugs. Here, the molecular class of aptamers could be a solution.
Collapse
|
26
|
Jolly M, Sehgal V, Arora S, Azizoddin D, Pinto B, Sharma A, Devilliers H, Inoue M, Toloza S, Bertoli A, Blazevic I, Vilá LM, Moldovan I, Torralba KD, Mazzoni D, Cicognani E, Hasni S, Goker B, Haznedaroglu S, Bourre-Tessier J, Navarra SV, Clarke A, Weisman M, Wallace D, Mok CC. Does hydroxychloroquine improve patient reported outcomes in patients with lupus? Lupus 2021; 30:1790-1798. [PMID: 34304629 DOI: 10.1177/09612033211033983] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Hydroxychloroquine (HCQ) use is associated with less disease activity, flares, damage and improved survival in Systemic Lupus Erythematosus (SLE). However, its effect on patient reported health outcomes (PROs) such as quality of life (QOL) is not known. METHODS International data from Study on Outcomes of Lupus (SOUL) from 2,161 SLE patients were compared by HCQ use. Disease activity and damage were assessed using SELENA-SLEDAI and SLICC-ACR/SDI. QOL was evaluated using LupusPRO and Lupus Impact Tracker (LIT). Linear regression analyses were performed with LupusPRO summary scores health related HRQOL, non-health related NHRQOL and LIT as dependent and HCQ use as independent variable. Analyses were undertaken to test mediation of effects of HCQ use on QOL through disease activity. RESULTS Mean age was 40.5 ± 12.8 years, 93% were women. Sixty-three (1363/2161) percent were on HCQ. On univariate analysis, HCQ use was associated with (a) better QOL (LupusPRO-HRQOL: β 6.19, 95% CI 4.15, 8.24, P ≤ 0.001, LupusPRO NHRQOL: β 5.83, 95% CI 4.02, 7.64, P ≤ 0.001) and less impact on daily life (LIT: β -9.37, 95% CI -12.24, -6.50, P ≤ 0.001). On multivariate and mediational analyses, the effects of HCQ on QOL were indirectly and completely mediated through disease activity. CONCLUSIONS HCQ use in SLE is associated with better patient reported health outcomes (LupusPRO-HRQOL and NHRQOL and impact on daily life), and the effects are mediated through disease activity. This information can facilitate patients and physician's communication with decision-making regarding the use of HCQ for SLE management.
Collapse
Affiliation(s)
| | | | | | - Desiree Azizoddin
- Emergency medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, USA
| | | | - Aman Sharma
- Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Herve Devilliers
- CHU Dijon Bourgogne - Hospital François Mitterrand, Dijon, France
| | | | - Sergio Toloza
- Rheumatology, Hospital San Juan Batista, Catamarca, Argentina
| | - Ana Bertoli
- Rheumatology, Instituto Reumatologico Strusberg, Cordoba, Spain
| | - Ivana Blazevic
- Rheumatology, 28196Universidad de Buenos Aires, 28196Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Luis M Vilá
- Division of Rheumatology, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | | | - Karina D Torralba
- Division of Rheumatology, 12221Loma Linda University School of Medicine, 12221Loma Linda University School of Medicine, Loma Linda, USA
| | - Davide Mazzoni
- Department of Oncology and Hemato-Oncology, University of Bologna, Bologna, Italy
| | - Elvira Cicognani
- Department of Oncology and Hemato-Oncology, University of Bologna, Bologna, Italy
| | - Sarfaraz Hasni
- Rheumatology, National Institute of Health, Bethesda, USA
| | - Berna Goker
- Rheumatology, Gazi University, Ankara, Turkey
| | | | | | - Sandra V Navarra
- Section of Rheumatology, University of Santo Tomas, Manila, Philippines
| | - Ann Clarke
- Division of Rheumatology, 2129University of Calgary, 2129University of Calgary, Calgary, Canada
| | - Michael Weisman
- Division of Rheumatology, Cedars Sinai Medical Center, Los Angeles, USA
| | - Daniel Wallace
- Division of Rheumatology, Cedars Sinai Medical Center, Los Angeles, USA
| | - Chi Chiu Mok
- Medicine, Tuen Mun Hospital, Hong Kong, Hong Kong
| |
Collapse
|
27
|
Mangoni AA, Woodman RJ, Piga M, Cauli A, Fedele AL, Gremese E, Erre GL. Patterns of Anti-Inflammatory and Immunomodulating Drug Usage and Microvascular Endothelial Function in Rheumatoid Arthritis. Front Cardiovasc Med 2021; 8:681327. [PMID: 34350216 PMCID: PMC8326370 DOI: 10.3389/fcvm.2021.681327] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/22/2021] [Indexed: 01/19/2023] Open
Abstract
Objectives: Specific anti-inflammatory and/or immunomodulating drugs (AIDs) can influence endothelial function which is often impaired in patients with rheumatoid arthritis (RA). We sought to determine whether overall patterns of AID usage are similarly associated with endothelial function. Methods: The reactive hyperaemia index (RHI), a marker of microvascular endothelial function, was measured in 868 RA patients reporting their intake of seven AIDs known to affect endothelial function. Latent class analysis (LCA) was performed to characterise patterns of AID usage. Models for 2-6 classes were compared using the AIC and BIC statistics and Lo-Mendell-Rubin likelihood ratio tests. Associations between the classes and RHI were adjusted for age, gender, body mass index, diabetes, HDL-cholesterol, LDL-cholesterol, family history of ischaemic heart disease, smoking status, RA duration, DAS28 score, steroid dose, existing hypertension, and C-reactive protein. Results: LCA identified five distinct AID usage classes: Class 1, generally low medication usage; Class 2, using either sulfasalazine or non-tumour necrosis factor (TNF) inhibitors; Class 3, methotrexate users; Class 4, TNF-inhibitor users; and Class 5, hydroxychloroquine users. The geometric mean for the RHI for subjects in classes 1 to 5 was 1.92, 1.81, 1.94, 2.10, and 2.07, respectively, with subjects in classes 4 and 5 having better endothelial function than subjects in class 2 (p = 0.003 for each). The glucocorticoid dosage did not influence the classes formed or the association between the classes and the RHI in sensitivity analyses. Conclusion: There were five broad patterns (classes) of AID usage in RA patients. The RHI was relatively lower in users of either sulfasalazine or non-TNF inhibitors. TNF inhibitors or hydroxychloroquine may counteract the negative effects of RA on endothelial function.
Collapse
Affiliation(s)
- Arduino A Mangoni
- Discipline of Clinical Pharmacology, Flinders Medical Centre, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Richard J Woodman
- Centre of Epidemiology and Biostatistics, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Matteo Piga
- Dipartimento di Scienze Mediche e Sanità Pubblica, Università degli Studi di Cagliari, Cagliari, Italy.,Unità Operativa Complessa (UOC) di Reumatologia, Policlinico Universitario Azienda Ospedaliero-Universitaria (AOU) Cagliari, Cagliari, Italy
| | - Alberto Cauli
- Dipartimento di Scienze Mediche e Sanità Pubblica, Università degli Studi di Cagliari, Cagliari, Italy.,Unità Operativa Complessa (UOC) di Reumatologia, Policlinico Universitario Azienda Ospedaliero-Universitaria (AOU) Cagliari, Cagliari, Italy
| | - Anna Laura Fedele
- Fondazione Policlinico Gemelli-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Università Cattolica del Sacro Cuore, Rome, Italy
| | - Elisa Gremese
- Fondazione Policlinico Gemelli-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gian Luca Erre
- Dipartimento di Scienze Mediche, Chirurgiche e Sperimentali, Università degli Studi di Sassari, Sassari, Italy.,Dipartimento di Specialità Mediche, Unità Operativa Complessa (UOC) Reumatologia, Azienda Ospedaliero-Universitaria di Sassari, Sassari, Italy
| | | |
Collapse
|
28
|
Ulander L, Tolppanen H, Hartman O, Rissanen TT, Paakkanen R, Kuusisto J, Anttonen O, Nieminen T, Yrjölä J, Ryysy R, Drews T, Utriainen S, Karjalainen P, Anttila I, Nurmi K, Silventoinen K, Koskinen M, Kovanen PT, Lehtonen J, Eklund KK, Sinisalo J. Hydroxychloroquine reduces interleukin-6 levels after myocardial infarction: The randomized, double-blind, placebo-controlled OXI pilot trial. Int J Cardiol 2021; 337:21-27. [PMID: 33961943 DOI: 10.1016/j.ijcard.2021.04.062] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/06/2021] [Accepted: 04/28/2021] [Indexed: 12/27/2022]
Abstract
OBJECTIVES To determine the anti-inflammatory effect and safety of hydroxychloroquine after acute myocardial infarction. METHOD In this multicenter, double-blind, placebo-controlled OXI trial, 125 myocardial infarction patients were randomized at a median of 43 h after hospitalization to receive hydroxychloroquine 300 mg (n = 64) or placebo (n = 61) once daily for 6 months and, followed for an average of 32 months. Laboratory values were measured at baseline, 1, 6, and 12 months. RESULTS The levels of interleukin-6 (IL-6) were comparable at baseline between study groups (p = 0.18). At six months, the IL-6 levels were lower in the hydroxychloroquine group (p = 0.042, between groups), and in the on-treatment analysis, the difference at this time point was even more pronounced (p = 0.019, respectively). The high-sensitivity C-reactive protein levels did not differ significantly between study groups at any time points. Eleven patients in the hydroxychloroquine group and four in the placebo group had adverse events leading to interruption or withdrawal of study medication, none of which was serious (p = 0.10, between groups). CONCLUSIONS In patients with myocardial infarction, hydroxychloroquine reduced IL-6 levels significantly more than did placebo without causing any clinically significant adverse events. A larger randomized clinical trial is warranted to prove the potential ability of hydroxychloroquine to reduce cardiovascular endpoints after myocardial infarction.
Collapse
Affiliation(s)
- Lotta Ulander
- Heart and Lung Center, Helsinki University Hospital, Helsinki University, Helsinki, Finland
| | - Heli Tolppanen
- Heart and Lung Center, Helsinki University Hospital, Helsinki University, Helsinki, Finland
| | - Otto Hartman
- Heart and Lung Center, Helsinki University Hospital, Helsinki University, Helsinki, Finland
| | - Tuomas T Rissanen
- Heart Center, North Karelia Central Hospital, Siunsote, Joensuu, Finland
| | - Riitta Paakkanen
- Heart and Lung Center, Helsinki University Hospital, Helsinki University, Helsinki, Finland
| | - Jouni Kuusisto
- Heart and Lung Center, Helsinki University Hospital, Helsinki University, Helsinki, Finland
| | - Olli Anttonen
- Department of Internal Medicine, Päijät-Häme Central Hospital, Lahti, Finland
| | - Tuomo Nieminen
- Department of Internal Medicine, Päijät-Häme Central Hospital, Lahti, Finland
| | - Jaana Yrjölä
- Department of Internal Medicine, Kymenlaakson Central Hospital, Kotka, Finland
| | - Ransu Ryysy
- Department of Internal Medicine, Kymenlaakson Central Hospital, Kotka, Finland
| | - Teemu Drews
- Department of Internal Medicine, South Karelia Central Hospital, Lappeenranta, Finland
| | - Seppo Utriainen
- Department of Internal Medicine, South Karelia Central Hospital, Lappeenranta, Finland
| | - Pasi Karjalainen
- Heart and Lung Center, Helsinki University Hospital, Helsinki University, Helsinki, Finland
| | - Ismo Anttila
- Department of Internal Medicine, Seinäjoki Central Hospital, Seinäjoki, Finland
| | - Katariina Nurmi
- Department of Rheumatology, Helsinki University Hospital, Helsinki University, ORTON Orthopaedic Hospital of the Orton Foundation, Helsinki, Finland
| | - Kristiina Silventoinen
- Department of Rheumatology, Helsinki University Hospital, Helsinki University, ORTON Orthopaedic Hospital of the Orton Foundation, Helsinki, Finland
| | - Miika Koskinen
- Helsinki Biobank, Helsinki University Hospital, Helsinki University, Helsinki, Finland
| | | | - Jukka Lehtonen
- Heart and Lung Center, Helsinki University Hospital, Helsinki University, Helsinki, Finland
| | - Kari K Eklund
- Department of Rheumatology, Helsinki University Hospital, Helsinki University, ORTON Orthopaedic Hospital of the Orton Foundation, Helsinki, Finland
| | - Juha Sinisalo
- Heart and Lung Center, Helsinki University Hospital, Helsinki University, Helsinki, Finland.
| |
Collapse
|
29
|
Hydroxychloroquine Potentiates Apoptosis Induced by PPAR α Antagonist in 786-O Clear Cell Renal Cell Carcinoma Cells Associated with Inhibiting Autophagy. PPAR Res 2021; 2021:6631605. [PMID: 33959154 PMCID: PMC8075691 DOI: 10.1155/2021/6631605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 03/14/2021] [Accepted: 04/05/2021] [Indexed: 02/07/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the major pathological pattern of renal cell carcinoma. The ccRCC cells exhibit a certain degree of inherent drug resistance due to some genetic mutations. In recent years, peroxisome proliferator-activated receptor-α (PPARα) antagonists have been reported as a targeted therapeutic drug capable of inducing apoptosis and cell cycle arrest in the ccRCC cell line. Autophagy, which can be induced by stress in eukaryotic cells, plays a complex role in the proliferation, survival, and death of tumor cells. In our study, we found that the expression of PPARα was low in highly differentiated ccRCC tissues and 786-O cell line but high in poorly differentiated ccRCC tissues. The level of PPARα expression in ccRCC tissues is correlated to the grade of differentiation, but not to the sex or age of ccRCC patients. The findings also revealed that the PPARα antagonist GW6471 can lower cell viability and induce autophagy in the 786-O ccRCC cell line. This autophagy can be inhibited by hydroxychloroquine. When treated with a combination of hydroxychloroquine and GW6471, the viability of the 786-O cells was decreased further when compared to the treatment with GW6471 or hydroxychloroquine alone, and apoptosis was promoted. Meanwhile, when human kidney 2 cells were cotreated with hydroxychloroquine and GW6471, cell viability was only slightly influenced. Hence, our finding indicates that the combination of GW6471 and hydroxychloroquine may constitute a novel and potentially effective treatment for ccRCC. Furthermore, this approach is likely to be safe owing to its minimal effects on normal renal tissues.
Collapse
|
30
|
van Leent MMT, Beldman TJ, Toner YC, Lameijer MA, Rother N, Bekkering S, Teunissen AJP, Zhou X, van der Meel R, Malkus J, Nauta SA, Klein ED, Fay F, Sanchez-Gaytan BL, Pérez-Medina C, Kluza E, Ye YX, Wojtkiewicz G, Fisher EA, Swirski FK, Nahrendorf M, Zhang B, Li Y, Zhang B, Joosten LAB, Pasterkamp G, Boltjes A, Fayad ZA, Lutgens E, Netea MG, Riksen NP, Mulder WJM, Duivenvoorden R. Prosaposin mediates inflammation in atherosclerosis. Sci Transl Med 2021; 13:eabe1433. [PMID: 33692130 PMCID: PMC8209679 DOI: 10.1126/scitranslmed.abe1433] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/17/2020] [Accepted: 02/17/2021] [Indexed: 12/13/2022]
Abstract
Macrophages play a central role in the pathogenesis of atherosclerosis. The inflammatory properties of these cells are dictated by their metabolism, of which the mechanistic target of rapamycin (mTOR) signaling pathway is a key regulator. Using myeloid cell-specific nanobiologics in apolipoprotein E-deficient (Apoe -/-) mice, we found that targeting the mTOR and ribosomal protein S6 kinase-1 (S6K1) signaling pathways rapidly diminished plaque macrophages' inflammatory activity. By investigating transcriptome modifications, we identified Psap, a gene encoding the lysosomal protein prosaposin, as closely related with mTOR signaling. Subsequent in vitro experiments revealed that Psap inhibition suppressed both glycolysis and oxidative phosphorylation. Transplantation of Psap -/- bone marrow to low-density lipoprotein receptor knockout (Ldlr -/-) mice led to a reduction in atherosclerosis development and plaque inflammation. Last, we confirmed the relationship between PSAP expression and inflammation in human carotid atherosclerotic plaques. Our findings provide mechanistic insights into the development of atherosclerosis and identify prosaposin as a potential therapeutic target.
Collapse
Affiliation(s)
- Mandy M T van Leent
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Experimetal Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, 1105 AZ Amsterdam, Netherlands
| | - Thijs J Beldman
- Experimetal Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, 1105 AZ Amsterdam, Netherlands
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Yohana C Toner
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Marnix A Lameijer
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Experimetal Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, 1105 AZ Amsterdam, Netherlands
| | - Nils Rother
- Department of Nephrology and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Siroon Bekkering
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Abraham J P Teunissen
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xianxiao Zhou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Roy van der Meel
- Department of Chemical Biology, Eindhoven University of Technology, 5612 AZ Eindhoven, Netherlands
| | - Joost Malkus
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sheqouia A Nauta
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Emma D Klein
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Francois Fay
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Institut Galien Paris-Saclay, Faculté de Pharmacie, CNRS, Université Paris-Saclay, 92 296 Châtenay-Malabry, France
| | - Brenda L Sanchez-Gaytan
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Chemistry Center, Science Institute, Meritorious Autonomous University of Puebla, Puebla 72570, Mexico
| | - Carlos Pérez-Medina
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Ewelina Kluza
- Department of Chemical Biology, Eindhoven University of Technology, 5612 AZ Eindhoven, Netherlands
| | - Yu-Xiang Ye
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
- Department of Diagnostic and Interventional Radiology, University Hospitals Tuebingen, 72076 Tuebingen, Germany
| | - Gregory Wojtkiewicz
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Edward A Fisher
- Department of Medicine (Cardiology) and Cell Biology, Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Filip K Swirski
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yang Li
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
- Centre for Individualised Infection Medicine (CiiM) and TWINCORE, joint ventures between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), 30625 Hannover, Germany
| | - Bowen Zhang
- Centre for Individualised Infection Medicine (CiiM) and TWINCORE, joint ventures between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), 30625 Hannover, Germany
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
- Department of Medical Genetics, University of Medicine and Pharmacy, Iuliu Haţieganu, Cluj-Napoca 400000, Romania
| | - Gerard Pasterkamp
- Central Diagnostics Laboratory, Division Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, Netherlands
| | - Arjan Boltjes
- Central Diagnostics Laboratory, Division Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, Netherlands
| | - Zahi A Fayad
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Esther Lutgens
- Experimetal Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, 1105 AZ Amsterdam, Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, 80331 Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, 80539 Munich, Germany
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
- Department for Genomics and Immunoregulation, Life and Medical Sciences Institute, University of Bonn, 53127 Bonn, Germany
| | - Niels P Riksen
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Willem J M Mulder
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
- Department of Chemical Biology, Eindhoven University of Technology, 5612 AZ Eindhoven, Netherlands
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Raphaël Duivenvoorden
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- Department of Nephrology and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| |
Collapse
|
31
|
Ahmed S, Gasparyan AY, Zimba O. Comorbidities in rheumatic diseases need special consideration during the COVID-19 pandemic. Rheumatol Int 2021; 41:243-256. [PMID: 33388969 PMCID: PMC7778868 DOI: 10.1007/s00296-020-04764-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 11/26/2020] [Indexed: 02/08/2023]
Abstract
Comorbidities in rheumatic and musculoskeletal diseases (RMDs) not only increase morbidity and mortality but also confound disease activity, limit drug usage and increase chances of severe infections or drug-associated adverse effects. Most RMDs lead to accelerated atherosclerosis and variable manifestations of the metabolic syndrome. Literature on COVID-19 in patients with RMDs, and the effects of various comorbidities on COVID-19 was reviewed. The initial data of COVID-19 infections in RMDs have not shown an increased risk for severe disease or the use of different immunosuppression. However, there are some emerging data that patients with RMDs and comorbidities may fare worse. Various meta-analyses have reiterated that pre-existing hypertension, cardiovascular disease, stroke, diabetes, chronic kidney disease, heart failure, lung disease or obesity predispose to increased COVID-19 mortality. All these comorbidities are commonly encountered in the various RMDs. Presence of comorbidities in RMDs pose a greater risk than the RMDs themselves. A risk score based on comorbidities in RMDs should be developed to predict severe COVID-19 and death. Additionally, there should be active management of such comorbidities to mitigate these risks. The pandemic must draw our attention towards, and not away from, comorbidities.
Collapse
Affiliation(s)
- Sakir Ahmed
- Department of Clinical Immunology and Rheumatology, Kalinga Institute of Medical Sciences (KIMS), KIIT University, Bhubaneswar, 751024 India
| | - Armen Yuri Gasparyan
- Departments of Rheumatology and Research and Development, Dudley Group NHS Foundation Trust (Teaching Trust of the University of Birmingham, UK), Russells Hall Hospital, Dudley, West Midlands UK
| | - Olena Zimba
- Department of Internal Medicine No. 2, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| |
Collapse
|
32
|
Potent synthetic and endogenous ligands for the adopted orphan nuclear receptor Nurr1. Exp Mol Med 2021; 53:19-29. [PMID: 33479411 PMCID: PMC8080818 DOI: 10.1038/s12276-021-00555-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/19/2020] [Accepted: 11/05/2020] [Indexed: 01/30/2023] Open
Abstract
Until recently, Nurr1 (NR4A2) was known as an orphan nuclear receptor without a canonical ligand-binding domain, featuring instead a narrow and tight cavity for small molecular ligands to bind. In-depth characterization of its ligand-binding pocket revealed that it is highly dynamic, with its structural conformation changing more than twice on the microsecond-to-millisecond timescale. This observation suggests the possibility that certain ligands are able to squeeze into this narrow space, inducing a conformational change to create an accessible cavity. The cocrystallographic structure of Nurr1 bound to endogenous ligands such as prostaglandin E1/A1 and 5,6-dihydroxyindole contributed to clarifying the crucial roles of Nurr1 and opening new avenues for therapeutic interventions for neurodegenerative and/or inflammatory diseases related to Nurr1. This review introduces novel endogenous and synthetic Nurr1 agonists and discusses their potential effects in Nurr1-related diseases.
Collapse
|
33
|
Piga M, Floris A, Sebastiani GD, Prevete I, Iannone F, Coladonato L, Govoni M, Bortoluzzi A, Mosca M, Tani C, Doria A, Iaccarino L, Franceschini F, Fredi M, Conti F, Spinelli FR, Galeazzi M, Bellisai F, Zanetti A, Carrara G, Scirè CA, Mathieu A. Risk factors of damage in early diagnosed systemic lupus erythematosus: results of the Italian multicentre Early Lupus Project inception cohort. Rheumatology (Oxford) 2021; 59:2272-2281. [PMID: 31840179 DOI: 10.1093/rheumatology/kez584] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/25/2019] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE To investigate risk factors for damage development in a prospective inception cohort of early diagnosed SLE patients. METHODS The Early Lupus Project recruited an inception cohort of patients within 12 months of SLE classification (1997 ACR criteria). At enrolment and every 6 months thereafter, the SLICC/ACR Damage Index was recorded. The contribution of baseline and time-varying covariates to the development of damage, defined as any SLICC/ACR Damage Index increase from 0 to ≥1, was assessed using univariate analysis. Forward-backward Cox regression models were fitted with covariates with P < 0.05 to identify factors independently associated with the risk of damage development. RESULTS Overall, 230 patients with a mean (s.d.) age of 36.5 (14.4) years were eligible for this study; the mean number of visits per patient was 5.3 (2.7). There were 51 (22.2%) patients with SLICC/ACR Damage Index ≥1 after 12 months, 59 (25.6%) after 24 months and 67 (29.1%) after 36 months. Dyslipidaemia [P = 0.001; hazard ratio (HR) 2.9; 95% CI 1.5, 5.6], older age (P = 0.001; HR 3.0; 95% CI 1.6, 5.5), number of organs/systems involved (P = 0.002; HR 1.4; 95% CI 1.1, 1.8) and cardiorespiratory involvement (P = 0.041; HR 1.9; 95% CI 1.0, 3.7) were independently associated with an increased risk of developing damage. Risk profiles for damage development differed for glucocorticoid-related and -unrelated damage. HCQ use (P = 0.005; HR 0.4; 95% CI 0.2, 0.8) reduced the risk of glucocorticoid-unrelated damage. CONCLUSION We identified risk factors of damage development, but little effect of glucocorticoids, in this early SLE cohort. Addressing modifiable risk factors from the time of SLE diagnosis might improve patient outcomes.
Collapse
Affiliation(s)
- Matteo Piga
- Rheumatology Unit, University of Cagliari and AOU University Clinic, Cagliari
| | - Alberto Floris
- Rheumatology Unit, University of Cagliari and AOU University Clinic, Cagliari
| | | | - Imma Prevete
- UOC di Reumatologia, Azienda Ospedaliera San Camillo-Forlanini, Rome
| | - Florenzo Iannone
- Dipartimento dell'Emergenza e dei Trapianto di Organi - Sezione di Reumatologia, Università di Bari, Bari
| | - Laura Coladonato
- Dipartimento dell'Emergenza e dei Trapianto di Organi - Sezione di Reumatologia, Università di Bari, Bari
| | - Marcello Govoni
- UOC e Sezione di Reumatologia - Dipartimento di Scienze Mediche, Università degli Studi di Ferrara, Ferrara
| | - Alessandra Bortoluzzi
- UOC e Sezione di Reumatologia - Dipartimento di Scienze Mediche, Università degli Studi di Ferrara, Ferrara
| | - Marta Mosca
- UOC di Reumatologia, Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa
| | - Chiara Tani
- UOC di Reumatologia, Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine, University of Padova, Padova
| | - Luca Iaccarino
- Rheumatology Unit, Department of Medicine, University of Padova, Padova
| | - Franco Franceschini
- UOC di Reumatologia e Immunologia Clinica, Dipartimento di Scienze Cliniche e Sperimentali, Università degli Studi di Brescia, ASST Spedali Civili, Brescia, Italy
| | - Micaela Fredi
- UOC di Reumatologia e Immunologia Clinica, Dipartimento di Scienze Cliniche e Sperimentali, Università degli Studi di Brescia, ASST Spedali Civili, Brescia, Italy
| | - Fabrizio Conti
- Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Roma
| | - Francesca Romana Spinelli
- Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Roma
| | - Mauro Galeazzi
- UOC di Reumatologia, Azienda Ospedaliera Universitaria Senese, Siena
| | | | - Anna Zanetti
- Società Italiana di Reumatologia, Unità Epidemiologica, Milano.,Divisione di Biostatistica, Epidemiologia e Salute Pubblica, Dipartimento di Statistica e Metodi Quantitativi, Università degli Studi di Milano-Bicocca, Milano, Italy
| | - Greta Carrara
- Società Italiana di Reumatologia, Unità Epidemiologica, Milano
| | | | - Alessandro Mathieu
- Rheumatology Unit, University of Cagliari and AOU University Clinic, Cagliari
| |
Collapse
|
34
|
Piga M, Arnaud L. The Main Challenges in Systemic Lupus Erythematosus: Where Do We Stand? J Clin Med 2021; 10:E243. [PMID: 33440874 PMCID: PMC7827672 DOI: 10.3390/jcm10020243] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/04/2021] [Accepted: 01/08/2021] [Indexed: 12/12/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an immune-mediated multi-systemic disease characterized by a wide variability of clinical manifestations and a course frequently subject to unpredictable flares. Despite significant advances in the understanding of the pathophysiology and optimization of medical care, patients with SLE still have significant mortality and carry a risk of progressive organ damage accrual and reduced health-related quality of life. New tools allow earlier classification of SLE, whereas tailored early intervention and treatment strategies targeted to clinical remission or low disease activity could offer the opportunity to reduce damage, thus improving long-term outcomes. Nevertheless, the early diagnosis of SLE is still an unmet need for many patients. Further disentangling the SLE susceptibility and complex pathogenesis will allow to identify more accurate biomarkers and implement new ways to measure disease activity. This could represent a major step forward to find new trials modalities for developing new drugs, optimizing the use of currently available therapeutics and minimizing glucocorticoids. Preventing and treating comorbidities in SLE, improving the management of hard-to-treat manifestations including management of SLE during pregnancy are among the remaining major unmet needs. This review provides insights and a research agenda for the main challenges in SLE.
Collapse
Affiliation(s)
- Matteo Piga
- Rheumatology Unit, AOU University Clinic and University of Cagliari, 09042 Cagliari, Italy;
| | - Laurent Arnaud
- Service de Rhumatologie, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, 67000 Strasbourg, France
- Centre National de Références des Maladies Systémiques et Auto-immunes Rares Est Sud-Ouest (RESO), 67000 Strasbourg, France
| |
Collapse
|
35
|
Haugaard JH, Dreyer L, Ottosen MB, Gislason G, Kofoed K, Egeberg A. Use of hydroxychloroquine and risk of major adverse cardiovascular events in patients with lupus erythematosus: A Danish nationwide cohort study. J Am Acad Dermatol 2020; 84:930-937. [PMID: 33321159 DOI: 10.1016/j.jaad.2020.12.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/08/2020] [Accepted: 12/01/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Limited data suggest that hydroxychloroquine may affect risk of cardiovascular disease in patients with lupus erythematosus (LE). OBJECTIVE To investigate whether hydroxychloroquine treatment is associated with major adverse cardiovascular events (MACE) (myocardial infarction, ischemic stroke, or cardiovascular-associated death) in patients with cutaneous LE (CLE) or systemic LE (SLE). METHODS Based on the Danish nationwide registers, an observational cohort study was conducted including patients with first-time diagnosis of CLE or SLE (between 1997 and 2017). Cox regression models calculating the hazard ratio (HR) analyzing the risk of MACE were performed comparing time on and off hydroxychloroquine (including never users). The models were adjusted for age, sex, socioeconomic status, concomitant treatment, and cardiovascular risk factors. RESULTS Among 4587 patients with LE, 51% (n = 2343) were treated with hydroxychloroquine during the study period. An inverse association between use of hydroxychloroquine and MACE risk was observed among patients with SLE (adjusted HR, 0.65; 95% confidence interval, 0.46-0.90) and patients with CLE (adjusted HR, 0.71; 95% confidence interval, 0.42-1.19). Consistent results were found in sensitivity analyses including a case-time control design. LIMITATIONS No information on disease activity/severity was available. CONCLUSION Our findings indicate an opportunity to reduce the risk of cardiovascular events in patients with LE through use of hydroxychloroquine.
Collapse
Affiliation(s)
- Jeanette Halskou Haugaard
- Department of Dermatology, Allergy and Venerology, Herlev and Gentofte University Hospital, Copenhagen, Denmark.
| | - Lene Dreyer
- Departments of Clinical Medicine and Rheumatology, Aalborg University and Aalborg University Hospital, Aalborg, Denmark
| | - Mathias Bo Ottosen
- Department of Dermatology, Allergy and Venerology, Herlev and Gentofte University Hospital, Copenhagen, Denmark
| | - Gunnar Gislason
- Department of Cardiology, Herlev and Gentofte University Hospital, Copenhagen, Denmark
| | - Kristian Kofoed
- Department of Dermatology, Allergy and Venerology, Herlev and Gentofte University Hospital, Copenhagen, Denmark
| | - Alexander Egeberg
- Department of Dermatology, Allergy and Venerology, Herlev and Gentofte University Hospital, Copenhagen, Denmark
| |
Collapse
|
36
|
Morris G, Athan E, Walder K, Bortolasci CC, O'Neil A, Marx W, Berk M, Carvalho AF, Maes M, Puri BK. Can endolysosomal deacidification and inhibition of autophagy prevent severe COVID-19? Life Sci 2020; 262:118541. [PMID: 33035581 PMCID: PMC7537668 DOI: 10.1016/j.lfs.2020.118541] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 02/06/2023]
Abstract
The possibility is examined that immunomodulatory pharmacotherapy may be clinically useful in managing the pandemic coronavirus disease 2019 (COVID-19), known to result from infection by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a positive-sense single-stranded RNA virus. The dominant route of cell entry of the coronavirus is via phagocytosis, with ensconcement in endosomes thereafter proceeding via the endosomal pathway, involving transfer from early (EEs) to late endosomes (LEs) and ultimately into lysosomes via endolysosomal fusion. EE to LE transportation is a rate-limiting step for coronaviruses. Hence inhibition or dysregulation of endosomal trafficking could potentially inhibit SARS-CoV-2 replication. Furthermore, the acidic luminal pH of the endolysosomal system is critical for the activity of numerous pH-sensitive hydrolytic enzymes. Golgi sub-compartments and Golgi-derived secretory vesicles also depend on being mildly acidic for optimal function and structure. Activation of endosomal toll-like receptors by viral RNA can upregulate inflammatory mediators and contribute to a systemic inflammatory cytokine storm, associated with a worsened clinical outcome in COVID-19. Such endosomal toll-like receptors could be inhibited by the use of pharmacological agents which increase endosomal pH, thereby reducing the activity of acid-dependent endosomal proteases required for their activity and/or assembly, leading to suppression of antigen-presenting cell activity, decreased autoantibody secretion, decreased nuclear factor-kappa B activity and decreased pro-inflammatory cytokine production. It is also noteworthy that SARS-CoV-2 inhibits autophagy, predisposing infected cells to apoptosis. It is therefore also suggested that further pharmacological inhibition of autophagy might encourage the apoptotic clearance of SARS-CoV-2-infected cells.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Eugene Athan
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia; Department of Infectious Disease, Barwon Health, Geelong, Australia
| | - Ken Walder
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Chiara C Bortolasci
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia; Deakin University, Centre for Molecular and Medical Research, School of Medicine, Geelong, Victoria, Australia
| | - Adrienne O'Neil
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Wolf Marx
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Michael Berk
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, the Department of Psychiatry, the Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - André F Carvalho
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Michael Maes
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia; Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | | |
Collapse
|
37
|
Hammam N, Abdel-Wahab N, Gheita TA. Atherogenic Index of Plasma in Women with Rheumatoid Arthritis and Systemic Lupus Erythematosus: A 10-Year Potential Predictor of Cardiovascular Disease. Curr Rheumatol Rev 2020; 17:122-130. [PMID: 33030132 DOI: 10.2174/1573397116666201007123403] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 06/18/2020] [Accepted: 06/29/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Women with rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE) are at high risk of cardiovascular diseases (CVD). The atherogenic index of plasma (AIP) is a new marker for the assessment of CVD. OBJECTIVE This study aimed to determine the predictive value of AIP with long-term CVD risk among women with RA and SLE. METHODS This is a cross-sectional study of 99 RA and 59 SLE women. Demographic, clinical, and biochemical data were obtained, and disease activities were calculated. For each patient, the longterm risk of CVD was calculated using the Framingham risk score (FRS); AIP was derived according to the logarithmic (triglycerides/high-density lipoproteins cholesterol). RESULTS The mean age of the RA and SLE patients was 47.97 ± 8.78 and 36.75 ± 9.09 years, respectively. The median (interquartile range) of AIP values in RA and SLE patients were 0.34 (-0.15, 1.02) and 0.33 (-0.53, 0.96), respectively, while FRS values of RA patients and SLE patients were 6.38 ± 5.58 and 4.86 ± 4.5, respectively (p >0.05). There was a moderate correlation between AIP and FRS in RA and SLE patients (r=0.42, p=0.002 and r=0.33, p=0.007, respectively). According to the multivariate regression analyses, we found that AIP value is an independent factor for FRS in RA (β: 4.13, 95% confidence interval; 1.71, 6.18; p=0.008) and in SLE patients (β: 6.19, 95% confidence interval; 2.58, 9.81; p<0.001). CONCLUSIONS We reported that AIP can be used as an independent indicator for long-term CVD risk in RA and SLE patients.
Collapse
Affiliation(s)
- Nevin Hammam
- Rheumatology and Rehabilitation Department, Faculty of Medicine, Assiut University, Asyut, Egypt
| | - Noha Abdel-Wahab
- Rheumatology and Rehabilitation Department, Faculty of Medicine, Assiut University, Asyut, Egypt
| | - Tamer A Gheita
- Rheumatology Department Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
38
|
Sakai R, Honda S, Tanaka E, Majima M, Konda N, Takada H, Harigai M. The risk of hospitalized infection in patients with systemic lupus erythematosus treated with hydroxychloroquine. Lupus 2020; 29:1712-1718. [DOI: 10.1177/0961203320952853] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Objective To compare the risk of hospitalized infection (HI) between users and non-users of hydroxychloroquine (HCQ) in systemic lupus erythematosus (SLE). Methods Using claims data, patients were defined as SLE cases by the following criteria: 1) they had at least one SLE diagnostic code; 2) they had a prescription for specific drugs, including corticosteroids, steroid pulse therapy, and immunosuppressive drugs; and 3) they were at least 16 years old between September 2015 and July 2017 (n = 17,483). The SLE cases with at least one prescription for HCQ were defined as the HCQ group (n = 1,431), while the others were defined as the non-HCQ group. Among the SLE cases, propensity score-matched cases were observed for 1 year (n = 1,095 in each group). Results The median age and proportion of female patients in both groups were about 42 years and 88%, respectively. The proportions of cases with HIs were similar (HCQ group, 4.5%; non-HCQ group, 5.6%; p = 0.240, McNemar test). The hazard ratio of the HCQ group for HIs after adjusting for patients’ characteristics was not significant at 0.9 (0.6–1.3). Conclusion The use of HCQ was not associated with a risk of HIs in patients with SLE.
Collapse
Affiliation(s)
- Ryoko Sakai
- Department of Rheumatology, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
- Division of Epidemiology and Pharmacoepidemiology of Rheumatic Diseases, Department of Rheumatology, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
| | - Suguru Honda
- Department of Rheumatology, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
| | - Eiichi Tanaka
- Department of Rheumatology, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
| | - Masako Majima
- Department of Rheumatology, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
| | - Naoko Konda
- Department of Rheumatology, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
| | - Hideto Takada
- Department of Rheumatology, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
| | - Masayoshi Harigai
- Department of Rheumatology, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
- Division of Epidemiology and Pharmacoepidemiology of Rheumatic Diseases, Department of Rheumatology, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
| |
Collapse
|
39
|
Chen HJ, Tas SW, de Winther MPJ. Type-I interferons in atherosclerosis. J Exp Med 2020; 217:132613. [PMID: 31821440 PMCID: PMC7037237 DOI: 10.1084/jem.20190459] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 10/05/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022] Open
Abstract
Chen et al. review the effects of type-I IFNs and the potential of anti–type-I IFN therapies in atherosclerosis. The contribution of dyslipidemia and inflammation in atherosclerosis is well established. Along with effective lipid-lowering treatments, the recent success of clinical trials with anti-inflammatory therapies and the accelerated atherosclerosis in many autoimmune diseases suggest that targeting inflammation may open new avenues for the prevention and the treatment for cardiovascular diseases (CVDs). In the past decades, studies have widened the role of type-I interferons (IFNs) in disease, from antivirus defense to autoimmune responses and immuno-metabolic syndromes. While elevated type-I IFN level in serum is associated with CVD incidence in patients with interferonopathies, experimental data have attested that type-I IFNs affect plaque-residing macrophages, potentiate foam cell and extracellular trap formation, induce endothelial dysfunction, alter the phenotypes of dendritic cells and T and B lymphocytes, and lead to exacerbated atherosclerosis outcomes. In this review, we discuss the production and the effects of type-I IFNs in different atherosclerosis-associated cell types from molecular biology studies, animal models, and clinical observations, and the potential of new therapies against type-I IFN signaling for atherosclerosis.
Collapse
Affiliation(s)
- Hung-Jen Chen
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Sander W Tas
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology and Clinical Immunology, and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, Netherlands
| | - Menno P J de Winther
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany
| |
Collapse
|
40
|
López P, Rodríguez-Carrio J, Martínez-Zapico A, Pérez-Álvarez ÁI, Benavente L, Caminal-Montero L, Suárez A. IgM anti-phosphorylcholine antibodies associate with senescent and IL-17+ T cells in SLE patients with a pro-inflammatory lipid profile. Rheumatology (Oxford) 2020; 59:407-417. [PMID: 31302689 DOI: 10.1093/rheumatology/kez264] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 06/04/2019] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE The aim was to evaluate whether T cell subsets and the lipid profile could be linked to the cardioprotective effect of IgM anti-phosphorylcholine (PC) antibodies in SLE. METHODS Anti-PC antibodies were quantified by ELISA in 197 patients and 99 controls and analysed in relationship to clinical features, treatments and serum lipids. Carotid atheromatosis was evaluated by ultrasonography; Th1, Th17, Treg and CD4+CD28null cells by flow cytometry; and cytokine serum levels by immunoassays, in a subgroup of 120 SLE patients and 33 controls. RESULTS IgM anti-PC serum levels were reduced in SLE patients compared with controls (P < 0.001) and were associated with age (β= -0.252; P = 0.002), high-density lipoprotein (HDL; β = 0.271; P = 0.001), low-density lipoprotein (LDL; β= -0.192; P = 0.017) and glucocorticoid treatment (β= -0.201; P = 0.012), whereas the IgG-to-IgM anti-PC ratio was increased (P = 0.007) and associated with age (β = 0.194; P = 0.028) and SLEDAI (β = 0.250; P = 0.005). Also, patients with clinical or subclinical cardiovascular disease exhibited reduced IgM anti-PC levels compared with their cardiovascular disease-free counterparts, regardless of glucocorticoid usage (P = 0.001). CD4+CD28null and Th17 cells were increased in SLE patients compared with controls (P < 0.01) and correlated inversely with IgM anti-PC levels. These associations were observed in patients displaying high triglyceride or low HDL levels, even after adjusting for clinical parameters and treatments (CD4+CD28null: β = -0.455, P = 0.001; Th17: β= -0.280, P = 0.035), but not in those with a normal lipid profile. High triglyceride and low HDL profiles were related to low IgM anti-PC and Treg levels, respectively, whereas both lipid profiles were associated with inflammatory markers and cytokines. CONCLUSION The present study provides evidence for an association of IgM anti-PC antibodies with pro-atherogenic T cell subsets in SLE, with a high triglyceride/low HDL lipid profile playing a facilitating major role.
Collapse
Affiliation(s)
- Patricia López
- Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo.,Group of Basic and Translational Research in Inflammatory Diseases, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)
| | - Javier Rodríguez-Carrio
- Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo.,Group of Basic and Translational Research in Inflammatory Diseases, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)
| | - Aleida Martínez-Zapico
- Department of Internal Medicine, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | - Lorena Benavente
- Department of Neurology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Luis Caminal-Montero
- Group of Basic and Translational Research in Inflammatory Diseases, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA).,Department of Internal Medicine, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Ana Suárez
- Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo.,Group of Basic and Translational Research in Inflammatory Diseases, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)
| |
Collapse
|
41
|
Clarke AE, Yazdany J, Kabadi SM, Durden E, Winer I, Griffing K, Costenbader KH. The economic burden of systemic lupus erythematosus in commercially- and medicaid-insured populations in the United States. Semin Arthritis Rheum 2020; 50:759-768. [PMID: 32531505 DOI: 10.1016/j.semarthrit.2020.04.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 04/07/2020] [Accepted: 04/30/2020] [Indexed: 12/01/2022]
Abstract
OBJECTIVE To estimate the economic burden of systematic lupus erythematous (SLE), stratified by disease severity, in commercially- and Medicaid-insured US populations. METHODS Adults (≥18 years) with SLE treated with antimalarials, selected biologics, immunosuppressants, and systemic glucocorticoids (2010-2014) were identified within the commercial and Medicaid insurance IBM MarketScan® databases (index date = first SLE medication claim). Both cohorts were stratified into mild (receiving antimalarial or glucocorticoid monotherapy ≤5 mg/day) versus moderate/severe SLE (receiving glucocorticoids >5 mg/day, biologic, immunosuppressant, or combination therapy) during a 6-month exposure period. All-cause healthcare utilization and costs were evaluated during the 12 months following the exposure period. RESULTS Among 8231 commercially-insured patients, 32.6% had mild and 67.4% had moderate/severe SLE by our definition. Among 802 Medicaid-insured patients, 25.2% had mild and 74.8% had moderate/severe SLE. Adjusted mean total healthcare costs, excluding pharmacy, for moderate/severe SLE patients were higher than for mild SLE patients in the commercially-insured ($39,021 versus $23,519; p < 0.0001) and Medicaid-insured populations ($56,050 versus $44,932; p = 0.06). In both SLE severity populations total unadjusted costs were significantly higher among Medicaid-insured than commercially-insured patients. CONCLUSION Commercially-insured patients with treatment suggesting moderate/severe SLE incurred significantly higher adjusted mean healthcare costs, excluding pharmacy, compared with mild SLE patients. While not reaching statistical significance, moderate/severe Medicaid-insured patients had higher costs then mild SLE patients. Total unadjusted healthcare costs were significantly higher among Medicaid-insured than commercially-insured patients. These differential costs are important to consider and monitor when implementing interventions to improve health and reduce healthcare spending for SLE.
Collapse
Affiliation(s)
- Ann E Clarke
- Division of Rheumatology, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, Alberta, Canada.
| | - Jinoos Yazdany
- University of California - San Francisco, San Francisco, CA, USA
| | | | | | | | | | | |
Collapse
|
42
|
Mangoni AA, Erre GL. Repurposing Available Anti-inflammatory and Immunomodulating Agents for Cardiovascular Risk Management: A Call for Submissions to Current Clinical Pharmacology. CURRENT CLINICAL PHARMACOLOGY 2020; 15:2-3. [PMID: 32407260 DOI: 10.2174/157488471501200319150358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Affiliation(s)
- Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, Australia
- Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Gian L Erre
- UOC di Reumatologia, Dipartimento di Specialità Mediche, Azienda Ospedaliero-Universitaria di Sassari, Sassari, Italy
- Department of Clinical and Experimental Medicine, School of Medicine, University of Sassari, Sassari, Italy
| |
Collapse
|
43
|
Wondafrash DZ, Desalegn TZ, Yimer EM, Tsige AG, Adamu BA, Zewdie KA. Potential Effect of Hydroxychloroquine in Diabetes Mellitus: A Systematic Review on Preclinical and Clinical Trial Studies. J Diabetes Res 2020; 2020:5214751. [PMID: 32190699 PMCID: PMC7064866 DOI: 10.1155/2020/5214751] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 01/14/2020] [Accepted: 02/11/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Diabetes mellitus is a chronic metabolic disorder characterized by persistent hyperglycemia. It affects millions of people globally. In spite of many antidiabetic drugs that are available, an adequate level of control remains challenging. Hydroxychloroquine is an immunomodulatory drug that has been used for the treatment of malaria and autoimmune diseases. There is an emerging evidence that suggests its beneficial effect against diabetes mellitus. Therefore, this systematic review is aimed at discoursing the role of hydroxychloroquine against diabetes mellitus and its potential mechanisms of actions. METHODS A systematic and manual searching was carried out to retrieve relevant articles (preclinical and clinical studies) published from January 2014 to July 2019. Electronic databases including PubMed and Scopus as well as clinicaltrials.gov have been searched using different searching terms: "hydroxychloroquine," "diabetes mellitus," "hyperglycemia," and "insulin resistance." The MeSH terms (PubMed) and text words were combined with "AND" or "OR." In addition, manual searching of Google Engine and Google Scholar was conducted. Quality assessment of all the included studies was performed using CAMARADES (preclinical studies) and the Newcastle-Ottawa Scale and Cochrane Collaboration's tools (clinical studies). RESULTS A total of eighteen studies (three experimental and fifteen clinical studies) were found to be eligible for the present systematic review. Among the included clinical studies (six randomized control trials, five observational studies, and four cohort studies), about 55,776 study participants were involved. Most of these studies showed significant improvement of lipid profile and insulin levels and substantial diminution of hemoglobin A1c, fasting plasma glucose, and postprandial blood glucose levels. Reduction in lysosomal degradation of the internal insulin-insulin receptor complex and enhancement in insulin sensitivity and adiponectin levels are some of the hypothesized mechanisms for the antidiabetic effect of hydroxychloroquine. CONCLUSION The current review provides preliminary evidence for potential antidiabetic properties of hydroxychloroquine. Though the provided available data were promising, further clinical trials and mechanistic studies are needed to determine its long-term effects.
Collapse
Affiliation(s)
- Dawit Zewdu Wondafrash
- Department of Pharmacology and Toxicology, School of Pharmacy, Mekelle University, Mekelle, Ethiopia
| | - Tsion Zewdu Desalegn
- Department of Pharmacology and Toxicology, School of Pharmacy, Mekelle University, Mekelle, Ethiopia
| | - Ebrahim M. Yimer
- Department of Pharmacy, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Arega Gashaw Tsige
- Clinical Pharmacy Research and Course Unit, School of Pharmacy, Mekelle University, Mekelle, Ethiopia
| | | | - Kaleab Alemayehu Zewdie
- Department of Pharmacology and Toxicology, School of Pharmacy, Mekelle University, Mekelle, Ethiopia
| |
Collapse
|
44
|
Shukla AM, Wagle Shukla A. Expanding horizons for clinical applications of chloroquine, hydroxychloroquine, and related structural analogues. Drugs Context 2019; 8:2019-9-1. [PMID: 31844421 PMCID: PMC6905642 DOI: 10.7573/dic.2019-9-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 10/08/2019] [Accepted: 10/09/2019] [Indexed: 12/11/2022] Open
Abstract
Several experimental and clinical studies have transformed the traditional antimalarial role of chloroquine (CHQ) and related structural analogues to potent therapeutic agents for a host of nonmalarial indications. The expanding clinical applicability for these drugs includes rheumatological and cardiovascular disorders (CVD), chronic kidney disease (CKD), oncology, and a variety of nonmalarial infections. These clinical advancements are primarily related to pleiotropic pharmacological actions of these drugs, including immunomodulation, anti-inflammatory properties, and capabilities of inducing autophagy and apoptosis at a cellular level. Historically, many clinical benefits in nonmalarial indications were first recognized through serendipitous observations; however, with numerous ongoing systematic clinical studies, the clinical horizons of these drugs have a promising future.
Collapse
Affiliation(s)
- Ashutosh M Shukla
- North Florida/South Georgia, Veteran Healthcare System, Gainesville, FL, USA
- Department of Medicine, Division of Nephrology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Aparna Wagle Shukla
- Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
45
|
Quartuccio L, Gandolfo S, Zabotti A, Zandonella Callegher S, Fabro C, De Vita S. Articular and Peripheral Nervous System Involvement Are Linked to the Long-Term Outcome in Primary Sjögren's Syndrome: The Relevance of Single Organ Manifestations Rather Than a Composite Score as Predictors. Front Immunol 2019; 10:1527. [PMID: 31354708 PMCID: PMC6637305 DOI: 10.3389/fimmu.2019.01527] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 06/18/2019] [Indexed: 11/17/2022] Open
Abstract
Objective: The disease course in primary Sjögren's Syndrome (pSS) differs in different subsets of patients. The aim of this study was to clarify whether the pattern of organ involvement may improve the prediction of the very long-term disease outcome. Methods: We collected the data of 255 patients. The total European League Against Rheumatism (EULAR), EULAR Sjögren's Syndrome Disease Activity Index (ESSDAI) score was compared with the pattern of organ involvement, as differentiated by the single ESSDAI domains: (i) at disease diagnosis, and (ii) in the follow-up, by verifying the appearance of new ESSDAI domains and/or the worsening of already active ESSDAI domains. Results: The mean follow-up duration was 9.1 ± 6.9 years. At disease diagnosis, only the articular activity at baseline could predict the long-term outcome of pSS detected at last follow-up visit, being protective in terms of stable or improved disease activity, as measured by ESSDAI [OR 2.9 (1.6–5.4), p = 0.01]. In the follow-up, the onset, and/or worsening of either the peripheral nervous system (PNS) domain (by multivariate and univariate analysis), or the biological domain (only by univariate analysis) correlated with a higher disease activity at the last visit [PNS domain: OR 5.9 (2.4–14.5), p < 0.0001; biological domain: OR 1.9 (1.0–3.8), p = 0.043]. A significantly higher number of patients with articular involvement were taking hydroxychloroquine at the last follow-up visits, if compared with patients without (41/130, 31.5 vs. 13/125, 10.4%, p < 0.0001). Conclusion: Single organ disease manifestations of SS, herein identified as the articular, PNS and biologic involvement, are relevant to predict the very long-term outcome in pSS.
Collapse
Affiliation(s)
- Luca Quartuccio
- Department of Medical Area (DAME), Rheumatology Clinic, University of Udine, Udine, Italy
| | - Saviana Gandolfo
- Department of Medical Area (DAME), Rheumatology Clinic, University of Udine, Udine, Italy
| | - Alen Zabotti
- Department of Medical Area (DAME), Rheumatology Clinic, University of Udine, Udine, Italy
| | | | - Cinzia Fabro
- Department of Medical Area (DAME), Rheumatology Clinic, University of Udine, Udine, Italy
| | - Salvatore De Vita
- Department of Medical Area (DAME), Rheumatology Clinic, University of Udine, Udine, Italy
| |
Collapse
|
46
|
Long-Term Hydroxychloroquine Therapy and Risk of Coronary Artery Disease in Patients with Systemic Lupus Erythematosus. J Clin Med 2019; 8:jcm8060796. [PMID: 31195632 PMCID: PMC6616930 DOI: 10.3390/jcm8060796] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 05/22/2019] [Accepted: 06/02/2019] [Indexed: 11/26/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic systemic inflammatory disease associated with a high prevalence of cardiovascular disease (CVD). Hydroxychloroquine (HCQ) is commonly used to control disease activity in patients with SLE. We evaluated its potential additional therapeutic effect for reducing CVD in SLE patients. We conducted a retrospective cohort study, in which one million participants were sampled from 23 million beneficiaries and data were collected from 2000 to 2013. In total, 826 SLE patients receiving HCQ medication were included after exclusion for previous CVD. The total number of SLE patients was 795 after follow-up for more than one year. After adjusting for chronic comorbidity, a significantly decreased hazard ratio (HR) for coronary artery disease (CAD) was found among SLE patients with a high usage of HCQ for at least 318 days (HR = 0.31, 95% confidence interval, CI: 0.12–0.76). A low HR for CAD was observed in SLE patients with a high cumulative dose of at least 100,267 mg HCQ (HR = 0.25, 95% CI: 0.09–0.66). However, there was no significant lowering of the HR for stroke. Long-term HCQ therapy decreases the HR of CVD in SLE patients. The cardiovascular protective effect of HCQ therapy was associated with decrease in CAD, but not stroke.
Collapse
|
47
|
Shi N, Zhang S, Silverman G, Li M, Cai J, Niu H. Protective effect of hydroxychloroquine on rheumatoid arthritis-associated atherosclerosis. Animal Model Exp Med 2019; 2:98-106. [PMID: 31392302 PMCID: PMC6600633 DOI: 10.1002/ame2.12065] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 03/04/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Patients with rheumatoid arthritis (RA) have an increased risk for cardiovascular disease. We examined the effect of gut microbiota in a mouse model of RA that develops atherosclerosis. METHODS We created three groups of K/BxN female mice that were positive for the anti-glucose-6-phosphate isomerase (GPI) antibody: control diet (CD), high fat diet (HFD), and HFD with hydroxychloroquine (HFD + HCQ). Serological tests were used to detect the serum levels of total cholesterol (TCHO), low-density lipoprotein cholesterol (LDL-C), triglyceride (TG), high-density lipoprotein cholesterol (HDL-C), anti-GPI antibody titers, and serum cytokines. Atherosclerotic plaque was determined by histological analysis, and gut microbiota were determined by 16sV4 sequencing. RESULTS Relative to mice given the CD, those receiving the HFD had increased serum levels of LDL-C, TCHO, and TG, decreased serum levels of HDL-C, increased atherosclerotic lesions in the aortic root, and altered gut microbiota. Addition of HCQ to HFD decreased the serum levels of LDL-C, TCHO, and TG, increased serum levels of HDL-C, and decreased the atherosclerotic lesions in the aortic root. Mice receiving HFD + HCQ also had the greatest bacterial diversity among the three experimental groups. Moreover, HCQ treatment significantly increased the abundance of Akkermansia and Parabacteroides, and decreased the abundance of Clostridium sensu stricto cluster 1, and therefore may be responsible for the reduced RA-associated atherosclerosis and dyslipidemia. CONCLUSION Our mouse model of RA indicated that HFD increased ankle width and aggravated atherosclerosis and dyslipidemia, and that HCQ alleviated the dyslipidemia and atherosclerosis, but had no effect on ankle width.
Collapse
Affiliation(s)
- Na Shi
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS&PUMC), Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese MedicineBeijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Shuangyue Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS&PUMC), Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese MedicineBeijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| | - Gregg Silverman
- Department of Rheumatology, Langone Medical CenterNew York UniversityNew York CityNew York
| | - Mengtao Li
- Peking Union Medical College HospitalBeijingChina
| | - Jun Cai
- Fuwai HospitalChinese Academy of Medical SciencesBeijingChina
| | - Haitao Niu
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS&PUMC), Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese MedicineBeijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
| |
Collapse
|
48
|
Abstract
Introduction: The metabolic syndrome (MetS) is now recognized as a chronic proinflammatory and prothrombotic state that aggravates insulin resistance, oxidative injury, and cardiovascular risk. MetS is more prevalent in patients with systemic lupus erythematosus (SLE), a prototype of systemic autoimmune disease associated with premature atherosclerosis that cannot be accounted by traditional vascular risk factors alone. Dysregulation of the cytokines and adipokines is a common feature in both SLE and MetS, suggesting a complex relationship among autoimmunity, obesity, inflammation, and atherosclerosis. Areas covered: This review summarizes the prevalence of MetS and its effect on cardiovascular outcome and organ damage in patients with SLE. The pathophysiology of MetS and its relevance to SLE is also briefly discussed. Expert opinion: Imbalance of adipokine production in MetS contributes to inflammation and atherosclerosis. MetS predisposes SLE patients to new cardiovascular events and vascular mortality, as well as the development of chronic kidney disease and diabetes mellitus. However, conflicting results have been reported in the literature regarding the levels of the proinflammatory leptin and anti-inflammatory adiponectin, and their relationship with disease activity in SLE patients. While lifestyle modifications and targeting dyslipidemia, hypertension and diabetes mellitus is essential, there is little information on the efficacy and safety of metformin and hydroxychloroquine in alleviating insulin resistance in SLE or MetS. Further research on adipokines in SLE and the role of anti-obesity medications and probiotics in MetS is necessary.
Collapse
Affiliation(s)
- Chi Chiu Mok
- a Department of Medicine , Tuen Mun Hospital , Hong Kong , SAR China
| |
Collapse
|
49
|
Fang C, Luo T, Lin L. Elevation of serum proprotein convertase subtilisin/kexin type 9 (PCSK9) concentrations and its possible atherogenic role in patients with systemic lupus erythematosus. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:452. [PMID: 30603640 DOI: 10.21037/atm.2018.11.04] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Systemic lupus erythematosus (SLE) patients have tendencies of accelerated atherosclerosis (AS) which can only partly be explained by traditional cardiovascular disease (CVD) risk factors. Imbalanced inflammation also plays a vital role. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a new therapeutic target for AS for its dual mechanisms in lipids and inflammation. We aimed to assess serum PCSK9 concentrations in SLE patients and its possible role in atherogenesis of SLE. Methods Ninety SLE patients and 50 healthy controls were included. SLE patients were further divided into SLE-AS and SLE-NonAS subgroups, according to the carotid intima-media thickness (cIMT). Traditional CVD risk factors, inflammatory biomarkers and PCSK9 concentrations were compared between: (I) SLE patients and controls; (II) SLE-AS subgroup and SLE-NonAS subgroup; (III) SLE patients with and without lupus nephritis (LN). Correlational analysis, univariate and multivariate linear regression analysis were applied to analyze the association between PCSK9 levels and disease parameter in SLE patients. Effects on PCSK9 concentrations by monotherapy with hydroxychloroquine (HCQ), which is thought having protective effects against AS in SLE, were investigated by follow-up analysis in 15 SLE patients. Results We found that SLE patients had significantly elevated serum PCSK9 levels than controls, especially in SLE-As subgroup or those with LN, accompanied with higher ratio of cIMT thickening. Correlational analysis showed PCSK9 concentrations correlated with C-reactive protein (CRP) levels, age and erythrocyte sedimentation rate (ESR). Univariate and multivariate linear regression revealed that only CRP, but not age or ESR was positive predictors of PCSK9. Interestingly, monotherapy with HCQ for three months significantly reduced PCSK9 and CRP levels in inactive SLE patients. Conclusions Our results suggested that elevated PCSK9 levels in SLE are probably associated with atherogenic inflammation in SLE. HCQ, which is thought having protective effects against AS in SLE, can effectively reduce PCSK9 levels in SLE patients.
Collapse
Affiliation(s)
- Chenglong Fang
- Department of Rheumatology, Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Tingting Luo
- Department of Ultrasonic Cardiogram, Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Ling Lin
- Department of Rheumatology, Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| |
Collapse
|
50
|
Mangoni AA, Tommasi S, Zinellu A, Sotgia S, Carru C, Piga M, Erre GL. Repurposing existing drugs for cardiovascular risk management: a focus on methotrexate. Drugs Context 2018; 7:212557. [PMID: 30459819 PMCID: PMC6239018 DOI: 10.7573/dic.212557] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/25/2018] [Accepted: 10/26/2018] [Indexed: 12/12/2022] Open
Abstract
About 20% of patients with a history of atherosclerotic cardiovascular disease will experience further cardiovascular events despite maximal pharmacological treatment with cardioprotective drugs. This highlights the presence of residual cardiovascular risk in a significant proportion of patients and the need for novel, more effective therapies. These therapies should ideally target different pathophysiological pathways involved in the onset and the progression of atherosclerosis, particularly the inflammatory and immune pathways. Methotrexate is a first-line disease-modifying antirheumatic drug that is widely used for the management of autoimmune and chronic inflammatory disorders. There is some in vitro and in vivo evidence that methotrexate might exert a unique combination of anti-inflammatory, blood pressure lowering, and vasculoprotective effects. Pending the results of large prospective studies investigating surrogate end-points as well as morbidity and mortality, repurposing methotrexate for cardiovascular risk management might represent a cost-effective strategy with immediate public health benefits. This review discusses the current challenges in the management of cardiovascular disease; the available evidence on the effects of methotrexate on inflammation, blood pressure, and surrogate markers of arterial function; suggestions for future research directions; and practical considerations with the use of methotrexate in this context.
Collapse
Affiliation(s)
- Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, Australia
| | - Sara Tommasi
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, Australia
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Salvatore Sotgia
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Quality Control Unit, University Hospital (AOUSS), Sassari, Italy
| | - Matteo Piga
- Rheumatology Unit, University Clinic and AOU of Cagliari, Italy
| | - Gian Luca Erre
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University Hospital (AOUSS) and University of Sassari, Sassari, Italy
| |
Collapse
|