1
|
Saliba F, Khattar G, Mourad O, Aoun L, Bou Sanayeh E, Arafa F, Al Saidi I, Abidor E, Al Achkar M, Rizvi T, Sangaraju K, Di Pietro G, Haddadin F, Almardini S, El Gharib K, El-Hage H. Evaluating the impact of type 2 diabetes mellitus on interstitial lung disease prevalence in patients with systemic lupus erythematosus: A national inpatient sample analysis. Lupus 2024; 33:1547-1555. [PMID: 39401267 DOI: 10.1177/09612033241292162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2024]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) increases the risk of interstitial lung disease (ILD). SLE is also linked to an elevated risk of type 2 diabetes mellitus (T2DM). However, the impact of T2DM on ILD risk in patients with SLE is still unclear. This study aimed to compare the prevalence of ILD in patients with SLE based on the presence of T2DM (SLE + T2DM+) or its absence (SLE + T2DM-). METHODS This was a retrospective cohort study using the 2019-2020 National Inpatient Sample database. Adult SLE patients were identified and stratified by T2DM status. Comparable cohorts were created using propensity score matching, resulting in 10,532 patients in each cohort. Multivariate logistic regression assessed the association between T2DM and ILD. RESULTS T2DM was associated with a lower prevalence of ILD in patients with SLE (OR 0.798, 95% CI: 0.695-0.918, p = .002), occurring in 371 (3.5%) patients with T2DM compared to 463 (4.4%) patients without T2DM. Specifically, this difference was mainly driven by pulmonary fibrosis, which was significantly less frequent in the T2DM group (1.3% vs 1.8%, OR 0.7, 95% CI: 0.560-0.875, p = .002). No differences were found in secondary outcomes, including death rates, length of hospital stay, ARDS, pneumothorax, pleural effusion, or pulmonary arterial hypertension. CONCLUSION Our study suggests that T2DM significantly reduced ILD risk in patients with SLE, specifically diminishing pulmonary fibrosis prevalence. Further research should explore mechanisms for this protective association between T2DM and ILD development in SLE. These findings may guide management strategies for this vulnerable population.
Collapse
Affiliation(s)
- Fares Saliba
- Department of Internal Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Georges Khattar
- Department of Internal Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Omar Mourad
- Department of Internal Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Laurence Aoun
- Department of Internal Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Elie Bou Sanayeh
- Department of Internal Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Fatema Arafa
- Department of Internal Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Ibrahim Al Saidi
- Department of Internal Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Erica Abidor
- Department of Internal Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Michel Al Achkar
- Department of Internal Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Taqi Rizvi
- Department of Internal Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Koushik Sangaraju
- Department of Internal Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Gaetano Di Pietro
- Department of Internal Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Fadi Haddadin
- Department of Internal Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Shaza Almardini
- Department of Internal Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Khalil El Gharib
- Department of Internal Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Halim El-Hage
- Department of Pulmonary and Critical Care Medicine, Staten Island University Hospital, Staten Island, NY, USA
| |
Collapse
|
2
|
Reis-Neto ETD, Seguro LPC, Sato EI, Borba EF, Klumb EM, Costallat LTL, Medeiros MMDC, Bonfá E, Araújo NC, Appenzeller S, Montandon ACDOES, Yuki EFN, Teixeira RCDA, Telles RW, Egypto DCSD, Ribeiro FM, Gasparin AA, Junior ASDA, Neiva CLS, Calderaro DC, Monticielo OA. II Brazilian Society of Rheumatology consensus for lupus nephritis diagnosis and treatment. Adv Rheumatol 2024; 64:48. [PMID: 38890752 DOI: 10.1186/s42358-024-00386-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 05/25/2024] [Indexed: 06/20/2024] Open
Abstract
OBJECTIVE To develop the second evidence-based Brazilian Society of Rheumatology consensus for diagnosis and treatment of lupus nephritis (LN). METHODS Two methodologists and 20 rheumatologists from Lupus Comittee of Brazilian Society of Rheumatology participate in the development of this guideline. Fourteen PICO questions were defined and a systematic review was performed. Eligible randomized controlled trials were analyzed regarding complete renal remission, partial renal remission, serum creatinine, proteinuria, serum creatinine doubling, progression to end-stage renal disease, renal relapse, and severe adverse events (infections and mortality). The Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach was used to develop these recommendations. Recommendations required ≥82% of agreement among the voting members and were classified as strongly in favor, weakly in favor, conditional, weakly against or strongly against a particular intervention. Other aspects of LN management (diagnosis, general principles of treatment, treatment of comorbidities and refractory cases) were evaluated through literature review and expert opinion. RESULTS All SLE patients should undergo creatinine and urinalysis tests to assess renal involvement. Kidney biopsy is considered the gold standard for diagnosing LN but, if it is not available or there is a contraindication to the procedure, therapeutic decisions should be based on clinical and laboratory parameters. Fourteen recommendations were developed. Target Renal response (TRR) was defined as improvement or maintenance of renal function (±10% at baseline of treatment) combined with a decrease in 24-h proteinuria or 24-h UPCR of 25% at 3 months, a decrease of 50% at 6 months, and proteinuria < 0.8 g/24 h at 12 months. Hydroxychloroquine should be prescribed to all SLE patients, except in cases of contraindication. Glucocorticoids should be used at the lowest dose and for the minimal necessary period. In class III or IV (±V), mycophenolate (MMF), cyclophosphamide, MMF plus tacrolimus (TAC), MMF plus belimumab or TAC can be used as induction therapy. For maintenance therapy, MMF or azathioprine (AZA) are the first choice and TAC or cyclosporin or leflunomide can be used in patients who cannot use MMF or AZA. Rituximab can be prescribed in cases of refractory disease. In cases of failure in achieving TRR, it is important to assess adherence, immunosuppressant dosage, adjuvant therapy, comorbidities, and consider biopsy/rebiopsy. CONCLUSION This consensus provides evidence-based data to guide LN diagnosis and treatment, supporting the development of public and supplementary health policies in Brazil.
Collapse
Affiliation(s)
- Edgard Torres Dos Reis-Neto
- Division of Rheumatology, Department of Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM/Unifesp), Otonis Street, 863, 2 Floor, Vila Clementino, São Paulo, SP, 04025-002, Brazil.
| | - Luciana Parente Costa Seguro
- Division of Rheumatology, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Emília Inoue Sato
- Division of Rheumatology, Department of Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM/Unifesp), Otonis Street, 863, 2 Floor, Vila Clementino, São Paulo, SP, 04025-002, Brazil
| | - Eduardo Ferreira Borba
- Division of Rheumatology, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Evandro Mendes Klumb
- Department of Rheumatology, Hospital Universitário Pedro Ernesto, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lilian Tereza Lavras Costallat
- Division of Rheumatology, Department of Orthopedics, Rheumatology and Traumatology, Universidade Estadual de Campinas (Unicamp), Campinas, Brazil
| | | | - Eloisa Bonfá
- Division of Rheumatology, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Nafice Costa Araújo
- Division of Rheumatology, Hospital do Servidor Público Estadual de São Paulo - Instituto de Assistência Médica ao Servidor Público Estadual de São Paulo, São Paulo, Brazil
| | - Simone Appenzeller
- Division of Rheumatology, Department of Orthopedics, Rheumatology and Traumatology, Universidade Estadual de Campinas (Unicamp), Campinas, Brazil
| | | | - Emily Figueiredo Neves Yuki
- Division of Rheumatology, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | | | - Rosa Weiss Telles
- Division of Rheumatology, Faculdade de Medicina da Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | - Francinne Machado Ribeiro
- Department of Rheumatology, Hospital Universitário Pedro Ernesto, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andrese Aline Gasparin
- Division of Rheumatology, Department of Internal Medicine, Hospital de Clínicas de Porto Alegre, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Antonio Silaide de Araujo Junior
- Division of Rheumatology, Department of Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM/Unifesp), Otonis Street, 863, 2 Floor, Vila Clementino, São Paulo, SP, 04025-002, Brazil
| | | | - Debora Cerqueira Calderaro
- Division of Rheumatology, Faculdade de Medicina da Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Odirlei Andre Monticielo
- Division of Rheumatology, Department of Internal Medicine, Hospital de Clínicas de Porto Alegre, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| |
Collapse
|
3
|
Abstract
Vascular disease is a major cause of morbidity and mortality in patients with systemic autoimmune diseases, particularly systemic lupus erythematosus (SLE). Although comorbid cardiovascular risk factors are frequently present in patients with SLE, they do not explain the high burden of premature vascular disease. Profound innate and adaptive immune dysregulation seems to be the primary driver of accelerated vascular damage in SLE. In particular, evidence suggests that dysregulation of type 1 interferon (IFN-I) and aberrant neutrophils have key roles in the pathogenesis of vascular damage. IFN-I promotes endothelial dysfunction directly via effects on endothelial cells and indirectly via priming of immune cells that contribute to vascular damage. SLE neutrophils are vasculopathic in part because of their increased ability to form immunostimulatory neutrophil extracellular traps. Despite improvements in clinical care, cardiovascular disease remains the leading cause of mortality among patients with SLE, and treatments that improve vascular outcomes are urgently needed. Improved understanding of the mechanisms of vascular injury in inflammatory conditions such as SLE could also have implications for common cardiovascular diseases, such as atherosclerosis and hypertension, and may ultimately lead to personalized therapeutic approaches to the prevention and treatment of this potentially fatal complication.
Collapse
Affiliation(s)
- William G Ambler
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
4
|
Venturelli V, Abrantes AM, Rahman A, Isenberg DA. The impact of antiphospholipid antibodies/antiphospholipid syndrome on systemic lupus erythematosus. Rheumatology (Oxford) 2024; 63:SI72-SI85. [PMID: 38320586 DOI: 10.1093/rheumatology/kead618] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/31/2023] [Indexed: 02/08/2024] Open
Abstract
aPLs are a major determinant of the increased cardiovascular risk in patients with SLE. They adversely affect clinical manifestations, damage accrual and prognosis. Apart from the antibodies included in the 2006 revised classification criteria for APS, other non-classical aPLs might help in identifying SLE patients at increased risk of thrombotic events. The best studied are IgA anti-β2-glycoprotein I, anti-domain I β2-glycoprotein I and aPS-PT. Major organ involvement includes kidney and neuropsychiatric systems. aPL/APS severely impacts pregnancy outcomes. Due to increased thrombotic risk, these patients require aggressive cardiovascular risk factor control. Primary prophylaxis is based on low-dose aspirin in high-risk patients. Warfarin is the gold-standard drug for secondary prophylaxis.
Collapse
Affiliation(s)
- Veronica Venturelli
- Rheumatology Unit, Department of Medical Sciences, Università degli Studi di Ferrara, Azienda Ospedaliero-Universitaria S. Anna, Cona, Italy
| | - Ana Mafalda Abrantes
- Division of Internal Medicine II, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
- Instituto de Semiótica Clínica, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Anisur Rahman
- Centre for Rheumatology, Department of Medicine, University College London, London, UK
| | - David A Isenberg
- Centre for Rheumatology, Department of Medicine, University College London, London, UK
| |
Collapse
|
5
|
Mokgopa KP, Lobb KA, Tshiwawa T. A Computational Study of Green Tea Extracts and their Derivatives as Potential Inhibitors for Squalene Monooxygenase. Med Chem 2024; 20:721-732. [PMID: 38584555 DOI: 10.2174/0115734064280290240211170037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/09/2024] [Accepted: 01/24/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND According to the World Health Organisation, cardiovascular complications have been recognized as the leading course of death between 2000 and 2019. Cardiovascular complications are caused by excess LDL cholesterol in the body or arteries that can build up to form a plaque. There are drugs currently in clinical use called statins that target HMGCoA reductase. However, these drugs result in several side effects. This work investigated using computational approaches to lower cholesterol by investigating green tea extracts as an inhibitors for squalene monooxygenase (the second-rate-controlling step in cholesterol synthesis). METHODS Pharmacophore modeling was done to identify possible pharmacophoric sites based on the pIC50 values. The best hypothesis generated by pharmacophore modeling was further validated by atom-based 3D QSAR, where 70% of the data set was treated as the training set. Prior molecular docking ADMET studies were done to investigate the physiochemical properties of these molecules. Glide docking was performed, followed by molecular dynamics to evaluate the protein conformational changes. RESULTS Pharmacophore results suggest that the best molecules to interact with the biological target should have at least one hydrogen acceptor (A5), two hydrogen donors (D9 and D10), and two benzene rings (R14 and R15) for green tea polyphenols and theasinensin A. ADMET result shows that all molecules in this class have low oral adsorption. Molecular docking results showed that some green tea polyphenols have good binding affinities, with most of these structures having a docking score of less than -10 kcal/mol. Molecular dynamics further illustrated that the best-docked ligands perfectly stay within the active site over a 100 ns simulation. CONCLUSION The results obtained from this study suggest that green tea polyphenols have the potential for inhibition of squalene monooxygenase, except for theasinensin A.
Collapse
Affiliation(s)
| | - Kevin A Lobb
- Department of Chemistry, Rhodes University, Makhanda, South Africa
- Research Unit in Bioinformatics (RUBi), Rhodes University, Makhanda, 6140, South Africa
| | | |
Collapse
|
6
|
Garg S, Bartels CM, Bao G, Helmick CG, Drenkard C, Lim SS. Timing and Predictors of Incident Cardiovascular Disease in Systemic Lupus Erythematosus: Risk Occurs Early and Highlights Racial Disparities. J Rheumatol 2023; 50:84-92. [PMID: 35914786 PMCID: PMC10773489 DOI: 10.3899/jrheum.220279] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2022] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) affects Black people 2 to 3 times more frequently than non-Black people and is associated with higher morbidity and mortality. In total, 4 studies with predominantly non-Black SLE cohorts highlighted that cardiovascular disease (CVD) is no longer primarily a late complication of SLE. This study assessed the timing and predictors of incident CVD in a predominantly Black population-based SLE cohort. METHODS Incident SLE cases from the population-based Georgia Lupus Registry were validated as having a CVD event through review of medical records and matching with the Georgia Hospital Discharge Database and the National Death Index. The surveillance period for an incident CVD event spanned a 15-year period, starting from 2 years prior to SLE diagnosis. RESULTS Among 336 people with SLE, 253 (75%) were Black and 56 (17%) had an incident CVD event. The frequency of CVD events peaked in years 2 and 11 after SLE diagnosis. There was a 7-fold higher risk of incident CVD over the entire 15-year period; this risk was 19-fold higher in the first 12 years in Black people as compared to non-Black people with SLE. Black people with SLE (P < 0.001) and those with discoid rash (hazard ratio 3.2, 95% CI 1.4-7.1) had a higher risk of incident CVD events. CONCLUSION The frequency of incident CVD events peaked in years 2 and 11 after SLE diagnosis. Being Black or having a discoid rash were strong predictors of an incident CVD event. Surveillance for CVD and preventive interventions, directed particularly toward Black people with recent SLE diagnoses, are needed to reduce racial disparities.
Collapse
Affiliation(s)
- Shivani Garg
- S. Garg, MD, MS, Assistant Professor, C.M. Bartels, MD, MS, Associate Professor, Rheumatology Division, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin;
| | - Christie M Bartels
- S. Garg, MD, MS, Assistant Professor, C.M. Bartels, MD, MS, Associate Professor, Rheumatology Division, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Gaobin Bao
- G. Bao, MPH, Senior Statistician, Division of Rheumatology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Charles G Helmick
- C.G. Helmick, MD, Professor, Division of Population Health, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Cristina Drenkard
- C. Drenkard, MD, PhD, Associate Professor, S.S. Lim, MD, MPH, Professor, Division of Rheumatology, Department of Medicine, Emory University School of Medicine, and Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, Georgia, USA
| | - S Sam Lim
- C. Drenkard, MD, PhD, Associate Professor, S.S. Lim, MD, MPH, Professor, Division of Rheumatology, Department of Medicine, Emory University School of Medicine, and Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, Georgia, USA
| |
Collapse
|
7
|
Garcia-de los Ríos C, Medina-Casado M, Díaz-Chamorro A, Sierras-Jiménez M, Lardelli-Claret P, Cáliz-Cáliz R, Sabio JM. Sclerostin as a biomarker of cardiovascular risk in women with systemic lupus erythematosus. Sci Rep 2022; 12:21621. [PMID: 36517533 PMCID: PMC9749620 DOI: 10.1038/s41598-022-25651-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 12/02/2022] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular disease is one of the main causes of death in patients with systemic lupus erythematosus (SLE). On the other hand, sclerostin is a reliable and early biomarker of vascular calcification. This study aimed to estimate the association between sclerostin and two markers of cardiovascular risk, carotid atherosclerotic plaque (CP) and carotid-femoral pulse wave velocity (PWV), in women with SLE. The presence of CP (determined by carotid artery ultrasound) and PWV were measured in 68 women with SLE and preserved renal function. None of the participants had a history of cardiovascular disease. Serum levels of sclerostin were determined using the ELISA method. Other factors associated with increased cardiovascular risk were also measured. The association between sclerostin, CP and PWV was assessed using Receiver Operating Characteristic (ROC) curves and multivariate regression models. The area under the ROC curve was 0.785 (95% confidence interval [CI] 0.662-0.871) for CP and 0.834 (95% CI 0.729-0.916) for dichotomized PWV. After adjusting for other cardiovascular risk factors, it was found that a 10-units increase in sclerostin values was associated with a 44% increase in the odds of CP (95% CI 1-105), but no adjusted association was observed between sclerostin and PWV. Predictive models included age (for both outcomes), hypertension, Framingham risk score and C-reactive protein (for PWV), but not sclerostin. Sclerostin is associated with the presence of CP in women with SLE. Further research should confirm its possible role as a biomarker of cardiovascular risk in these patients.
Collapse
Affiliation(s)
- Carlos Garcia-de los Ríos
- grid.411380.f0000 0000 8771 3783Department of Internal Medicine, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | | | - Antonio Díaz-Chamorro
- grid.411380.f0000 0000 8771 3783Department of Internal Medicine, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - María Sierras-Jiménez
- grid.411380.f0000 0000 8771 3783Emergency Department, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Pablo Lardelli-Claret
- grid.4489.10000000121678994Department of Preventive Medicine and Public Health, University of Granada, Granada, Spain ,Centros de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain ,grid.507088.2Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain
| | - Rafael Cáliz-Cáliz
- grid.411380.f0000 0000 8771 3783Department of Rheumatology, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - José Mario Sabio
- grid.411380.f0000 0000 8771 3783Systemic Autoimmune Diseases Unit, Department of Internal Medicine, 9th Floor, Hospital Universitario Virgen de las Nieves, Avda. Fuerzas Armadas, Nº 2, 18014 Granada, Spain
| |
Collapse
|
8
|
Guzmán-Martínez G, Marañón C. Immune mechanisms associated with cardiovascular disease in systemic lupus erythematosus: A path to potential biomarkers. Front Immunol 2022; 13:974826. [PMID: 36420265 PMCID: PMC9677819 DOI: 10.3389/fimmu.2022.974826] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/13/2022] [Indexed: 10/29/2023] Open
Abstract
Systemic lupus erythematosus (SLE) patients display an increased risk of cardiovascular disease (CVD). With the improved clinical management of other classical severe manifestation of the disease, CVD is becoming one of the most relevant complications of SLE, and it is an important factor causing morbidity and mortality. Several immune constituents have been shown to be involved in the pathogenesis of atherosclerosis and endothelial damage in SLE patients, including specific circulating cell populations, autoantibodies, and inflammatory mediators. In this review, we summarize the presentation of CVD in SLE and the role of the autoimmune responses present in SLE patients in the induction of atherogenesis, endothelial impairment and cardiac disease. Additionally, we discuss the utility of these immune mediators as early CVD biomarkers and targets for clinical intervention in SLE patients.
Collapse
Affiliation(s)
- Gabriela Guzmán-Martínez
- Atrys Health, Madrid, Spain
- Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
- Department of Cardiology, La Paz University Hospital, IdiPaz, Madrid, Spain
| | - Concepción Marañón
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| |
Collapse
|
9
|
Hasni S, Temesgen-Oyelakin Y, Davis M, Chu J, Poncio E, Naqi M, Gupta S, Wang X, Oliveira C, Claybaugh D, Dey A, Lu S, Carlucci P, Purmalek M, Manna ZG, Shi Y, Ochoa-Navas I, Chen J, Mukherjee A, Han KL, Cheung F, Koroleva G, Belkaid Y, Tsang JS, Apps R, Thomas DE, Heller T, Gadina M, Playford MP, Li X, Mehta NN, Kaplan MJ. Peroxisome proliferator activated receptor-γ agonist pioglitazone improves vascular and metabolic dysfunction in systemic lupus erythematosus. Ann Rheum Dis 2022; 81:1576-1584. [PMID: 35914929 PMCID: PMC9606512 DOI: 10.1136/ard-2022-222658] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/08/2022] [Indexed: 11/18/2022]
Abstract
OBJECTIVES Premature cardiovascular events in systemic lupus erythematosus (SLE) contribute to morbidity and mortality, with no effective preventive strategies described to date. Immune dysregulation and metabolic disturbances appear to play prominent roles in the induction of vascular disease in SLE. The peroxisome proliferator activated receptor-gamma agonist pioglitazone (PGZ suppresses vascular damage and immune dysregulation in murine lupus and improves endothelial dysfunction in other inflammatory diseases. We hypothesised that PGZ could improve vascular dysfunction and cardiometabolic parameters in SLE. METHODS Eighty SLE subjects with mild to severe disease activity were randomised to a sequence of PGZ followed by placebo for 3 months, or vice versa, in a double-blind, cross-over design with a 2-month wash-out period. Primary endpoints were parameters of endothelial function and arterial inflammation, measured by multimodal assessments. Additional outcome measures of disease activity, neutrophil dysregulation, metabolic disturbances and gene expression studies were performed. RESULTS Seventy-two subjects completed the study. PGZ was associated with a significant reduction in Cardio-Ankle Vascular Index (a measure of arterial stiffness) compared with placebo. Various metabolic parameters improved with PGZ, including insulin resistance and lipoprotein profiles. Circulating neutrophil extracellular trap levels also significantly decreased with PGZ compared with placebo. Most adverse events experienced while on PGZ were mild and resolved with reduction in PGZ dose. CONCLUSION PGZ was well tolerated and induced significant improvement in vascular stiffness and cardiometabolic parameters in SLE. The results suggest that PGZ should be further explored as a modulator of cardiovascular disease risk in SLE. TRIAL REGISTRATION NUMBER NCT02338999.
Collapse
Affiliation(s)
- Sarfaraz Hasni
- Lupus Clinical Trials Unit, Office of the Clinical Director, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Yenealem Temesgen-Oyelakin
- Lupus Clinical Trials Unit, Office of the Clinical Director, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Michael Davis
- Lupus Clinical Trials Unit, Office of the Clinical Director, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jun Chu
- Lupus Clinical Trials Unit, Office of the Clinical Director, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Elaine Poncio
- Lupus Clinical Trials Unit, Office of the Clinical Director, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Mohammad Naqi
- Lupus Clinical Trials Unit, Office of the Clinical Director, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sarthak Gupta
- Systemic Autoimmunity Branch/NIAMS, National Institutes of Health, Bethesda, Maryland, USA
| | - Xinghao Wang
- Systemic Autoimmunity Branch/NIAMS, National Institutes of Health, Bethesda, Maryland, USA
| | - Christopher Oliveira
- Systemic Autoimmunity Branch/NIAMS, National Institutes of Health, Bethesda, Maryland, USA
| | - Dillon Claybaugh
- Systemic Autoimmunity Branch/NIAMS, National Institutes of Health, Bethesda, Maryland, USA
| | - Amit Dey
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Shajia Lu
- Translational Immunology Section, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA
| | - Philip Carlucci
- Systemic Autoimmunity Branch/NIAMS, National Institutes of Health, Bethesda, Maryland, USA
| | - Monica Purmalek
- Systemic Autoimmunity Branch/NIAMS, National Institutes of Health, Bethesda, Maryland, USA
| | - Zerai G Manna
- Lupus Clinical Trials Unit, Office of the Clinical Director, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Yinghui Shi
- Office of the Clinical Director, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA
| | - Isabel Ochoa-Navas
- Lupus Clinical Trials Unit, Office of the Clinical Director, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jinguo Chen
- National Institutes of Health, Bethesda, Maryland, USA
| | | | - Kyu Lee Han
- National Institutes of Health, Bethesda, Maryland, USA
| | - Foo Cheung
- National Institutes of Health, Bethesda, Maryland, USA
| | | | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - John S Tsang
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, National Institutes of Health, Bethesda, Maryland, USA
- NIH Center for Human Immunology, National Institutes of Health, Bethesda, Maryland, USA
| | - Richard Apps
- National Institutes of Health, Bethesda, Maryland, USA
| | - Donald E Thomas
- Arthritis and Pain Associates of PG County, Greenbelt, Maryland, USA
| | - Theo Heller
- NIDDK, National Institutes of Health, Bethesda, Maryland, USA
| | - Massimo Gadina
- NIAMS, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Xiaobai Li
- Biostatistics and Clinical Epidemiology Service, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Nehal N Mehta
- National Institutes of Health, Bethesda, Maryland, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch/NIAMS, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
10
|
Ciurtin C, Pineda-Torra I, Jury EC, Robinson GA. CD8+ T-Cells in Juvenile-Onset SLE: From Pathogenesis to Comorbidities. Front Med (Lausanne) 2022; 9:904435. [PMID: 35801216 PMCID: PMC9254716 DOI: 10.3389/fmed.2022.904435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/26/2022] [Indexed: 11/25/2022] Open
Abstract
Diagnosis of systemic lupus erythematosus (SLE) in childhood [juvenile-onset (J) SLE], results in a more severe disease phenotype including major organ involvement, increased organ damage, cardiovascular disease risk and mortality compared to adult-onset SLE. Investigating early disease course in these younger JSLE patients could allow for timely intervention to improve long-term prognosis. However, precise mechanisms of pathogenesis are yet to be elucidated. Recently, CD8+ T-cells have emerged as a key pathogenic immune subset in JSLE, which are increased in patients compared to healthy individuals and associated with more active disease and organ involvement over time. CD8+ T-cell subsets have also been used to predict disease prognosis in adult-onset SLE, supporting the importance of studying this cell population in SLE across age. Recently, single-cell approaches have allowed for more detailed analysis of immune subsets in JSLE, where type-I IFN-signatures have been identified in CD8+ T-cells expressing high levels of granzyme K. In addition, JSLE patients with an increased cardiometabolic risk have increased CD8+ T-cells with elevated type-I IFN-signaling, activation and apoptotic pathways associated with atherosclerosis. Here we review the current evidence surrounding CD8+ T-cell dysregulation in JSLE and therapeutic strategies that could be used to reduce CD8+ T-cell inflammation to improve disease prognosis.
Collapse
Affiliation(s)
- Coziana Ciurtin
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, United Kingdom
| | - Ines Pineda-Torra
- Centre for Cardiometabolic and Vascular Science, Division of Medicine, University College London, London, United Kingdom
| | - Elizabeth C. Jury
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom
| | - George A. Robinson
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, United Kingdom
| |
Collapse
|
11
|
Robinson GA, Pineda-Torra I, Ciurtin C, Jury EC. Sex Differences in Lipid Metabolism: Implications for Systemic Lupus Erythematosus and Cardiovascular Disease Risk. Front Med (Lausanne) 2022; 9:914016. [PMID: 35712086 PMCID: PMC9197418 DOI: 10.3389/fmed.2022.914016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/02/2022] [Indexed: 11/13/2022] Open
Abstract
It is known that healthy women during childbearing years have a lower risk of cardiovascular disease (CVD) and coronary heart disease compared to age matched men. Various traditional risk factors have been shown to confer differential CVD susceptibilities by sex. Atherosclerosis is a major cause of CVD and mortality and sex differences in CVD risk could be due to reduced atherogenic low and very low-density lipoproteins (LDL and VLDL) and increased atheroprotective high density lipoproteins (HDLs) in women. In contrast, patients with systemic lupus erythematosus (SLE), a chronic inflammatory disease that predominately affects women, have an increased atherosclerotic and CVD risk. This increased CVD risk is largely associated with dyslipidaemia, the imbalance of atherogenic and atheroprotective lipoproteins, a conventional CVD risk factor. In many women with SLE, dyslipidaemia is characterised by elevated LDL and reduced HDL, eradicating the sex-specific CVD protection observed in healthy women compared to men. This review will explore this paradox, reporting what is known regarding sex differences in lipid metabolism and CVD risk in the healthy population and transgender individuals undergoing cross-sex hormone therapy, and provide evidence for how these differences may be compromised in an autoimmune inflammatory disease setting. This could lead to better understanding of mechanistic changes in lipid metabolism driving the increased CVD risk by sex and in autoimmunity and highlight potential therapeutic targets to help reduce this risk.
Collapse
Affiliation(s)
- George A. Robinson
- Division of Medicine, Centre for Rheumatology Research, University College London, London, United Kingdom
- Division of Medicine, Centre for Adolescent Rheumatology Versus Arthritis, University College London, London, United Kingdom
| | - Ines Pineda-Torra
- Division of Medicine, Centre for Cardiometabolic and Vascular Science, University College London, London, United Kingdom
| | - Coziana Ciurtin
- Division of Medicine, Centre for Rheumatology Research, University College London, London, United Kingdom
- Division of Medicine, Centre for Adolescent Rheumatology Versus Arthritis, University College London, London, United Kingdom
| | - Elizabeth C. Jury
- Division of Medicine, Centre for Rheumatology Research, University College London, London, United Kingdom
| |
Collapse
|
12
|
Oliveira CB, Kaplan MJ. Cardiovascular disease risk and pathogenesis in systemic lupus erythematosus. Semin Immunopathol 2022; 44:309-324. [PMID: 35355124 PMCID: PMC9064999 DOI: 10.1007/s00281-022-00922-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/08/2022] [Indexed: 02/06/2023]
Abstract
Systemic lupus erythematosus (SLE) often features extensive cardiovascular (CV) comorbidity and patients with SLE are at significantly increased risk of CV event occurrence and CV-related mortality. While the specific mechanisms leading to this increased cardiovascular disease (CVD) risk remain to be fully characterized, this heightened risk cannot be fully explained by traditional CV risk factors and is likely driven by immunologic and inflammatory features of SLE. Widespread innate and adaptive immune dysregulation characterize SLE, and factors including excessive type I interferon burden, inappropriate formation and ineffective clearance of neutrophil extracellular traps, and autoantibody formation have been linked to clinical and metabolic features impacting CV risk in SLE and may represent pathogenic drivers of SLE-related CVD. Indeed, functional and phenotypic aberrations in almost every immune cell type are present in SLE and may impact CVD progression. As understanding of the contribution of SLE-specific factors to CVD in SLE improves, improved screening and monitoring of CV risk alongside development of therapeutic treatments aimed at prevention of CVD in SLE patients are required and remain the focus of several ongoing studies and lines of inquiry.
Collapse
Affiliation(s)
- Christopher B Oliveira
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 10 Center Drive, 12N248C, Bethesda, MD, 20892, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 10 Center Drive, 12N248C, Bethesda, MD, 20892, USA.
| |
Collapse
|
13
|
Oliveira CB, Kaplan MJ. Cardiovascular disease risk and pathogenesis in systemic lupus erythematosus. Semin Immunopathol 2022. [PMID: 35355124 DOI: 10.1007/s00281-02200922-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Systemic lupus erythematosus (SLE) often features extensive cardiovascular (CV) comorbidity and patients with SLE are at significantly increased risk of CV event occurrence and CV-related mortality. While the specific mechanisms leading to this increased cardiovascular disease (CVD) risk remain to be fully characterized, this heightened risk cannot be fully explained by traditional CV risk factors and is likely driven by immunologic and inflammatory features of SLE. Widespread innate and adaptive immune dysregulation characterize SLE, and factors including excessive type I interferon burden, inappropriate formation and ineffective clearance of neutrophil extracellular traps, and autoantibody formation have been linked to clinical and metabolic features impacting CV risk in SLE and may represent pathogenic drivers of SLE-related CVD. Indeed, functional and phenotypic aberrations in almost every immune cell type are present in SLE and may impact CVD progression. As understanding of the contribution of SLE-specific factors to CVD in SLE improves, improved screening and monitoring of CV risk alongside development of therapeutic treatments aimed at prevention of CVD in SLE patients are required and remain the focus of several ongoing studies and lines of inquiry.
Collapse
Affiliation(s)
- Christopher B Oliveira
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 10 Center Drive, 12N248C, Bethesda, MD, 20892, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 10 Center Drive, 12N248C, Bethesda, MD, 20892, USA.
| |
Collapse
|
14
|
Whittier M, Bautista Sanchez R, Arora S, Manadan AM. Systemic Lupus Erythematosus and Antiphospholipid Antibody Syndrome as Risk Factors for Acute Coronary Syndrome in Young Patients: Analysis of the National Inpatient Sample. J Clin Rheumatol 2022; 28:143-146. [PMID: 35293887 DOI: 10.1097/rhu.0000000000001824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE This study aimed to compare the odds of acute coronary syndrome (ACS) in patients aged 18 to 40 years to patients older than 40 years with and without secondary diagnoses of systemic lupus erythematosus (SLE) or antiphospholipid antibody syndrome (APLS) while controlling for traditional cardiovascular (CV) risk factors. METHODS Data were extracted from the National Inpatient Sample database from 2016 to 2018. The National Inpatient Sample was searched for hospitalizations of adult patients with ACS as the principal diagnosis, with and without SLE or APLS as secondary diagnoses. Age was divided categorically into 2 groups: adults aged 18 to 40 years and those older than 40 years. The primary outcome was the development of ACS. Multivariate logistic regression analyses were used to adjust for confounders. RESULTS There were 90,879,561 hospital discharges in the 2016 to 2018 database. Of those, 55,050 between the ages of 18 to 40 years and 1,966,234 aged older than 40 years were hospitalized with a principal diagnosis of ACS. Traditional CV risk factors were associated with ACS hospitalizations in both age groups. In multivariate analysis of the 18 to 40 years age group, both SLE (odds ratio, 2.18; 95% confidence interval, 1.814-2.625) and APLS (odds ratio, 2.18; 95% confidence interval, 1.546-3.087) were strongly associated with ACS hospitalizations. After the age of 40 years, there were no increased odds of ACS hospitalizations for SLE or APLS. CONCLUSIONS In the younger population, SLE and APLS were strongly associated with ACS hospitalizations in addition to the traditional CV risk factors. In the older age group, traditional CV risk factors dominated and diluted the effect of SLE and APLS.
Collapse
|
15
|
Huang YJ, Chen JS, Luo SF, Kuo CF. Comparison of Indexes to Measure Comorbidity Burden and Predict All-Cause Mortality in Rheumatoid Arthritis. J Clin Med 2021; 10:jcm10225460. [PMID: 34830741 PMCID: PMC8618526 DOI: 10.3390/jcm10225460] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/04/2021] [Accepted: 11/11/2021] [Indexed: 11/20/2022] Open
Abstract
Objectives: To examine the comorbidity burden in patients with rheumatoid arthritis (RA) patients using a nationwide population-based cohort by assessing the Charlson Comorbidity Index (CCI), Elixhauser Comorbidity Index (ECI), Multimorbidity Index (MMI), and Rheumatic Disease Comorbidity Index (RDCI) scores and to investigate their predictive ability for all-cause mortality. Methods: We identified 24,767 RA patients diagnosed from 1998 to 2008 in Taiwan and followed up until 31 December 2013. The incidence of comorbidities was estimated in three periods (before, during, and after the diagnostic period). The incidence rate ratios were calculated by comparing during vs. before and after vs. before the diagnostic period. One- and 5-year mortality rates were calculated and discriminated by low and high-score groups and modified models for each index. Results: The mean score at diagnosis was 0.8 in CCI, 2.8 in ECI, 0.7 in MMI, and 1.3 in RDCI, and annual percentage changes are 11.0%, 11.3%, 9.7%, and 6.8%, respectively. The incidence of any increase in the comorbidity index was significantly higher in the periods of “during” and “after” the RA diagnosis (incidence rate ratios for different indexes: 1.33–2.77). The mortality rate significantly differed between the high and low-score groups measured by each index (adjusted hazard ratios: 2.5–4.3 for different indexes). CCI was slightly better in the prediction of 1- and 5-year mortality rates. Conclusions: Comorbidities are common before and after RA diagnosis, and the rate of accumulation accelerates after RA diagnosis. All four comorbidity indexes are useful to measure the temporal changes and to predict mortality.
Collapse
|
16
|
Ammar Y, Mohamed A, Khalil G, Maharem D. Accelerated Atherosclerosis in Systemic Lupus Erythematosus: Role of Fibroblast Growth Factor 23- Phosphate Axis. Int J Nephrol Renovasc Dis 2021; 14:331-347. [PMID: 34475774 PMCID: PMC8407679 DOI: 10.2147/ijnrd.s326399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/05/2021] [Indexed: 01/16/2023] Open
Abstract
PURPOSE Despite management advances, accelerated atherosclerotic cardiovascular disease (ACVD) remains a major cause of morbimortality in systemic lupus erythematosus (SLE) patients; that is not fully explained by traditional risk factors. Fibroblast growth factor-23 (FGF23) is a bone-derived phosphaturic hormone with multiple klotho-dependent and independent effects, including promotion of atherosclerosis and vascular calcification, particularly in the context of chronic kidney disease. Increased circulating FGF23 was reported in SLE patients, particularly with lupus nephritis (LN); but its atherogenic role in these disorders was not explored. SUBJECTS AND METHODS Three study groups of predominantly middle-aged females were categorized by the 2012 SLE International Collaborating Clinics (SLICC) criteria as SLE (without LN), LN, or controls matching for traditional CVD risk profile. Measures of SLE activity, damage, steroid therapy, and glomerular filtration rate were calculated. Fasting blood samples were checked for serum lipid profile, anti-DNA, urea, creatinine, uric acid, proteins, albumin, calcium, phosphorus, C3, C4, CRP, vitamin-D3, intact parathyroid hormone and FGF23 (iFGF23). By carotid ultrasonography, mean common carotid artery intima-media thickness (CC-IMT), plaque score (PS) and internal carotid resistive index (ICRI) were recorded. RESULTS CC-IMT, ICRI and serum iFGF23 differed along the study groups (LN>SLE>controls). In both SLE and LN patients, serum iFGF23 had a significant positive correlation with serum phosphorus, CC-IMT and PS. On multivariate analysis, the strongest predictor of increased CC-IMT was cumulative steroid dose in SLE and serum iFGF23 in LN patients. Most significant independent predictors of increased serum iFGF23 were hyperphosphatemia in SLE and proteinuria in LN patients. CONCLUSION FGF23-phosphate axis has a key role in accelerated ACVD in SLE patients. Serum phosphorus and iFGF23 should be included in ACVD risk profile assessment of these patients. Prospective studies shall define the role of dietary and/or pharmacologic control of hyperphosphatemia and proteinuria in reducing circulating iFGF23 and ACVD in them.
Collapse
Affiliation(s)
- Yaser Ammar
- Internal Medicine Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Amira Mohamed
- Internal Medicine Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Gihane Khalil
- Chemical Pathology Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Dalia Maharem
- Internal Medicine Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|
17
|
Peçanha T, Bannell DJ, Sieczkowska SM, Goodson N, Roschel H, Sprung VS, Low DA. Effects of physical activity on vascular function in autoimmune rheumatic diseases: a systematic review and meta-analysis. Rheumatology (Oxford) 2021; 60:3107-3120. [PMID: 33521818 DOI: 10.1093/rheumatology/keab094] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/18/2020] [Accepted: 01/05/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVES To summarize existing evidence and quantify the effects of physical activity on vascular function and structure in autoimmune rheumatic diseases (ARDs). METHODS Databases were searched (through March 2020) for clinical trials evaluating the effects of physical activity interventions on markers of micro- and macrovascular function and macrovascular structure in ARDs. Studies were combined using random effects meta-analysis, which was conducted using Hedges' g. Meta-analyses were performed on each of the following outcomes: microvascular function [i.e. skin blood flow or vascular conductance responses to acetylcholine (ACh) or sodium nitropusside (SNP) administration]; macrovascular function [i.e. brachial flow-mediated dilation (FMD%) or brachial responses to glyceryl trinitrate (GTN%); and macrovascular structure [i.e. aortic pulse wave velocity (PWV)]. RESULTS Ten studies (11 trials) with a total of 355 participants were included in this review. Physical activity promoted significant improvements in microvascular [skin blood flow responses to ACh, g = 0.92 (95% CI 0.42, 1.42)] and macrovascular function [FMD%, g = 0.94 (95% CI 0.56, 1.02); GTN%, g = 0.53 (95% CI 0.09, 0.98)]. Conversely, there was no evidence for beneficial effects of physical activity on macrovascular structure [PWV, g = -0.41 (95% CI -1.13, 0.32)]. CONCLUSIONS Overall, the available clinical trials demonstrated a beneficial effect of physical activity on markers of micro- and macrovascular function but not on macrovascular structure in patients with ARDs. The broad beneficial impact of physical activity across the vasculature identified in this review support its role as an effective non-pharmacological management strategy for patients with ARDs.
Collapse
Affiliation(s)
- Tiago Peçanha
- Applied Physiology and Nutrition Research Group, School of Physical Education and Sport; Laboratory of Assessment and Conditioning in Rheumatology, Rheumatology Division, Hospital das Clínicas HCFMUSP, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Daniel J Bannell
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University
| | - Sofia Mendes Sieczkowska
- Applied Physiology and Nutrition Research Group, School of Physical Education and Sport; Laboratory of Assessment and Conditioning in Rheumatology, Rheumatology Division, Hospital das Clínicas HCFMUSP, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Nicola Goodson
- Liverpool University Hospitals NHS Foundation Trust, Aintree University Hospital, Liverpool, UK
| | - Hamilton Roschel
- Applied Physiology and Nutrition Research Group, School of Physical Education and Sport; Laboratory of Assessment and Conditioning in Rheumatology, Rheumatology Division, Hospital das Clínicas HCFMUSP, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Victoria S Sprung
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University
| | - David A Low
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University
| |
Collapse
|
18
|
Fernandes das Neves M, Jury EC, Delgado Alves J. High density lipoprotein influences CD4 T cell proliferation in Systemic Lupus Erythematosus and increases TGF-β1 expression: A potential role in the protection from atherosclerosis and autoimmunity. Lupus 2021; 30:2019-2021. [PMID: 34284678 DOI: 10.1177/09612033211034557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Marisa Fernandes das Neves
- Center of the Study of Chronic Diseases, New University of Lisbon, Lisbon, Portugal.,Medicine 4 Department, Fernando Fonseca Hospital, Amadora, Portugal
| | - Elisabeth C Jury
- Department of Inflammation, University College London, London, UK
| | - José Delgado Alves
- Center of the Study of Chronic Diseases, New University of Lisbon, Lisbon, Portugal.,Medicine 4 Department, Fernando Fonseca Hospital, Amadora, Portugal
| |
Collapse
|
19
|
Greenan-Barrett J, Doolan G, Shah D, Virdee S, Robinson GA, Choida V, Gak N, de Gruijter N, Rosser E, Al-Obaidi M, Leandro M, Zandi MS, Pepper RJ, Salama A, Jury EC, Ciurtin C. Biomarkers Associated with Organ-Specific Involvement in Juvenile Systemic Lupus Erythematosus. Int J Mol Sci 2021; 22:7619. [PMID: 34299237 PMCID: PMC8306911 DOI: 10.3390/ijms22147619] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 12/16/2022] Open
Abstract
Juvenile systemic lupus erythematosus (JSLE) is characterised by onset before 18 years of age and more severe disease phenotype, increased morbidity and mortality compared to adult-onset SLE. Management strategies in JSLE rely heavily on evidence derived from adult-onset SLE studies; therefore, identifying biomarkers associated with the disease pathogenesis and reflecting particularities of JSLE clinical phenotype holds promise for better patient management and improved outcomes. This narrative review summarises the evidence related to various traditional and novel biomarkers that have shown a promising role in identifying and predicting specific organ involvement in JSLE and appraises the evidence regarding their clinical utility, focusing in particular on renal biomarkers, while also emphasising the research into cardiovascular, haematological, neurological, skin and joint disease-related JSLE biomarkers, as well as genetic biomarkers with potential clinical applications.
Collapse
Affiliation(s)
- James Greenan-Barrett
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
| | - Georgia Doolan
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
| | - Devina Shah
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
| | - Simrun Virdee
- Department of Ophthalmology, Royal Free Hospital, London NW3 2QG, UK;
| | - George A. Robinson
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
| | - Varvara Choida
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
| | - Nataliya Gak
- Department of Rheumatology, University College London Hospital NHS Foundation Trust, London NW1 2BU, UK; (N.G.); (M.L.)
| | - Nina de Gruijter
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
| | - Elizabeth Rosser
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
| | - Muthana Al-Obaidi
- Department of Paediatric Rheumatology, Great Ormond Street Hospital, London WC1N 3JH, UK;
- NIHR Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Maria Leandro
- Department of Rheumatology, University College London Hospital NHS Foundation Trust, London NW1 2BU, UK; (N.G.); (M.L.)
- Centre for Rheumatology, Division of Medicine, University College London, London WC1E 6DH, UK;
| | - Michael S. Zandi
- Department of Neurology, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London NW1 2BU, UK;
| | - Ruth J. Pepper
- Department of Renal Medicine, Royal Free Hospital, University College London, London NW3 2QG, UK; (R.J.P.); (A.S.)
| | - Alan Salama
- Department of Renal Medicine, Royal Free Hospital, University College London, London NW3 2QG, UK; (R.J.P.); (A.S.)
| | - Elizabeth C. Jury
- Centre for Rheumatology, Division of Medicine, University College London, London WC1E 6DH, UK;
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
- Department of Rheumatology, University College London Hospital NHS Foundation Trust, London NW1 2BU, UK; (N.G.); (M.L.)
| |
Collapse
|
20
|
Yeh YL, Lu MC, Tsai BCK, Tzang BS, Cheng SM, Zhang X, Yang LY, Mahalakshmi B, Kuo WW, Xiang P, Huang CY. Heat-Killed Lactobacillus reuteri GMNL-263 Inhibits Systemic Lupus Erythematosus-Induced Cardiomyopathy in NZB/W F1 Mice. Probiotics Antimicrob Proteins 2021; 13:51-59. [PMID: 32514746 DOI: 10.1007/s12602-020-09668-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
It has been increasingly recognized that accelerated atherosclerosis is a major cause of morbidity and mortality in patients with systemic lupus erythematosus, a multisystem autoimmune disease. In this study, we investigated the anti-apoptotic effects of heat-killed Lactobacillus reuteri GMNL-263 on the cardiac tissue of NZB/W F1 mice. The myocardial architecture of the mice heart was observed and evaluated using different staining techniques such as hematoxylin and eosin, TUNEL assay, Masson's trichrome, and fluorescent immunohistochemistry. Additionally, the probiotics-related pathway proteins were analyzed via western blot analysis. Our results showed prevention of enlarged interstitial spaces and abnormal myocardial structures in the hearts of NZB/W F1 mice with L. reuteri GMNL-263 feeding. Significant reduction in TUNEL-positive cells, Fas death receptor-related components, and apoptosis was also detected in the cardiac tissues of the NZB/W F1 mice after L. reuteri GMNL-263 feeding compared with the control group. These findings are the first to reveal the protective effects of L. reuteri GMNL-263 against cardiac abnormalities in NZB/W F1 mice and suggest the potential clinical applications of L. reuteri GMNL-263 in the treatment of SLE-related cardiovascular diseases.
Collapse
Affiliation(s)
- Yu-Lan Yeh
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan.,Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Taipei, Taiwan
| | - Min-Chi Lu
- Division of Infectious Diseases, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan.,Department of Microbiology and Immunology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Bruce Chi-Kang Tsai
- Graduate Institute of Aging Medicine, China Medical University, Taichung, Taiwan
| | - Bor-Show Tzang
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Shiu-Min Cheng
- Department of Healthcare Administration, Asia University, Taichung, Taiwan
| | - Xiaoyong Zhang
- Department of Cardiology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Liang-Yo Yang
- Department of Physiology, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - B Mahalakshmi
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Peng Xiang
- Nephrology Center, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital , Qingyuan, Guangdong, China
| | - Chih-Yang Huang
- Graduate Institute of Aging Medicine, China Medical University, Taichung, Taiwan. .,Department of Biotechnology, Asia University, Taichung, Taiwan. .,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan. .,Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan.
| |
Collapse
|
21
|
Lu L, Kong W, Zhou K, Chen J, Hou Y, Dou H, Liang J. Association of lipoproteins and thyroid hormones with cognitive dysfunction in patients with systemic lupus erythematosus. BMC Rheumatol 2021; 5:18. [PMID: 34103098 PMCID: PMC8188676 DOI: 10.1186/s41927-021-00190-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/11/2021] [Indexed: 12/19/2022] Open
Abstract
Background Neuropsychiatric manifestations occur in up to 75% of adult systemic lupus erythematosus (SLE) patients and are one of the major causes of death in SLE patients. Cognitive dysfunction is a typical clinical feature of neuropsychiatric SLE (NPSLE), which seriously affects the quality of life of patients. Dyslipidaemia and thyroid symptoms, which are prevalent in SLE patients, have both been related to neuropsychiatric disturbances, including significant psychiatric and cognitive disturbances. This study aimed to investigate whether cognitive dysfunction in patients with SLE was related to the expression of serum thyroid hormone and lipoprotein levels. Methods A total of 121 patients with SLE and 65 healthy controls (HCs) at Nanjing Drum Tower Hospital completed a cognitive function test, and 81 SLE patients were divided into a high-cognition (n = 33) group and a low-cognition group (n = 48). The clinical and laboratory characteristics of the patients were compared; moreover, correlations between serum HDL-C, LDL-C, F-T3 and F-T4 levels and cognitive function were analysed. Serum levels of APOE, APOA1, IGF-1, and IGFBP7 in 81 patients were detected by ELISA, and the correlation between these four proteins and cognition was analysed separately. Results The patients with SLE with abnormal cognitive function were less educated than the HCs. For low-cognition patients, the levels of albumin, F-T3 (P < 0.05) and F-T4 decreased, while D-dimer, anti-dsDNA antibody, and IgM levels increased. Serum F-T3 and F-T4 levels positively correlated with cognition. Furthermore, serum protein levels of APOE and APOA1 showed no difference between the high- and low-cognition groups. However, the serum APOE levels were negatively correlated with line orientation scores, and APOA1 levels were positively correlated with coding scores. Conclusions Serum F-T3 and F-T4 levels were both positively correlated with four indexes of cognition (language was the exception), while serum APOE levels were negatively correlated with line orientation scores, APOA1 levels were positively correlated with coding scores, and IGFBP7 levels were negatively correlated with figure copy scores. These results demonstrated that F-T3 and F-T4 might be clinical biomarkers of cognitive dysfunction in SLE. Supplementary Information The online version contains supplementary material available at 10.1186/s41927-021-00190-7.
Collapse
Affiliation(s)
- Li Lu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, PR China.,The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, PR China
| | - Wei Kong
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, PR China.,The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Kangxing Zhou
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, PR China.,The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Jinglei Chen
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, PR China
| | - Yayi Hou
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, PR China. .,The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China. .,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, PR China.
| | - Huan Dou
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, PR China. .,The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China. .,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, PR China.
| | - Jun Liang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, PR China. .,The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China.
| |
Collapse
|
22
|
Hasni SA, Gupta S, Davis M, Poncio E, Temesgen-Oyelakin Y, Carlucci PM, Wang X, Naqi M, Playford MP, Goel RR, Li X, Biehl AJ, Ochoa-Navas I, Manna Z, Shi Y, Thomas D, Chen J, Biancotto A, Apps R, Cheung F, Kotliarov Y, Babyak AL, Zhou H, Shi R, Stagliano K, Tsai WL, Vian L, Gazaniga N, Giudice V, Lu S, Brooks SR, MacKay M, Gregersen P, Mehta NN, Remaley AT, Diamond B, O’Shea JJ, Gadina M, Kaplan MJ. Phase 1 double-blind randomized safety trial of the Janus kinase inhibitor tofacitinib in systemic lupus erythematosus. Nat Commun 2021; 12:3391. [PMID: 34099646 PMCID: PMC8185103 DOI: 10.1038/s41467-021-23361-z] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 04/22/2021] [Indexed: 02/05/2023] Open
Abstract
Increased risk of premature cardiovascular disease (CVD) is well recognized in systemic lupus erythematosus (SLE). Aberrant type I-Interferon (IFN)-neutrophil interactions contribute to this enhanced CVD risk. In lupus animal models, the Janus kinase (JAK) inhibitor tofacitinib improves clinical features, immune dysregulation and vascular dysfunction. We conducted a randomized, double-blind, placebo-controlled clinical trial of tofacitinib in SLE subjects (ClinicalTrials.gov NCT02535689). In this study, 30 subjects are randomized to tofacitinib (5 mg twice daily) or placebo in 2:1 block. The primary outcome of this study is safety and tolerability of tofacitinib. The secondary outcomes include clinical response and mechanistic studies. The tofacitinib is found to be safe in SLE meeting study's primary endpoint. We also show that tofacitinib improves cardiometabolic and immunologic parameters associated with the premature atherosclerosis in SLE. Tofacitinib improves high-density lipoprotein cholesterol levels (p = 0.0006, CI 95%: 4.12, 13.32) and particle number (p = 0.0008, CI 95%: 1.58, 5.33); lecithin: cholesterol acyltransferase concentration (p = 0.024, CI 95%: 1.1, -26.5), cholesterol efflux capacity (p = 0.08, CI 95%: -0.01, 0.24), improvements in arterial stiffness and endothelium-dependent vasorelaxation and decrease in type I IFN gene signature, low-density granulocytes and circulating NETs. Some of these improvements are more robust in subjects with STAT4 risk allele.
Collapse
Affiliation(s)
- Sarfaraz A. Hasni
- grid.420086.80000 0001 2237 2479Lupus Clinical Trials Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD USA
| | - Sarthak Gupta
- grid.420086.80000 0001 2237 2479Lupus Clinical Trials Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD USA ,grid.420086.80000 0001 2237 2479Systemic Autoimmunity Branch, NIAMS, NIH, Bethesda, MD USA
| | - Michael Davis
- grid.420086.80000 0001 2237 2479Lupus Clinical Trials Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD USA
| | - Elaine Poncio
- grid.420086.80000 0001 2237 2479Lupus Clinical Trials Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD USA
| | - Yenealem Temesgen-Oyelakin
- grid.420086.80000 0001 2237 2479Lupus Clinical Trials Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD USA
| | - Philip M. Carlucci
- grid.420086.80000 0001 2237 2479Systemic Autoimmunity Branch, NIAMS, NIH, Bethesda, MD USA
| | - Xinghao Wang
- grid.420086.80000 0001 2237 2479Systemic Autoimmunity Branch, NIAMS, NIH, Bethesda, MD USA
| | - Mohammad Naqi
- grid.420086.80000 0001 2237 2479Lupus Clinical Trials Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD USA
| | - Martin P. Playford
- grid.279885.90000 0001 2293 4638Section of Inflammation and Cardiometabolic Diseases, National Heart Lung and Blood Institute (NHLBI), NIH, Bethesda, MD USA
| | - Rishi R. Goel
- grid.420086.80000 0001 2237 2479Systemic Autoimmunity Branch, NIAMS, NIH, Bethesda, MD USA
| | - Xiaobai Li
- grid.410305.30000 0001 2194 5650NIH Clinical Center Biostatistics and Clinical Epidemiology Service, Bethesda, MD USA
| | - Ann J. Biehl
- grid.420086.80000 0001 2237 2479Office of the Clinical Director, NIAMS, NIH, Bethesda, MD USA
| | - Isabel Ochoa-Navas
- grid.420086.80000 0001 2237 2479Lupus Clinical Trials Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD USA
| | - Zerai Manna
- grid.420086.80000 0001 2237 2479Lupus Clinical Trials Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD USA
| | - Yinghui Shi
- grid.420086.80000 0001 2237 2479Translational Immunology Section, NIAMS, NIH, Bethesda, MD USA
| | - Donald Thomas
- Arthritis and Pain Associates of PG County, Greenbelt, MD USA
| | - Jinguo Chen
- grid.94365.3d0000 0001 2297 5165Trans-NIH Center for Human Immunology, Autoimmunity and Inflammation, NIH, Bethesda, MD USA
| | - Angélique Biancotto
- grid.94365.3d0000 0001 2297 5165Trans-NIH Center for Human Immunology, Autoimmunity and Inflammation, NIH, Bethesda, MD USA
| | - Richard Apps
- grid.94365.3d0000 0001 2297 5165Trans-NIH Center for Human Immunology, Autoimmunity and Inflammation, NIH, Bethesda, MD USA
| | - Foo Cheung
- grid.94365.3d0000 0001 2297 5165Trans-NIH Center for Human Immunology, Autoimmunity and Inflammation, NIH, Bethesda, MD USA
| | - Yuri Kotliarov
- grid.94365.3d0000 0001 2297 5165Trans-NIH Center for Human Immunology, Autoimmunity and Inflammation, NIH, Bethesda, MD USA
| | - Ashley L. Babyak
- grid.94365.3d0000 0001 2297 5165Trans-NIH Center for Human Immunology, Autoimmunity and Inflammation, NIH, Bethesda, MD USA
| | - Huizhi Zhou
- grid.94365.3d0000 0001 2297 5165Trans-NIH Center for Human Immunology, Autoimmunity and Inflammation, NIH, Bethesda, MD USA
| | - Rongye Shi
- grid.94365.3d0000 0001 2297 5165Trans-NIH Center for Human Immunology, Autoimmunity and Inflammation, NIH, Bethesda, MD USA
| | - Katie Stagliano
- grid.94365.3d0000 0001 2297 5165Trans-NIH Center for Human Immunology, Autoimmunity and Inflammation, NIH, Bethesda, MD USA
| | - Wanxia Li Tsai
- grid.420086.80000 0001 2237 2479Translational Immunology Section, NIAMS, NIH, Bethesda, MD USA
| | - Laura Vian
- grid.420086.80000 0001 2237 2479Translational Immunology Section, NIAMS, NIH, Bethesda, MD USA
| | - Nathalia Gazaniga
- grid.420086.80000 0001 2237 2479Translational Immunology Section, NIAMS, NIH, Bethesda, MD USA
| | - Valentina Giudice
- grid.279885.90000 0001 2293 4638Hematology Branch, NHLBI, NIH, Bethesda, MD USA
| | - Shajia Lu
- grid.420086.80000 0001 2237 2479Translational Immunology Section, NIAMS, NIH, Bethesda, MD USA
| | - Stephen R. Brooks
- grid.420086.80000 0001 2237 2479Biodata Mining and Discovery Section, NIAMS, NIH, Bethesda, MD USA
| | - Meggan MacKay
- grid.250903.d0000 0000 9566 0634Feinstein Institute for Medical Research, Manhasset, NY USA
| | - Peter Gregersen
- grid.250903.d0000 0000 9566 0634Feinstein Institute for Medical Research, Manhasset, NY USA
| | - Nehal N. Mehta
- grid.279885.90000 0001 2293 4638Section of Inflammation and Cardiometabolic Diseases, National Heart Lung and Blood Institute (NHLBI), NIH, Bethesda, MD USA
| | - Alan T. Remaley
- grid.279885.90000 0001 2293 4638Translational Vascular Medicine Branch, NHLBI, NIH, Bethesda, MD USA
| | - Betty Diamond
- grid.250903.d0000 0000 9566 0634Feinstein Institute for Medical Research, Manhasset, NY USA
| | - John J. O’Shea
- grid.420086.80000 0001 2237 2479Molecular Immunology and Inflammation Branch, NIAMS, NIH, Bethesda, MD USA
| | - Massimo Gadina
- grid.420086.80000 0001 2237 2479Translational Immunology Section, NIAMS, NIH, Bethesda, MD USA
| | - Mariana J. Kaplan
- grid.420086.80000 0001 2237 2479Systemic Autoimmunity Branch, NIAMS, NIH, Bethesda, MD USA
| |
Collapse
|
23
|
Sivakumaran J, Harvey P, Omar A, Tayer-Shifman O, Urowitz MB, Gladman DD, Anderson N, Su J, Touma Z. Assessment of cardiovascular risk tools as predictors of cardiovascular disease events in systemic lupus erythematosus. Lupus Sci Med 2021; 8:8/1/e000448. [PMID: 34045359 PMCID: PMC8162102 DOI: 10.1136/lupus-2020-000448] [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: 09/28/2020] [Accepted: 05/06/2021] [Indexed: 11/03/2022]
Abstract
BACKGROUND SLE is an independent risk factor for cardiovascular disease (CVD). This study aimed to determine which among QRISK2, QRISK3, Framingham Risk Score (FRS), modified Framingham Risk Score (mFRS) and SLE Cardiovascular Risk Equation (SLECRE) best predicts CVD. METHODS This is a single-centre analysis on 1887 patients with SLE followed prospectively according to a standard protocol. Tools' scores were evaluated against CVD development at/within 10 years for patients with CVD and without CVD. For patients with CVD, the index date for risk score calculation was chosen as close to 10 years prior to CVD event. For patients without CVD, risk scores were calculated as close to 10 years prior to the most recent clinic appointment. Proportions of low-risk (<10%), intermediate-risk (10%-20%) and high-risk (>20%) patients for developing CVD according to each tool were determined, allowing sensitivity, specificity, positive/negative predictive value and concordance (c) statistics analysis. RESULTS Among 1887 patients, 232 CVD events occurred. QRISK2 and FRS, and QRISK3 and mFRS, performed similarly. SLECRE classified the highest number of patients as intermediate and high risk. Sensitivities and specificities were 19% and 93% for QRISK2, 22% and 93% for FRS, 46% and 83% for mFRS, 47% and 78% for QRISK3, and 61% and 64% for SLECRE. Tools were similar in negative predictive value, ranging from 89% (QRISK2) to 92% (SLECRE). FRS and mFRS had the greatest c-statistics (0.73), while QRISK3 and SLECRE had the lowest (0. 67). CONCLUSION mFRS was superior to FRS and was not outperformed by the QRISK tools. SLECRE had the highest sensitivity but the lowest specificity. mFRS is an SLE-adjusted practical tool with a simple, intuitive scoring system reasonably appropriate for ambulatory settings, with more research needed to develop more accurate CVD risk prediction tools in this population.
Collapse
Affiliation(s)
| | - Paula Harvey
- Women's College Research Institute, Women's College Hospital, Toronto, Ontario, Canada
| | - Ahmed Omar
- University of Toronto, Toronto, Ontario, Canada
| | - Oshrat Tayer-Shifman
- Department of Internal Medicine B and Rheumatology Service, Meir Medical Center, Kfar Saba, Israel
| | - Murray B Urowitz
- Medicine, University of Toronto, Toronto, Ontario, Canada.,Toronto Western Hospital Centre for Prognosis Studies in the Rheumatic Diseases, Toronto, Ontario, Canada
| | - Dafna D Gladman
- University of Toronto, Toronto, Ontario, Canada.,Toronto Western Hospital Centre for Prognosis Studies in the Rheumatic Diseases, Toronto, Ontario, Canada
| | - Nicole Anderson
- Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Jiandong Su
- Toronto Western Hospital Centre for Prognosis Studies in the Rheumatic Diseases, Toronto, Ontario, Canada
| | - Zahi Touma
- University of Toronto, Toronto, Ontario, Canada .,Toronto Western Hospital Centre for Prognosis Studies in the Rheumatic Diseases, Toronto, Ontario, Canada
| |
Collapse
|
24
|
Nived O, Ingvarsson RF, Jöud A, Linge P, Tydén H, Jönsen A, Bengtsson AA. Disease duration, age at diagnosis and organ damage are important factors for cardiovascular disease in SLE. Lupus Sci Med 2021; 7:7/1/e000398. [PMID: 32587062 PMCID: PMC7319716 DOI: 10.1136/lupus-2020-000398] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/07/2020] [Accepted: 05/24/2020] [Indexed: 12/19/2022]
Abstract
Objective To report the incidence rate ratios (IRR) of acute myocardial infarctions (AMI) and cerebrovascular events (CVE) in incident SLE cases from a defined population. To study the risk factors for cardiovascular events in all patients with SLE at our unit. Methods Patients with SLE diagnosed from 1981 to 2006 were followed through to 2016. IRRs of AMI and CVE were calculated. The AMI and CVE incidence patterns for patients with SLE were studied in relation to hypertension, smoking, renal dysfunction, anticardiolipin (aCL) antibodies at diagnosis, disease duration and organ damage before an event. Results 262 patients with SLE were included in the study; of these 175 were from the defined population. Overall, 37 AMI and 44 CVE were recorded. An increased IRR of 3 for AMI was found (p<0.001). Smoking, hypertension and reduced renal function were risk factors for AMI. An increased IRR of 3.3 for ischaemic CVE was found for women (p<0.001). Hypertension and aCL were risk factors for CVE. Organ damage before events was increased. Conclusions Cardiovascular events are increased in SLE and are associated with hypertension, smoking and increased damage rate.
Collapse
Affiliation(s)
- Ola Nived
- Department of Rheumatology, Institution for Clinical Sciences, Lund, Sweden
| | - Ragnar Freyr Ingvarsson
- Department of Rheumatology, Institution for Clinical Sciences, Lund, Sweden.,Landspitali University Hospital, Reykjavik, Iceland
| | - Anna Jöud
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Petrus Linge
- Department of Rheumatology, Institution for Clinical Sciences, Lund, Sweden
| | - Helena Tydén
- Department of Rheumatology, Institution for Clinical Sciences, Lund, Sweden
| | - Andreas Jönsen
- Department of Rheumatology, Institution for Clinical Sciences, Lund, Sweden
| | - Anders A Bengtsson
- Department of Rheumatology, Institution for Clinical Sciences, Lund, Sweden
| |
Collapse
|
25
|
Rodríguez-Calvo R, Guardiola M, Oliva I, Arrando H, Arranz I, Ferré A, Pellicer P, Parra S, Ribalta J, Castro A. Low-density lipoprotein from active SLE patients is more atherogenic to endothelial cells than low-density lipoprotein from the same patients during remission. Rheumatology (Oxford) 2021; 60:866-871. [PMID: 32844232 DOI: 10.1093/rheumatology/keaa380] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/02/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES SLE patients have an enhanced risk of atherosclerosis and cardiovascular disease. However, the increased prevalence of cardiovascular disease is not fully explained by traditional Framingham cardiovascular risk factors. Specific features of low-density lipoprotein (LDL) particles, other than plasma concentration, may induce accelerated atherosclerosis at early stages in these patients. Thus, we aimed to explore the impact of LDL from both active and inactive SLE patients on human aortic endothelial cells. METHODS Human aortic endothelial cells were stimulated with the same concentration of LDL particles isolated from pooled serum that was collected from 13 SLE patients during both active and inactive states. Gene expression and cell migration assays were performed. RESULTS Circulating LDL particles obtained from healthy volunteers and SLE patients in both remission and flare states were comparable in terms of number, cholesterol and triglyceride content, and net electric charge. Stimulation of cells with LDL from active SLE patients induced the expression of vascular cell adhesion molecule 1 (∼2.0-fold, P < 0.05), monocyte chemoattractant protein 1 (∼2.0-fold, P < 0.05) and matrix metallopeptidase 2 (∼1.6-fold, P < 0.01) compared with cells stimulated with LDL from inactive SLE patients. Additionally, LDL extracted from active patients increased cell migration in a wound-healing assay (1.4-fold, P < 0.05). CONCLUSION Our data show that, at the same LDL concentration, LDL from active SLE patients had increased proatherogenic effects on endothelial cells compared with LDL from the same patients when in an inactive or remission state.
Collapse
Affiliation(s)
- Ricardo Rodríguez-Calvo
- Unitat de Recerca en Lípids i Arteriosclerosi, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, Reus, Spain.,Institut d'Investigació Sanitària Pere Virgili, Reus, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Reus, Spain
| | - Montse Guardiola
- Unitat de Recerca en Lípids i Arteriosclerosi, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, Reus, Spain.,Institut d'Investigació Sanitària Pere Virgili, Reus, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Reus, Spain
| | - Iris Oliva
- Unitat de Recerca en Lípids i Arteriosclerosi, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, Reus, Spain.,Institut d'Investigació Sanitària Pere Virgili, Reus, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Reus, Spain
| | - Hugo Arrando
- Unitat de Recerca en Lípids i Arteriosclerosi, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, Reus, Spain
| | - Idoia Arranz
- Unitat de Recerca en Lípids i Arteriosclerosi, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, Reus, Spain
| | - Anna Ferré
- Unitat de Recerca en Lípids i Arteriosclerosi, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, Reus, Spain
| | - Paula Pellicer
- Unitat de Recerca en Lípids i Arteriosclerosi, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, Reus, Spain
| | - Sandra Parra
- Institut d'Investigació Sanitària Pere Virgili, Reus, Spain.,Unitat de Malalties Autoinmunes, Medicina Interna, Hospital Universitari Sant Joan de Reus, Universitat Rovira i Virgili, Reus, Spain
| | - Josep Ribalta
- Unitat de Recerca en Lípids i Arteriosclerosi, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, Reus, Spain.,Institut d'Investigació Sanitària Pere Virgili, Reus, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Reus, Spain
| | - Antoni Castro
- Institut d'Investigació Sanitària Pere Virgili, Reus, Spain.,Unitat de Malalties Autoinmunes, Medicina Interna, Hospital Universitari Sant Joan de Reus, Universitat Rovira i Virgili, Reus, Spain
| |
Collapse
|
26
|
Coelewij L, Waddington KE, Robinson GA, Chocano E, McDonnell T, Farinha F, Peng J, Dönnes P, Smith E, Croca S, Bakshi J, Griffin M, Nicolaides A, Rahman A, Jury EC, Pineda-Torra I. Serum Metabolomic Signatures Can Predict Subclinical Atherosclerosis in Patients With Systemic Lupus Erythematosus. Arterioscler Thromb Vasc Biol 2021; 41:1446-1458. [PMID: 33535791 PMCID: PMC7610443 DOI: 10.1161/atvbaha.120.315321] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/12/2020] [Indexed: 12/19/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Leda Coelewij
- Centre for Cardiometabolic and Vascular Science, Department of Medicine, University College London, London W1CE 6JF, U.K
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Kirsty E Waddington
- Centre for Cardiometabolic and Vascular Science, Department of Medicine, University College London, London W1CE 6JF, U.K
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - George A Robinson
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
- Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Elvira Chocano
- Centre for Cardiometabolic and Vascular Science, Department of Medicine, University College London, London W1CE 6JF, U.K
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Thomas McDonnell
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Filipa Farinha
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Junjie Peng
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
- Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Pierre Dönnes
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
- Scicross AB, Skövde, Sweden
| | - Edward Smith
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Sara Croca
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Jyoti Bakshi
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Maura Griffin
- Vascular Screening and Diagnostic Centre, Weymouth Street, London, UK
| | - Andrew Nicolaides
- Vascular Screening and Diagnostic Centre, Weymouth Street, London, UK
- St Georges London/Nicosia Medical School, University of Nicosia, Cyprus
| | - Anisur Rahman
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Elizabeth C Jury
- Centre for Rheumatology Research, Department of Medicine, University College London, London W1CE 6JF, U.K
| | - Ines Pineda-Torra
- Centre for Cardiometabolic and Vascular Science, Department of Medicine, University College London, London W1CE 6JF, U.K
| |
Collapse
|
27
|
Forte F, Buonaiuto A, Calcaterra I, Iannuzzo G, Ambrosino P, Di Minno MND. Association of systemic lupus erythematosus with peripheral arterial disease: a meta-analysis of literature studies. Rheumatology (Oxford) 2021; 59:3181-3192. [PMID: 32793980 DOI: 10.1093/rheumatology/keaa414] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/27/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE SLE patients have an increased cardiovascular morbidity and mortality. Contrasting data are available about the association between peripheral arterial disease (PAD) and SLE. We aimed to perform a meta-analysis of studies evaluating the association between SLE and PAD. METHODS Studies were systematically searched in the PubMed, Web of Science, Scopus and EMBASE databases according to preferred reporting items for systematic reviews and meta-analyses guidelines. RESULTS Eight studies reporting on 263 258 SLE patients and 768 487 controls showed that the prevalence of PAD was 15.8% (95% CI: 10.5%, 23.2%) in SLE patients and 3.9% (95% CI: 1.8%, 7.9%) in controls with a corresponding odds ratio of 4.1 (95% CI: 1.5, 11.6; P <0.001). In addition, five studies reporting on ankle-brachial index showed significantly lower values in 280 SLE patients as compared with 201 controls (mean difference: -0.018; 95% CI: -0.034, -0.001; P =0.033). Meta-regression models showed that age, hypertension and diabetes were inversely associated with the difference in the prevalence of PAD between SLE patients and non-SLE controls, whereas no effect for all the other clinical and demographic variables on the evaluated outcome was found. CONCLUSION SLE patients exhibit an increased prevalence of PAD and lower ankle-brachial index values as compared with non-SLE controls. This should be considered when planning prevention, interventional and rehabilitation strategies for these chronic patients with functional disability and poor long-term outcomes.
Collapse
Affiliation(s)
- Francesco Forte
- Department of Clinical Medicine and Surgery, Federico II University, Naples
| | - Alessio Buonaiuto
- Department of Clinical Medicine and Surgery, Federico II University, Naples
| | - Ilenia Calcaterra
- Department of Clinical Medicine and Surgery, Federico II University, Naples
| | - Gabriella Iannuzzo
- Department of Clinical Medicine and Surgery, Federico II University, Naples
| | | | | |
Collapse
|
28
|
López P, Rodríguez-Carrio J, Martínez-Zapico A, Pérez-Álvarez ÁI, Suárez-Díaz S, Mozo L, Benavente L, Caminal-Montero L, Suárez A. Low-density granulocytes and monocytes as biomarkers of cardiovascular risk in systemic lupus erythematosus. Rheumatology (Oxford) 2020; 59:1752-1764. [PMID: 32031658 DOI: 10.1093/rheumatology/keaa016] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/08/2020] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE The aim was to evaluate the most relevant cell populations involved in vascular homeostasis as potential biomarkers of SLE-related cardiovascular disease (CVD). METHODS Low-density granulocytes (LDGs), monocyte subsets, endothelial progenitor cells, angiogenic T (Tang) cells, CD4+CD28null and Th1/Th17 lymphocytes and serum cytokine levels were quantified in 109 SLE patients and 33 controls in relationship to the presence of subclinical carotid atheromatosis or cardiovascular disease. A second cohort including 31 recent-onset SLE patients was also included. RESULTS Raised monocyte and LDG counts, particularly those LDGs negative for CD16/CD14 expression (nLDGs), in addition to the ratios of monocytes and nLDGs to high-density lipoprotein-cholesterol (HDLc) molecules (MHR and nLHR, respectively), were present in SLE patients with traditional risk factors or subclinical atheromatosis but not in those who were CV-free, thus revealing their value in the identification of patients at risk of CVD, even at the onset of disease. Accordingly, nLDGs were correlated positively with carotid intima-media thickness (cIMT) and with inflammatory markers (CRP and IL-6). A bias towards more differentiated monocyte subsets, related to increased IFN-α and IL-17 serum levels, was also observed in patients. Intermediate monocytes were especially expanded, but independently of their involvement in CVD. Finally, CD4+CD28null, Th17 and Th1 lymphocytes were increased, with CD4+CD28null and Th17 cells being associated with cIMT, whereas endothelial progenitor and Tang cell levels were reduced in all SLE patients. CONCLUSION The present study highlights the potential use of MHR and nLHR as valuable biomarkers of CVD risk in SLE patients, even at diagnosis. The increased amounts of nLDGs, monocytes, Th17 and senescent-CD28null subsets, coupled with reduced pro-angiogenic endothelial progenitor cells and Tang cells, could underlie the development of atheromatosis in SLE.
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
- 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
| | - Ángel I Pérez-Álvarez
- Group of Basic and Translational Research in Inflammatory Diseases, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA).,Department of Neurology, Hospital Universitario Central de Asturias
| | - Silvia Suárez-Díaz
- 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
| | - Lourdes Mozo
- Group of Basic and Translational Research in Inflammatory Diseases, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA).,Department of Immunology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Lorena Benavente
- Group of Basic and Translational Research in Inflammatory Diseases, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA).,Department of Neurology, Hospital Universitario Central de Asturias
| | - 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
| | - 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
|
29
|
Moreira MLP, Sztajnbok F, Giannini DT. RELATIONSHIP BETWEEN FIBER INTAKE AND CARDIOVASCULAR RISK FACTORS IN ADOLESCENTS WITH SYSTEMIC LUPUS ERYTHEMATOSUS. REVISTA PAULISTA DE PEDIATRIA : ORGAO OFICIAL DA SOCIEDADE DE PEDIATRIA DE SAO PAULO 2020; 39:e2019316. [PMID: 32876308 PMCID: PMC7457466 DOI: 10.1590/1984-0462/2021/39/2019316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 01/23/2020] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To evaluate the fiber intake and the relationship with cardiovascular risk factors in adolescents with juvenile systemic lupus erythematosus. METHODS This is a cross-sectional in which adolescents with juvenile systemic lupus erythematosus were evaluated. The dietary consumption was assessed by the 24-hour recall; nutritional status was classified according to the Body Mass index/Age by Sex; abdominal obesity was assessed through waist circumference, waist-to-height ratio and glucose and lipid metabolism. The data were analyzed using Statistical Software for Professionals 14 and all statistical analyses used an alpha error of 5%. RESULTS 52 patients were evaluated, with a mean age of 16.7±1.5 years. Inadequate fiber consumption occurred in 61.5% (n=32) of them. Average of waist circumference measures (81.4 vs. 75.5 cm; p=0.02), waist-to-height ratio (0.51 vs. 0.47; p=0.02) and systolic blood pressure (122.1 vs. 114.8 mmHg; p=0.03) were higher in those who had inadequate fiber intake. Among the cardiovascular risk factors evaluated, the waist/height ratio showed a significant negative correlation with fiber consumption (r=-0.3; p=0.04), that is, the higher the fiber consumption, the lower the value of the waist ratio /stature. CONCLUSIONS Low dietary fiber intake in adolescents with systemic lupus erythematosus juvenile is related to higher abdominal adiposity and consequently with increased cardiovascular risk.
Collapse
Affiliation(s)
| | - Flávio Sztajnbok
- Center for Adolescent Health Studies, Universidade do Estado do
Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Denise Tavares Giannini
- Center for Adolescent Health Studies, Universidade do Estado do
Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
30
|
Ganjali S, Shirmohammadi L, Read MI, Sahebkar A. High-density lipoprotein functionality in systemic lupus erythematosus. Semin Arthritis Rheum 2020; 50:769-775. [PMID: 32531506 DOI: 10.1016/j.semarthrit.2020.05.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 05/04/2020] [Accepted: 05/11/2020] [Indexed: 11/18/2022]
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous disease which is characterized with excessive inflammation and autoantibodies, macrophage and complement activation, and subsequently immunologically mediated tissue damage. In spite of improved treatments of SLE, these patients experience premature atherosclerosis and the rate of mortality among them remains high. Autoantibodies and circulating immune complexes might contribute to the pathogenesis of atherosclerosis by injuring the endothelium, as well as inducing pro-inflammatory and pro-adhesive endothelial cell phenotypes, as well as altering the metabolism of lipoproteins involved in atherogenesis. Hence, high levels of atherogenic lipoproteins (like low-density lipoprotein (LDL) and very low-density lipoprotein (VLDL)) and low levels of high-density lipoprotein (HDL-C) are important risk factors for atherosclerotic cardiovascular complications in SLE patients but these traditional risk factors fail to fully explain the increased risk of cardiovascular disease (CVD) in these patients. The exact mechanism by which inflammation decreases HDL levels is not defined, but decreases in apoA-I production and lecithin cholesterol acyltransferase (LCAT) activity, as well as increased serum amyloid A (SAA), endothelial lipase and secretory phospholipase A2 activity (PLA2) could all contribute. In addition, during inflammation multiple changes in HDL structure occur, leading to alterations in HDL function which may be implicated in the CVD complications of SLE. Therefore, this review will aim to identify the mechanisms implicated in HDL dysfunction which occurs in SLE patients.
Collapse
Affiliation(s)
- Shiva Ganjali
- Department of Medical Biotechnology & Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Shirmohammadi
- Department of Medical Biotechnology & Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Morgayn I Read
- Department of Pharmacology, University of Otago School of Medical Sciences, Dunedin, New Zealand
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
31
|
Zhang X, Liu L, Ma X, Hu W, Xu X, Huang S, Hua B, Wang H, Chen Z, Sun L. Clinical significance of non-thyroidal illness syndrome on disease activity and dyslipidemia in patients with SLE. PLoS One 2020; 15:e0231622. [PMID: 32298352 PMCID: PMC7162454 DOI: 10.1371/journal.pone.0231622] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 03/29/2020] [Indexed: 12/19/2022] Open
Abstract
Objectives Nonthyroidal illness syndrome (NTIS), also known as low triiodothyronine (T3) syndrome, frequently affects patients with systemic lupus erythematosus (SLE) and may affect lipid metabolism. Dyslipidemia is highly prevalent and associated with the long-term prognosis of SLE. The aim of the present study was to explore the clinical significance of NTIS on disease activity and dyslipidemia in patients with SLE. Methods Clinical and laboratory data were collected retrospectively from 223 patients with SLE. The correlation between free triiodothyronine (FT3), SLE disease activity, and lipid profiles were estimated. The correlation coefficient (r) was calculated using a Pearson’s regression model. Univariate and multivariate logistic regression analyses were performed to identify the risk factors for dyslipidemia in SLE. Results Serum FT3 levels were negatively correlated with the levels of 24 h urine protein (UP), blood urea nitrogen (BUN), creatinine (Cr) and SLE disease activity index (SLEDAI) (all p < 0.001) in NTIS patients but not in euthyroid patients. ApoB/ApoA1 was significantly correlated with SLEDAI (p < 0.01) in NTIS patients and CRP (p < 0.001) and ESR (p < 0.01) in euthyroid patients. A multivariate analysis revealed that only FT3 exhibited an independent negative association with dyslipidemia (P = 0.01; OR = 0.48; 95% CI 0.27–0.85). Conclusion NTIS frequently occurs in patients with SLE. Low FT3 is associated with disease activity in SLE patients complicated with NTIS. Low FT3 is an independent risk factor for dyslipidemia in patients with SLE.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Lirong Liu
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
- Department of Rheumatology and Immunology, The first Hospital of Changshu, Changshu, China
| | - Xiaolei Ma
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Wei Hu
- Department of Clinical Laboratory, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Xue Xu
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Saisai Huang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Bingzhu Hua
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Hong Wang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Zhiyong Chen
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
- * E-mail: (LS); (ZC)
| | - Lingyun Sun
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
- * E-mail: (LS); (ZC)
| |
Collapse
|
32
|
Alpizar-Rodriguez D, Romero-Diaz J. Are cardiovascular events and mortality in patients with systemic lupus erythematosus predictable at diagnosis? Rheumatology (Oxford) 2020; 59:467-468. [DOI: 10.1093/rheumatology/kez539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 09/12/2019] [Accepted: 09/17/2019] [Indexed: 11/13/2022] Open
Affiliation(s)
- Deshiré Alpizar-Rodriguez
- Division of Rheumatology, Department of Internal Medicine Specialties, University Hospitals of Geneva, Geneva, Switzerland
| | - Juanita Romero-Diaz
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición, Mexico City, Mexico
| |
Collapse
|
33
|
Kieninger-Gräfitsch A, Vogt S, Ribi C, Dubler D, Chizzolini C, Huynh-Do U, Osthoff M, Trendelenburg M. No association of complement mannose-binding lectin deficiency with cardiovascular disease in patients with Systemic Lupus Erythematosus. Sci Rep 2020; 10:3693. [PMID: 32111865 PMCID: PMC7048794 DOI: 10.1038/s41598-020-60523-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 02/10/2020] [Indexed: 11/09/2022] Open
Abstract
Cardiovascular (CV) morbidity is the major cause of death in patients with Systemic Lupus Erythematosus (SLE). Previous studies on mannose-binding lectin (MBL) gene polymorphisms in SLE patients suggest that low levels of complement MBL are associated with cardiovascular disease (CVD). However, as large studies on MBL deficiency based on resulting MBL plasma concentrations are lacking, the aim of our study was to analyze the association of MBL concentrations with CVD in SLE patients. Plasma MBL levels SLE patients included in the Swiss SLE Cohort Study were quantified by ELISA. Five different CV organ manifestations were documented. Of 373 included patients (85.5% female) 62 patients had at least one CV manifestation. Patients with MBL deficiency (levels below 500 ng/ml or 1000 ng/ml) had no significantly increased frequency of CVD (19.4% vs. 15.2%, P = 0.3 or 17.7% vs. 15.7%, P = 0.7). After adjustment for traditional CV risk factors, MBL levels and positive antiphospholipid serology (APL+) a significant association of CVD with age, hypertension, disease duration and APL+ was demonstrated. In our study of a large cohort of patients with SLE, we could not confirm previous studies suggesting MBL deficiency to be associated with an increased risk for CVD.
Collapse
Affiliation(s)
- A Kieninger-Gräfitsch
- Division of Internal Medicine and Clinical Immunology Lab, Department of Biomedicine, University Hospital and University, Basel, Switzerland.
| | - S Vogt
- Division of Internal Medicine and Clinical Immunology Lab, Department of Biomedicine, University Hospital and University, Basel, Switzerland
| | - C Ribi
- Department of Immunology and Allergy, University Hospital, Lausanne, Switzerland
| | - D Dubler
- Division of Internal Medicine and Clinical Immunology Lab, Department of Biomedicine, University Hospital and University, Basel, Switzerland
| | - C Chizzolini
- Department of Internal Medicine Specialties, Clinical Immunology and Allergy, University Hospital and School of Medicine, Geneva, Switzerland
| | - U Huynh-Do
- Department of Nephrology and Hypertension, University Hospital, Bern, Switzerland
| | - M Osthoff
- Division of Internal Medicine and Clinical Immunology Lab, Department of Biomedicine, University Hospital and University, Basel, Switzerland
| | - M Trendelenburg
- Division of Internal Medicine and Clinical Immunology Lab, Department of Biomedicine, University Hospital and University, Basel, Switzerland
| |
Collapse
|
34
|
Ramirez GA, Manfredi AA, Maugeri N. Misunderstandings Between Platelets and Neutrophils Build in Chronic Inflammation. Front Immunol 2019; 10:2491. [PMID: 31695699 PMCID: PMC6817594 DOI: 10.3389/fimmu.2019.02491] [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: 08/04/2019] [Accepted: 10/07/2019] [Indexed: 12/14/2022] Open
Abstract
Regulated hemostasis, inflammation and innate immunity entail extensive interactions between platelets and neutrophils. Under physiological conditions, vascular inflammation offers a template for the establishment of effective intravascular immunity, with platelets providing neutrophils with an array of signals that increase their activation threshold, thus limiting collateral damage to tissues and promoting termination of the inflammatory response. By contrast, persistent systemic inflammation as observed in immune-mediated diseases, such as systemic vasculitides, systemic sclerosis, systemic lupus erythematosus or rheumatoid arthritis is characterized by platelet and neutrophil reciprocal activation, which ultimately culminates in the generation of thrombo-inflammatory lesions, fostering vascular injury and organ damage. Here, we discuss recent evidence regarding the multifaceted aspects of platelet-neutrophil interactions from bone marrow precursors to shed microparticles. Moreover, we analyse shared and disease-specific events due to an aberrant deployment of these interactions in human diseases. To restore communications between the pillars of the immune-hemostatic continuum constitutes a fascinating challenge for the near future.
Collapse
Affiliation(s)
- Giuseppe A Ramirez
- Vita-Salute San Raffaele University, Milan, Italy.,Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Angelo A Manfredi
- Vita-Salute San Raffaele University, Milan, Italy.,Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Norma Maugeri
- Vita-Salute San Raffaele University, Milan, Italy.,Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
35
|
Bagchi-Chakraborty J, Francis A, Bray T, Masters L, Tsiantoulas D, Nus M, Harrison J, Broekhuizen M, Leggat J, Clatworthy MR, Espéli M, Smith KG, Binder CJ, Mallat Z, Sage AP. B Cell Fcγ Receptor IIb Modulates Atherosclerosis in Male and Female Mice by Controlling Adaptive Germinal Center and Innate B-1-Cell Responses. Arterioscler Thromb Vasc Biol 2019; 39:1379-1389. [PMID: 31092015 PMCID: PMC6636804 DOI: 10.1161/atvbaha.118.312272] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/22/2019] [Indexed: 02/02/2023]
Abstract
Objective- Investigate the impact of modulating B cell FcγRIIb (Fcγ receptor IIb) expression on atherosclerosis. Approach and Results- Western diet-induced atherosclerosis was assessed in Ldlr-/- or Apoe-/- mice with B cell-specific overexpression of FcγRIIb or with an FcγRIIb promoter mutation that alters FcγRIIb expression in germinal center (GC) B cells. In males, overexpression of FcγRIIb on B cells severely reduced activated, class switched B cell responses, as indicated by reductions in GC B cells, plasma cells, and serum IgG but not IgM antibodies. Male mice overexpressing FcγRIIb developed less atherosclerosis, suggesting a pathogenic role for GC B cell IgG responses. In support of this hypothesis, male mice with a promoter polymorphism-driven reduction in FcγRIIb on GC B cells but not plasma cells have a converse phenotype of enhanced GC responses and IgG2c antibodies and enhanced atherosclerosis. IgG2c significantly enhanced TNF (tumor necrosis factor) secretion by CD11b+ CD11c+ cells expressing the high-affinity receptor FcγRIV. In females, overexpression of FcγRIIb on B cells not only reduced GC B cell responses but also substantially reduced B-1 cells and IgM antibodies, which translated into acceleration of atherosclerosis. Promoter-driven reduction in FcγRIIb did not alter GC B cell responses in females and, therefore, had no impact on atherosclerosis. Conclusions- B cell FcγRIIb differentially alters proatherogenic adaptive GC B cell and atheroprotective innate B-1 responses in male and female mice fed a western diet. Our results highlight the importance of a better understanding and ability to selectively target B cell responses in future immunotherapeutic approaches against human cardiovascular disease. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Jayashree Bagchi-Chakraborty
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Anna Francis
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Toni Bray
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Leanne Masters
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Dimitrios Tsiantoulas
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Meritxell Nus
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - James Harrison
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Michelle Broekhuizen
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Jennifer Leggat
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Menna R. Clatworthy
- Division of Immunology (M.R.C., K.G.C.S., Z.M.), Department of Medicine, University of Cambridge, United Kingdom
| | - Marion Espéli
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Paris, France (M.E.)
| | - Kenneth G.C. Smith
- Division of Immunology (M.R.C., K.G.C.S., Z.M.), Department of Medicine, University of Cambridge, United Kingdom
| | - Christoph J. Binder
- Department of Laboratory Medicine, Medical University of Vienna, Austria (C.J.B.)
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (C.J.B.)
| | - Ziad Mallat
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
- Division of Immunology (M.R.C., K.G.C.S., Z.M.), Department of Medicine, University of Cambridge, United Kingdom
- Institut National de la Santé et de la Recherche Médicale, Universite Paris-Descartes, Paris Cardiovascular Research Center, and Université Paris-Descartes, France (Z.M.)
| | - Andrew P. Sage
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| |
Collapse
|
36
|
Tani C, Elefante E, Signorini V, Zucchi D, Lorenzoni V, Carli L, Stagnaro C, Ferro F, Mosca M. Glucocorticoid withdrawal in systemic lupus erythematosus: are remission and low disease activity reliable starting points for stopping treatment? A real-life experience. RMD Open 2019; 5:e000916. [PMID: 31275608 PMCID: PMC6579574 DOI: 10.1136/rmdopen-2019-000916] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/02/2019] [Accepted: 05/09/2019] [Indexed: 12/26/2022] Open
Abstract
Objectives To evaluate the proportion of patients who have successfully withdrawn glucocorticoids (GCs) in a longitudinal cohort of patients with systemic lupus erythematosus (SLE) over a period of 6 years; to evaluate patient characteristics during GC withdrawal in relation to existing definitions of remission and Lupus Low Disease Activity State (LLDAS); and to evaluate the occurrence of flares after GC withdrawal. Methods Patients who attempted GC withdrawal were identified for the cohort, and the following information was assessed during withdrawal attempts: date of last disease flare, disease activity and damage and ongoing treatment. Information regarding the occurrence of disease flares after GC withdrawal was also recorded for patients who successfully stopped treatment.Definitions of remission were applied to GC withdrawal in line with European consensus criteria (Definitions of remission in SLE [DORIS]) and LLDAS in line with the Asian Pacific Lupus Consortium definition. Results 148 patients were involved in the study; GC withdrawal was attempted in 91 patients (61.5%) with 77 patients (84.6%) successfully stopping GCs. At the beginning of the GC reduction, the majority of patients were in complete or clinical remission (48.9% and 39.6%, respectively). Disease activity was significantly lower in patients who successfully stopped GCs, and the proportion of patients in complete remission was higher (54.2%) with respect to patients who failed in their attempt. Among patients who stopped GCs, 18 flares were recorded after a median of 1 year. The time period since the last flare was shorter in patients who experienced flares with respect to patients who did not flare (mean 0.93 years vs 6.0, p<0.001). Conclusions GC withdrawal is an achievable goal in SLE and may be attempted after a long-term remission or LLDAS to protect the patient from disease flares.
Collapse
Affiliation(s)
- Chiara Tani
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elena Elefante
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Viola Signorini
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Dina Zucchi
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Linda Carli
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Chiara Stagnaro
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesco Ferro
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Marta Mosca
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
37
|
De Lorenzis E, Gremese E, Bosello S, Nurmohamed MT, Sinagra G, Ferraccioli G. Microvascular heart involvement in systemic autoimmune diseases: The purinergic pathway and therapeutic insights from the biology of the diseases. Autoimmun Rev 2019; 18:317-324. [DOI: 10.1016/j.autrev.2019.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 10/16/2018] [Indexed: 02/08/2023]
|
38
|
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune rheumatic disease with a prevalence of approximately 1 in 1000. Over the last 30 years, advances in treatment such as use of corticosteroids and immunosuppressants have improved life expectancy and quality of life for patients with lupus and the key unmet needs have therefore changed. With the reduced mortality from disease activity, development of cardiovascular disease (CVD) has become an increasingly important cause of death in patients with SLE. The increased CVD risk in these patients is partly, but not fully explained by standard risk factors, and abnormalities in the immune response to lipids may play a role. Invariant natural killer T cells, which are triggered specifically by lipid antigens, may protect against progression of subclinical atherosclerosis. However, currently our recommendation is that clinicians should focus on optimal management of standard CVD risk factors such as smoking, blood pressure and lipid levels. Fatigue is one of the most common and most limiting symptoms suffered by patients with SLE. The cause of fatigue is multifactorial and disease activity does not explain this symptom. Consequently, therapies directed towards reducing inflammation and disease activity do not reliably reduce fatigue and new approaches are needed. Currently, we recommend asking about sleep pattern, optimising pain relief and excluding other causes of fatigue such as anaemia and metabolic disturbances. For the subgroup of patients whose disease activity is not fully controlled by standard treatment regimes, a range of different biologic agents have been proposed and subjected to clinical trials. Many of these trials have given disappointing results, though belimumab, which targets B lymphocytes, did meet its primary endpoint. New biologics targeting B cells, T cells or cytokines (especially interferon) are still going through trials raising the hope that novel therapies for patients with refractory SLE may be available soon.
Collapse
|
39
|
Liu W, Wang T, Sun P, Zhou Y. Expression of Hcy and blood lipid levels in serum of CHD patients and analysis of risk factors for CHD. Exp Ther Med 2018; 17:1756-1760. [PMID: 30783445 PMCID: PMC6364198 DOI: 10.3892/etm.2018.7111] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 11/15/2018] [Indexed: 11/25/2022] Open
Abstract
Expression of homocysteine (Hcy) and blood lipid levels in serum of atherosclerotic coronary heart disease (CHD) patients was investigated and the related risk factors were analyzed. A total of 148 CHD patients were selected as study group and another 120 healthy people attending Weifang People's Hospital for physical examination in the same period as control group. Hcy and blood lipid levels in serum were measured in the two groups, including total cholesterol (TC), triglyceride (TG), high-density lipoprotein cholesterol (HDL-C) and low-density lipoprotein cholesterol (LDL-C). At the same time, analysis of risk factors for CHD was performed. The levels of Hcy, TC, TG and LDL-C in patients in the study group were significantly higher than that in control group, while the HDL-C level was significantly lower than that in control group, and the differences were statistically significant (P<0.001). The difference was statistically significant in the levels of Hcy, TC, TG and LDL-C between acute and old myocardial infarction and angina pectoris group (P<0.05), and the difference was statistically significant in the levels of Hcy, TC, TG and LDL-C between old and acute myocardial infarction group (P<0.05). Analysis of risk factors for the disease was performed using logistic regression. The results of univariate analysis showed that CHD was associated with Hcy, hyperlipidemia, smoking, hypertension and diabetes (P<0.05), but had no relationship with drinking and obesity (P>0.05). The results of multivariate analysis showed that Hcy, hyperlipidemia, hypertension and diabetes were independent risk factors for CHD, and the difference was statistically significant (P<0.05). The Hcy level increased and the HDL-C level decreased in serum of CHD patients. Hcy, hyperlipidemia, hypertension and diabetes are independent risk factors for CHD, which can provide practical basis for the diagnosis, treatment and prevention.
Collapse
Affiliation(s)
- Weijing Liu
- Department of Emergency, Weifang People's Hospital, Weifang, Shandong 261000, P.R. China
| | - Tao Wang
- Department of Emergency, Weifang People's Hospital, Weifang, Shandong 261000, P.R. China
| | - Pengfei Sun
- Department of Emergency, Weifang People's Hospital, Weifang, Shandong 261000, P.R. China
| | - Yanju Zhou
- Department of Emergency, Weifang People's Hospital, Weifang, Shandong 261000, P.R. China
| |
Collapse
|
40
|
Constantin MM, Nita IE, Olteanu R, Constantin T, Bucur S, Matei C, Raducan A. Significance and impact of dietary factors on systemic lupus erythematosus pathogenesis. Exp Ther Med 2018; 17:1085-1090. [PMID: 30679978 DOI: 10.3892/etm.2018.6986] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 08/14/2018] [Indexed: 12/12/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease of unknown etiology, although its mechanisms involve genetic, epigenetic and environmental risk factors. Considering that SLE pathogenesis is yet to be explored, recent studies aimed to investigate the impact of diet, in terms of triggering or altering the course of the disease. To study the impact of diet on SLE pathogenesis, we conducted a search on Pubmed using the keywords 'diet and autoimmune diseases', 'diet and lupus', 'caloric restriction and lupus', 'polyunsaturated fatty acids and lupus', 'vitamin D and lupus', 'vitamin C and lupus' 'vitamin E and lupus' 'vitamin A and lupus' 'vitamin B and lupus', 'polyphenols and lupus', 'isoflavones and lupus', 'minerals and lupus', 'aminoacids and lupus', 'curcumin and lupus' and found 10,215 papers, from which we selected 47 relevant articles. The paper clearly emphasizes the beneficial role of personalized diet in patients with SLE, and the information presented could be used in daily practice. Proper diet in SLE can help preserve the body's homeostasis, increase the period of remission, prevent adverse effects of medication and improve the patient's physical and mental well-being.
Collapse
Affiliation(s)
- Maria-Magdalena Constantin
- University of Medicine and Pharmacy 'Carol Davila', 050474 Bucharest, Romania.,The Second Department of Dermatology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Iuliana Elena Nita
- The Second Department of Dermatology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Rodica Olteanu
- The Second Department of Dermatology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Traian Constantin
- University of Medicine and Pharmacy 'Carol Davila', 050474 Bucharest, Romania
| | - Stefana Bucur
- The Second Department of Dermatology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Clara Matei
- University of Medicine and Pharmacy 'Carol Davila', 050474 Bucharest, Romania
| | - Anca Raducan
- Dr. Anca Răducan Anti-Aging Dermatology Clinic, 900162 Constanta, Romania
| |
Collapse
|
41
|
Li H, Tong Q, Guo L, Yu S, Li Y, Cao Q, Li J, Li F. Risk of Coronary Artery Disease in Patients With Systemic Lupus Erythematosus: A Systematic Review and Meta-analysis. Am J Med Sci 2018; 356:451-463. [PMID: 30241668 DOI: 10.1016/j.amjms.2018.08.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 07/26/2018] [Accepted: 08/01/2018] [Indexed: 01/11/2023]
Abstract
BACKGROUND The association between chronic inflammation and the accelerated development of atherosclerosis is well recognized. However, it remains controversial as to whether the risk of coronary artery disease (CAD) is elevated in patients with systemic lupus erythematosus (SLE). The objective of this meta-analysis was to obtain a better estimate of the risk of CAD in patients with SLE. METHODS An English-restricted literature review was conducted according to PRISMA guidelines using key databases, surveying all articles published through October 31, 2017. Specific search terms included "SLE" and "coronary artery disease" as well as appropriate MeSH terms. The Newcastle-Ottawa scale was used for quality assessment. RESULTS Nine studies were identified and included in this meta-analysis. The pooled risk ratio of CAD in patients with SLE was 3.39 (95% CI: 2.15-5.35). The statistical heterogeneity of this meta-analysis was high, with an I2 value of 79.5%. An elevated risk of CAD was consistently observed in both female and male SLE patients (pooled risk ratio: 3.27 [95% CI: 2.01-5.30] and 3.16 [95% CI: 2.02-4.94], respectively). CONCLUSIONS SLE patients are at significantly higher risk of developing CAD. However, as relatively few studies were available for incorporation into this meta-analysis, there is a clear need for further studies with larger sample sizes that better parse gender-related differences in CAD susceptibility among SLE patients. Future work to standardize cardiovascular risk factor identification and monitoring in SLE patients is also needed.
Collapse
Affiliation(s)
- Huimin Li
- School of Nursing, Jilin University, Changchun City, Jilin Province, China
| | - Qian Tong
- School of Nursing, Jilin University, Changchun City, Jilin Province, China
| | - Lirong Guo
- School of Nursing, Jilin University, Changchun City, Jilin Province, China
| | - Shui Yu
- School of Nursing, Jilin University, Changchun City, Jilin Province, China
| | - Yuewei Li
- School of Nursing, Jilin University, Changchun City, Jilin Province, China
| | - Qinqin Cao
- School of Nursing, Jilin University, Changchun City, Jilin Province, China
| | - Jinwei Li
- School of Nursing, Jilin University, Changchun City, Jilin Province, China
| | - Feng Li
- School of Nursing, Jilin University, Changchun City, Jilin Province, China.
| |
Collapse
|
42
|
Monção CSA, Martins LN, Penteado MPS, Reis RCP, Santos FMM, Lanna CCD, Ribeiro AL, Telles RW. Incidence of cardiovascular risk factors in female patients with systemic lupus erythematosus: a 3-year follow-up cohort. Lupus 2018; 27:1790-1798. [DOI: 10.1177/0961203318790676] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Objectives To evaluate the incidence and variability of traditional coronary artery disease (CAD) risk factors in a cohort of lupus patients and to investigate if prednisone use predicts an increase in the number of risk factors. Methods A total of 151 women, 37.8 ± 11.1 (mean ± SD) years old at baseline, were reevaluated after a median period of 39 (interquartile range 36.5–42.0) months. The cumulative incidence of traditional risk factors, the incidence rate (with 95% confidence interval) of hypertension, diabetes, dyslipidemia and hypertriglyceridemia, and the frequency of the risk factors’ disappearance were calculated. Metabolic syndrome (MetS) and Framingham risk score (FRS) were computed. Logistic regression was used to investigate if maximum or cumulative prednisone dose used during follow-up predicted an increase in the cardiometabolic risk factors’ number. Results The cumulative incidence of risk factors varied from 39.1% (abdominal obesity) to zero (smoking), and the incidence rate varied from 133.2 (87.8–178.6) per 1000 person-years (dyslipidemia) to 10.4 (1.3–19.5) per 1000 person-years (diabetes). The cumulative incidence for MetS was 18.8%, and 11.7% of 143 patients with low FRS at baseline (T1) were classified in the high-risk category at the end of the study (T2). Dyslipidemia was the most variable risk factor, with 43.5% disappearance at T2. The maximum prednisone dose used during follow-up was borderline ( p = 0.050) for prediction of an increase in the number of cardiometabolic risk factors in an adjusted model for antimalarial use, modified Systemic Lupus Erythematosus Disease Activity Index 2000 (SLEDAI-2K) and age. Conclusion The authors described high incidence and variability of CAD risk factors in female lupus patients, with higher prednisone dose being borderline for an increase in the number of cardiometabolic risk factors.
Collapse
Affiliation(s)
- C S A Monção
- Universidade Federal de Minas Gerais, Faculdade de Medicina Belo Horizonte, Minas Gerais, Brazil
| | - L N Martins
- Universidade Federal de Minas Gerais, Faculdade de Medicina Belo Horizonte, Minas Gerais, Brazil
| | - M P S Penteado
- Universidade Federal de Minas Gerais, Faculdade de Medicina Belo Horizonte, Minas Gerais, Brazil
| | - R C P Reis
- Universidade Federal de Minas Gerais, Faculdade de Medicina Belo Horizonte, Minas Gerais, Brazil
| | - F M M Santos
- Universidade Federal de Minas Gerais, Faculdade de Medicina Belo Horizonte, Minas Gerais, Brazil
| | - C C D Lanna
- Universidade Federal de Minas Gerais, Faculdade de Medicina Belo Horizonte, Minas Gerais, Brazil
| | - A L Ribeiro
- Universidade Federal de Minas Gerais, Faculdade de Medicina Belo Horizonte, Minas Gerais, Brazil
| | - R W Telles
- Universidade Federal de Minas Gerais, Faculdade de Medicina Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
43
|
Influence of autoimmunity and inflammation on endothelial function and thrombosis in systemic lupus erythematosus patients. Clin Rheumatol 2018; 37:2087-2093. [PMID: 29675623 DOI: 10.1007/s10067-018-4104-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/28/2018] [Accepted: 04/10/2018] [Indexed: 10/17/2022]
Abstract
The aim of this study is to assess the relationship between autoimmunity and endothelial activation/damage (ICAM-1 and vWF serum levels) and the degree of prothrombotic activity (thrombin-antithrombin complexes-TAT serum levels) in SLE. In 60 clinically stable SLE patients, levels of the following parameters were estimated in their serum: lupus anticoagulant (LA), anticardiolipin antibodies in both IgG and IgM classes (aCL-IgG and aCL-IgM, respectively), antiβ2GPI antibodies in both IgG and IgM classes (antiβ2GPI-IgG and antiβ2GPI-IgM, respectively), ICAM, von Willebrand factor (vWF), TAT, CRP, C3c, C4, and IL-6. ICAM-1 values exceeded the upper reference limit in 9 (15%) patients. vWF levels were increased in 21 (35%) patients. In all patients with elevated ICAM-1 values, vWF were also increased. TAT concentrations were elevated in 12 (20%) people. ICAM-1 were significantly higher in patients with elevated aCL-IgM (> 30 MPL vs ≤ 30 MPL; p < 0.05). Similarly, ICAM-1 were significantly higher in patients with elevated antiβ2-GPI-IgM (> 20 SMU vs ≤ 20 SMU; p < 0.05). There was no significant difference in ICAM-1 levels in relation to LA-positivity. vWF were not significantly different in relation to antiphospholipid antibodies nor the inflammation marker levels. TAT were significantly higher in patients with elevated aCL-IgM (> 30 MPL vs ≤ 30 MPL; p < 0.05). In one third of young patients with stable SLE, signs of endothelial activation/damage were found, as shown by elevated plasma ICAM-1 or vWF. Increased prothrombotic tendency manifested by elevated TAT was found in one fifth of the patients. Elevated anticardiolipin (IgM) and anti-β2-glycoprotein I (IgM) antibodies influence endothelial dysfunction and enhance prothrombotic state.
Collapse
|
44
|
Impact of Diabetes Mellitus on the Risk of End-Stage Renal Disease in Patients with Systemic Lupus Erythematosus. Sci Rep 2018; 8:6008. [PMID: 29662119 PMCID: PMC5902607 DOI: 10.1038/s41598-018-24529-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/05/2018] [Indexed: 02/07/2023] Open
Abstract
Systemic lupus erythematosus (SLE) patients are associated with insulin resistance and are at higher risk to develop diabetes mellitus (DM). SLE and DM could lead to renal failure respectively. However, it is unknown whether DM increases the risk of end-stage renal disease (ESRD) in SLE patients. This study aimed to evaluate potential synergistic effect of DM on SLE patients for development of ESRD. We conducted this study by using National Health Insurance Research Database of Taiwan. We recruited SLE patients with newly-diagnosed DM as the study cohort. A comparison cohort at a 1:1 ratio of SLE patients without DM matched by age, sex, age at the diagnosis of SLE, duration between diagnosis of SLE and DM, and various comorbidities through propensity score matching were recruited. After 5.01 ± 3.13 years follow-up, the incidence of ESRD was significantly higher in the DM group than in the non-DM group (Incidence rate ratio: 2.71; 95% CI: 1.70-4.32). After control of confounding factors, DM was not an independent risk factor of ESRD. After starting dialysis, DM patients had a similar mortality rate to those without DM. In summary, SLE patients superimposed with subsequent DM are associated with potentially higher risk to develop ESRD.
Collapse
|
45
|
Towards the Clinical Management of Cardiac Involvement in Systemic Inflammatory Conditions—a Central Role for CMR. CURRENT CARDIOVASCULAR IMAGING REPORTS 2018. [DOI: 10.1007/s12410-018-9451-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
46
|
Protective Effects of Hydroxychloroquine against Accelerated Atherosclerosis in Systemic Lupus Erythematosus. Mediators Inflamm 2018; 2018:3424136. [PMID: 29670462 PMCID: PMC5835241 DOI: 10.1155/2018/3424136] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 12/10/2017] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular (CV) morbidity and mortality are a challenge in management of patients with systemic lupus erythematosus (SLE). Higher risk of CV disease in SLE patients is mostly related to accelerated atherosclerosis. Nevertheless, high prevalence of traditional cardiovascular risk factors in SLE patients does not fully explain the increased CV risk. Despite the pathological bases of accelerated atherosclerosis are not fully understood, it is thought that this process is driven by the complex interplay between SLE and atherosclerosis pathogenesis. Hydroxychloroquine (HCQ) is a cornerstone in treatment of SLE patients and has been thought to exert a broad spectrum of beneficial effects on disease activity, prevention of damage accrual, and mortality. Furthermore, HCQ is thought to protect against accelerated atherosclerosis targeting toll-like receptor signaling, cytokine production, T-cell and monocyte activation, oxidative stress, and endothelial dysfunction. HCQ was also described to have beneficial effects on traditional CV risk factors, such as dyslipidemia and diabetes. In conclusion, despite lacking randomized controlled trials unambiguously proving the protection of HCQ against accelerated atherosclerosis and incidence of CV events in SLE patients, evidence analyzed in this review is in favor of its beneficial effect.
Collapse
|
47
|
Fatemi A, Ghanbarian A, Sayedbonakdar Z, Kazemi M, Smiley A. Metabolic syndrome in Iranian patients with systemic lupus erythematosus and its determinants. Clin Rheumatol 2018; 37:1521-1528. [PMID: 29305669 DOI: 10.1007/s10067-017-3970-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/27/2017] [Accepted: 12/22/2017] [Indexed: 11/28/2022]
Abstract
The aim of this study was to determine the prevalence of metabolic syndrome (MetS) in Iranian patients with systemic lupus erythematosus (SLE) and its determinants. In a cross-sectional study, 98 patients with SLE and 95 controls were enrolled. Prevalence of MetS was determined based on American Heart Association and National Heart, Lung, and Blood Institute (AHA/NHLBI) and 2009 harmonizing criteria. In addition, demographic features and lupus characteristics such as disease duration, pharmacological treatment, laboratory data, SLE disease activity index (SLEDAI), and Systemic Lupus International Collaborating Clinics/American College of Rheumatology Damage index (SDI) were recorded. The predictors of MetS were obtained by backward stepwise regression analysis. Using AHA/NHLBI, MetS was observed in 35 (35.7%) patients and 28 (29.8%) controls (P = 0.4). Using harmonizing criteria, MetS was observed in 37 (37.7%) patients and 33 (35.1%) controls (P = 0.7). There was no difference in frequency distribution of MetS components between the patients and the controls. In multivariate regression analysis, low C3, blood urea nitrogen (BUN), and body mass index were independent determinants of MetS in lupus patients. BUN, low C3, and body mass index were the major determinants of MetS in lupus patients.
Collapse
Affiliation(s)
- Alimohammad Fatemi
- Department of Rheumatology, Alzahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Azadeh Ghanbarian
- Department of Internal Medicine, Alzahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Sayedbonakdar
- Department of Rheumatology, Alzahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehdi Kazemi
- Department of Internal Medicine, Alzahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abbas Smiley
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University, Bloomington, IN, USA
| |
Collapse
|
48
|
Managing conventional cardiovascular risk factors in the lupus clinic – what can we achieve? Lupus 2018. [DOI: 10.1177/0961203317717630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
49
|
|
50
|
Baragetti A, Ramirez GA, Magnoni M, Garlaschelli K, Grigore L, Berteotti M, Scotti I, Bozzolo E, Berti A, Camici PG, Catapano AL, Manfredi AA, Ammirati E, Norata GD. Disease trends over time and CD4 +CCR5 + T-cells expansion predict carotid atherosclerosis development in patients with systemic lupus erythematosus. Nutr Metab Cardiovasc Dis 2018; 28:53-63. [PMID: 29150407 DOI: 10.1016/j.numecd.2017.09.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 09/07/2017] [Accepted: 09/09/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIM Patients with Systemic Lupus Erythematosus (SLE) present increased cardiovascular mortality compared to the general population. Few studies have assessed the long-term development and progression of carotid atherosclerotic plaque in SLE patients. Our aim was to investigate the association of clinical and laboratory markers of disease activity and classical cardiovascular risk factors (CVRF) with carotid atherosclerosis development in SLE patients in a prospective 5-year study. METHODS AND RESULTS Clinical history and information on principal CVRFs were collected at baseline and after 5 years in 40 SLE patients (36 women, mean age 42 ± 9 years; 14.4 ± 7 years of mean disease duration) and 50 age-matched controls. Carotid Doppler ultrasonography was employed to quantify the atherosclerotic burden at baseline and at follow up. Clinimetrics were applied to assess SLE activity over time (SLEDAI). The association between basal circulating T cell subsets (including CD4+CCR5+; CD4+CXCR3+; CD4+HLADR+; CD4+CD45RA+RO-, CD4+CD45RO+RA- and their subsets) and atherosclerosis development was evaluated. During the 5-year follow up, 32% of SLE patients, developed carotid atherosclerosis compared to 4% of controls. Furthermore, considering SLEDAI changes over time, patients within the highest tertile were those with increased incidence of carotid atherosclerosis independently of CVRF. In addition, increased levels of CD4+CCR5+ T cells were independently associated with the development of carotid atherosclerosis in SLE patients. CONCLUSION Serial clinical evaluations over time, rather than a single point estimation of disease activity or CVRF burden, are required to define the risk of carotid atherosclerosis development in SLE patients. Specific T cell subsets are associated with long-term atherosclerotic progression and may further be of help in predicting vascular disease progression.
Collapse
Affiliation(s)
- A Baragetti
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy; Center for the Study of Atherosclerosis - Bassini Hospital, Cinisello Balsamo, Italy
| | - G A Ramirez
- Università Vita-Salute San Raffaele, Milan, Italy; Unit of Medicine and Clinical Immunology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - M Magnoni
- Department of Thoracic and Cardiovascular Surgery, Università Vita-Salute San Raffaele Scientific Institute Milan, Italy
| | - K Garlaschelli
- Center for the Study of Atherosclerosis - Bassini Hospital, Cinisello Balsamo, Italy
| | - L Grigore
- Center for the Study of Atherosclerosis - Bassini Hospital, Cinisello Balsamo, Italy; IRCCS - Multimedica Hospital, Sesto San Giovanni, Italy
| | - M Berteotti
- Department of Thoracic and Cardiovascular Surgery, Università Vita-Salute San Raffaele Scientific Institute Milan, Italy
| | - I Scotti
- Department of Thoracic and Cardiovascular Surgery, Università Vita-Salute San Raffaele Scientific Institute Milan, Italy
| | - E Bozzolo
- Università Vita-Salute San Raffaele, Milan, Italy
| | - A Berti
- Università Vita-Salute San Raffaele, Milan, Italy; Unit of Medicine and Clinical Immunology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - P G Camici
- Department of Thoracic and Cardiovascular Surgery, Università Vita-Salute San Raffaele Scientific Institute Milan, Italy
| | - A L Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy; IRCCS - Multimedica Hospital, Sesto San Giovanni, Italy
| | - A A Manfredi
- Università Vita-Salute San Raffaele, Milan, Italy; Unit of Medicine and Clinical Immunology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - E Ammirati
- Niguarda Ca' Granda Hospital, Milan, Italy
| | - G D Norata
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy; Center for the Study of Atherosclerosis - Bassini Hospital, Cinisello Balsamo, Italy; School of Biomedical Sciences, Curtin Health Innovation Research Institute, Faculty of Health Science, Curtin University, Perth, Western Australia, Australia.
| |
Collapse
|