1
|
Alonaizan A, Altamimi A, Alnaqla H, Alanazi MS, Benragosh N, Alanazi A, Alanazi F, Aldakhil L, Al Ghanim N. Hyperlipidemia in Saudi Arabian Patients With Systemic Lupus Erythematosus. Cureus 2024; 16:e74653. [PMID: 39735160 PMCID: PMC11681606 DOI: 10.7759/cureus.74653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2024] [Indexed: 12/31/2024] Open
Abstract
Background Systemic lupus erythematosus (SLE) is a chronic autoimmune disease associated with increased cardiovascular risk, partly due to dyslipidemia. This study aimed to evaluate the lipid profiles of Saudi Arabian patients with SLE and examine the impact of hydroxychloroquine (HCQ) and steroid use on these profiles, with a particular focus on patients with lupus nephritis. Methods A retrospective observational study was conducted at King Saud Medical City, Riyadh, Saudi Arabia, including SLE patients treated at the hospital's rheumatology clinic between July 2023 and December 2023. Patients aged 15-80 years diagnosed with SLE per the American College of Rheumatology revised criteria were included. Exclusion criteria comprised menopausal or pregnant women, individuals with significant comorbid conditions, and those on specific medications. Lipid profiles, including total cholesterol, low-density lipoprotein (LDL) cholesterol, high-density lipoprotein (HDL) cholesterol, triglycerides, and very low-density lipoprotein (VLDL) cholesterol, were analyzed, and correlations with disease activity parameters were assessed using STATA software (StataCorp LLC, College Station, TX, US). Statistical analyses included Wilcoxon signed-rank tests and Spearman rank correlations. Results The study included 138 SLE patients (84.5% females, mean age 37.13 ± 12.9 years). Lipid profiles showed varied results: mean total cholesterol was 4.48 mmol/L, LDL 2.56 mmol/L, HDL 1.32 mmol/L, triglycerides 1.40 mmol/L, and VLDL 0.63 mmol/L. HCQ use was associated with higher, albeit not statistically significant, lipid levels. Steroid use did not show significant effects on lipid levels. Patients with lupus nephritis had higher triglyceride and VLDL levels compared to those without nephritis (p = 0.02). No significant differences were observed in lipid profiles between patients with and without anti-double-stranded DNA (dsDNA) antibodies. Significant correlations were found between triglycerides and C-reactive protein (CRP), creatinine, and erythrocyte sedimentation rate (ESR). Conclusion This study highlights the complex relationship between SLE, dyslipidemia, and treatment. While HCQ use did not significantly alter lipid profiles, lupus nephritis was associated with worse lipid abnormalities. These findings underscore the need for ongoing monitoring and targeted management of lipid profiles in SLE patients to mitigate cardiovascular risk.
Collapse
Affiliation(s)
- Alham Alonaizan
- Rheumatology, Internal Medicine, Security Forces Hospital Program, Riyadh, SAU
| | | | | | | | | | | | - Fahad Alanazi
- Internal Medicine, King Saud Medical City, Riyadh, SAU
| | | | | |
Collapse
|
2
|
Pérez‐Ocampo J, Taborda NA, Yassin LM, Higuita‐Gutiérrez LF, Hernandez JC. Exploring the Association Between Systemic Lupus Erythematosus and High-Density Lipoproteins: A Systematic Review and Meta-Analysis. ACR Open Rheumatol 2024; 6:648-661. [PMID: 39030864 PMCID: PMC11471950 DOI: 10.1002/acr2.11700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 05/06/2024] [Accepted: 05/14/2024] [Indexed: 07/22/2024] Open
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is an autoimmune disease with inflammation as a critical feature. Recently, high-density lipoprotein cholesterol (HDLc) have been evidenced to have anti-inflammatory effects, suggesting a potential link between HDL and SLE that needs to be thoroughly studied. The aim was to explore the association between SLE and HDLc through a systematic review with meta-analysis. METHODS A systematic review with meta-analysis was conducted to assess mean differences in HDL levels between patients with SLE and healthy controls. Both qualitative and quantitative syntheses were performed, including an assessment of heterogeneity using I2, a publication bias evaluation, a methodologic quality assessment, and a forest plot under a random effects model. Subgroup analyses were conducted based on disease activity and the report of corticosteroid dosage. RESULTS A total of 53 studies were included in the qualitative synthesis, and 35 studies were included in the quantitative synthesis, comprising 3,002 patients with SLE and 2,123 healthy controls. Mean HDL levels were found to be lower in patients with SLE as follows: in the meta-analysis including all articles -6.55 (95% confidence interval [CI] -8.77 to -4.33); in patients with mild disease activity -5.46 (95% CI -8.26 to -2.65); in patients with moderate or severe disease activity -9.42 (95% CI -15.49 to -3.34); in patients using corticosteroids -5.32 (95% CI -10.35 to -0.29); and in studies with excellent methodologic quality -8.71 (95% CI -12.38 to -5.03). CONCLUSION HDL levels appear to be quantitatively altered in patients with SLE, suggesting a potential contribution to immune dysregulation, highlighting the importance of HDL in autoimmune diseases.
Collapse
Affiliation(s)
- Julián Pérez‐Ocampo
- Infettare, Facultad de MedicinaUniversidad Cooperativa de ColombiaMedellínColombia
| | - Natalia A. Taborda
- Grupo de Investigaciones Biomédicas Uniremington, Programa de Medicina, Facultad de Ciencias de la SaludCorporación Universitaria RemingtonMedellínColombia
| | - Lina M. Yassin
- Grupo de Investigaciones Biomédicas Uniremington, Programa de Medicina, Facultad de Ciencias de la SaludCorporación Universitaria RemingtonMedellínColombia
| | - Luis Felipe Higuita‐Gutiérrez
- Infettare, Facultad de MedicinaUniversidad Cooperativa de ColombiaMedellínColombia
- Escuela de microbiología, Universidad de Antioquia UdeAMedellinColombia
| | - Juan C. Hernandez
- Infettare, Facultad de MedicinaUniversidad Cooperativa de ColombiaMedellínColombia
- Grupo Inmunovirología, Facultad de Medicina (UdeA)Universidad de Antioquia UdeAMedellínColombia
| |
Collapse
|
3
|
Hu W, Chu T, Liao H, Wang W, Ha J, Kiburg K, Zhang X, Shang X, Huang Y, Zhang X, Tang S, Hu Y, Yu H, Yang X, He M, Zhu Z. Distinct and Overlapping Metabolites Associated with Visual Impairment and Cognitive Impairment. J Alzheimers Dis Rep 2024; 8:1093-1104. [PMID: 39434817 PMCID: PMC11491940 DOI: 10.3233/adr-230154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 06/27/2024] [Indexed: 10/23/2024] Open
Abstract
Background Previous studies found that visual impairment (VI) is associated with higher risk of cognitive impairment, but the molecular basis of these conditions is unknown. Objective We aim to compare the metabolite associations of VI and cognitive impairment. Methods The study population with comprehensive measurements was derived from the UK Biobank study. Visual acuity worse than 0.3 logMAR units were defined as VI. Failure in one or more of the four cognitive tests was defined as cognitive impairment. A panel of 249 metabolites was measured using a nuclear magnetic resonance metabolites profiling platform. Logistic regression models were applied to compare metabolite associations with VI and cognitive impairment. Results 23,775 participants with complete data on visual acuity, cognitive tests and metabolomics, and without a history of neurological disorders at baseline were included. After adjusting for confounding factors, VI was significantly associated with cognitive impairment (odds ratio[OR] = 1.49, 95% confidence interval [CI]: 1.27-1.74, p < 0.001). After multiple testing correction (p < 9×10-4), five metabolites including the ratio of omega-6 to omega-3 fatty acids (FAs) (OR = 1.18[1.10-1.27]), ratio of omega-3 to total FAs (OR = 0.84[0.77-0.91]), ratio of docosahexaenoic acid (DHA) to total FAs (OR = 0.86[0.80-0.94]), DHA (OR = 0.85[0.78-0.92]), and omega-3 FAs (OR = 0.84[0.77-0.91]) were uniquely associated with VI. Glycoprotein acetyls (OR = 1.06[1.03-1.10]) and alanine (OR = 0.95[0.92-0.98]) were exclusively associated with cognitive impairment. Albumin was identified as the common metabolite shared by the two phenotypes (OR = 0.90[0.85-0.95] for VI, and 0.95[0.92-0.98]) for cognitive impairment). Conclusions We identified distinct and overlapping metabolites associated with VI and cognitive impairment, unveiling their distinct metabolic profiles and potential common pathophysiology.
Collapse
Affiliation(s)
- Wenyi Hu
- Department of Ophthalmology, Guangdong Academy of Medical Sciences, Guangdong Provincial People’s Hospital, Guangzhou, China
- Centre for Eye Research Australia, Ophthalmology, University of Melbourne, Melbourne, Australia
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia
| | | | - Huan Liao
- Neural Regeneration Group, Institute of Reconstructive Neurobiology, University of Bonn, Bonn, Germany
| | - Wei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jason Ha
- Centre for Eye Research Australia, Ophthalmology, University of Melbourne, Melbourne, Australia
| | - Katerina Kiburg
- Centre for Eye Research Australia, Ophthalmology, University of Melbourne, Melbourne, Australia
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia
| | - Xiayin Zhang
- Department of Ophthalmology, Guangdong Academy of Medical Sciences, Guangdong Provincial People’s Hospital, Guangzhou, China
| | - Xianwen Shang
- Department of Ophthalmology, Guangdong Academy of Medical Sciences, Guangdong Provincial People’s Hospital, Guangzhou, China
- Centre for Eye Research Australia, Ophthalmology, University of Melbourne, Melbourne, Australia
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China
| | - Yu Huang
- Department of Ophthalmology, Guangdong Academy of Medical Sciences, Guangdong Provincial People’s Hospital, Guangzhou, China
| | - Xueli Zhang
- Department of Ophthalmology, Guangdong Academy of Medical Sciences, Guangdong Provincial People’s Hospital, Guangzhou, China
| | - Shulin Tang
- Department of Ophthalmology, Guangdong Academy of Medical Sciences, Guangdong Provincial People’s Hospital, Guangzhou, China
| | - Yijun Hu
- Department of Ophthalmology, Guangdong Academy of Medical Sciences, Guangdong Provincial People’s Hospital, Guangzhou, China
| | - Honghua Yu
- Department of Ophthalmology, Guangdong Academy of Medical Sciences, Guangdong Provincial People’s Hospital, Guangzhou, China
| | - Xiaohong Yang
- Department of Ophthalmology, Guangdong Academy of Medical Sciences, Guangdong Provincial People’s Hospital, Guangzhou, China
| | - Mingguang He
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China
- Research Centre for SHARP Vision, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Zhuoting Zhu
- Department of Ophthalmology, Guangdong Academy of Medical Sciences, Guangdong Provincial People’s Hospital, Guangzhou, China
- Centre for Eye Research Australia, Ophthalmology, University of Melbourne, Melbourne, Australia
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia
| |
Collapse
|
4
|
Xiong W, Yeo T, May JTM, Demmers T, Ceronie B, Ramesh A, McGinty RN, Michael S, Torzillo E, Sen A, Anthony DC, Irani SR, Probert F. Distinct plasma metabolomic signatures differentiate autoimmune encephalitis from drug-resistant epilepsy. Ann Clin Transl Neurol 2024; 11:1897-1908. [PMID: 39012808 PMCID: PMC11251473 DOI: 10.1002/acn3.52112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/24/2024] [Accepted: 05/17/2024] [Indexed: 07/18/2024] Open
Abstract
OBJECTIVE Differentiating forms of autoimmune encephalitis (AE) from other causes of seizures helps expedite immunotherapies in AE patients and informs studies regarding their contrasting pathophysiology. We aimed to investigate whether and how Nuclear Magnetic Resonance (NMR)-based metabolomics could differentiate AE from drug-resistant epilepsy (DRE), and stratify AE subtypes. METHODS This study recruited 238 patients: 162 with DRE and 76 AE, including 27 with contactin-associated protein-like 2 (CASPR2), 29 with leucine-rich glioma inactivated 1 (LGI1) and 20 with N-methyl-d-aspartate receptor (NMDAR) antibodies. Plasma samples across the groups were analyzed using NMR spectroscopy and compared with multivariate statistical techniques, such as orthogonal partial least squares discriminant analysis (OPLS-DA). RESULTS The OPLS-DA model successfully distinguished AE from DRE patients with a high predictive accuracy of 87.0 ± 3.1% (87.9 ± 3.4% sensitivity and 86.3 ± 3.6% specificity). Further, pairwise OPLS-DA models were able to stratify the three AE subtypes. Plasma metabolomic signatures of AE included decreased high-density lipoprotein (HDL, -(CH2)n-, -CH3), phosphatidylcholine and albumin (lysyl moiety). AE subtype-specific metabolomic signatures were also observed, with increased lactate in CASPR2, increased lactate, glucose, and decreased unsaturated fatty acids (UFA, -CH2CH=) in LGI1, and increased glycoprotein A (GlycA) in NMDAR-antibody patients. INTERPRETATION This study presents the first non-antibody-based biomarker for differentiating DRE, AE and AE subtypes. These metabolomics signatures underscore the potential relevance of lipid metabolism and glucose regulation in these neurological disorders, offering a promising adjunct to facilitate the diagnosis and therapeutics.
Collapse
Affiliation(s)
- Wenzheng Xiong
- Department of ChemistryUniversity of OxfordOxfordUK
- Department of Pharmacology, Medical Sciences DivisionUniversity of OxfordOxfordUK
| | - Tianrong Yeo
- Department of Pharmacology, Medical Sciences DivisionUniversity of OxfordOxfordUK
- Department of NeurologyNational Neuroscience InstituteSingaporeSingapore
- Duke‐NUS Medical SchoolSingaporeSingapore
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
| | - Jeanne Tan May May
- Department of NeurologyNational Neuroscience InstituteSingaporeSingapore
- Duke‐NUS Medical SchoolSingaporeSingapore
| | - Tor Demmers
- Department of Pharmacology, Medical Sciences DivisionUniversity of OxfordOxfordUK
| | - Bryan Ceronie
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Archana Ramesh
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Ronan N. McGinty
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Sophia Michael
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Emma Torzillo
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Arjune Sen
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Daniel C. Anthony
- Department of Pharmacology, Medical Sciences DivisionUniversity of OxfordOxfordUK
| | - Sarosh R. Irani
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
- Department of NeurologyJohn Radcliffe Hospital, Oxford University HospitalsOxfordUK
- Departments of Neurology and NeurosciencesMayo ClinicJacksonvilleFloridaUSA
| | - Fay Probert
- Department of ChemistryUniversity of OxfordOxfordUK
| |
Collapse
|
5
|
Akiash N, Abbaspour S, Mowla K, Moradi A, Madjidi S, Sharifi P, Pazoki M. Three-dimensional speckle tracking echocardiography for evaluation of ventricular function in patients with systemic lupus erythematosus: relationship between duration of lupus erythematosus and left ventricular dysfunction by using global longitudinal strain. Egypt Heart J 2024; 76:79. [PMID: 38914877 PMCID: PMC11196547 DOI: 10.1186/s43044-024-00511-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 06/19/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND Cardiovascular diseases are leading causes of morbidity and mortality in patients with systemic lupus erythematosus (SLE). Cardiac involvement in SLE can often go undetected. Three-dimensional (3D) speckle tracking echocardiography (STE) is a noninvasive imaging technique that can assess the function of the heart's ventricles in an accurate and reproducible way. This makes it an attractive option for detecting early signs of heart disease in SLE patients. By identifying these subclinical cardiac abnormalities, 3D-STE may help reduce the negative impact of cardiovascular diseases in SLE population. Therefore, this study aimed to compare the left ventricular (LV) function between patients with SLE compared to age- and gender-matched controls using two-dimensional (2D) and 3D-STE. RESULTS The current study found no significant differences in left ventricle ejection fraction, left ventricle end-diastolic volume, left ventricle end-systolic volume, left ventricle end-diastolic mass, and left ventricle end-systolic mass between the two groups. However, the SLE group exhibited a significantly lower LV global longitudinal strain (GLS) compared to the control group according to all types of echocardiographic assessments, including 3D and 2D long-axis strain, apical 2-chamber, and apical 4-chamber assessments (all P values < 0.05). Furthermore, a good inter-rater reliability and intra-rater reliability were observed regarding the LVGLS measurement with 3D-STE. Additionally, the study identified a significant correlation between LVGLS and SLE duration (r (50) = 0.46, P < 0.001). The use of prednisolone and nephrology disorders was also found to impact LVGLS measurements. CONCLUSIONS Despite a normal LVEF in patients with SLE, LVGLS measurements indicated that LV systolic dysfunction was observed more frequently in SLE patients compared to their healthy counterparts. Therefore, advanced 3D-STE techniques may be useful in identifying subtle abnormalities in LV function in SLE patients.
Collapse
Affiliation(s)
- Nehzat Akiash
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Golestan Blvd., Ahvaz, Iran
| | - Somayeh Abbaspour
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Golestan Blvd., Ahvaz, Iran
| | - Karim Mowla
- Department of Rheumatology, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Amir Moradi
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Golestan Blvd., Ahvaz, Iran.
| | | | - Parisa Sharifi
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Golestan Blvd., Ahvaz, Iran
| | - Mahboubeh Pazoki
- Department of Cardiology, School of Medicine, Hazarat-e Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Chen Y, Tao T, Liang Z, Chen X, Xu Y, Zhang T, Zhou D. Prednisone combined with Dihydroartemisinin attenuates systemic lupus erythematosus by regulating M1/M2 balance through the MAPK signaling pathway. Mol Immunol 2024; 170:144-155. [PMID: 38669759 DOI: 10.1016/j.molimm.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/02/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024]
Abstract
OBJECTIVE Dihydroartemisinin (DHA) plays a very important role in various diseases. However, the precise involvement of DHA in systemic lupus erythematosus (SLE), relation to the equilibrium between M1 and M2 cells, remains uncertain. Therefore, we aimed to investigate the role of DHA in SLE and its effect on the M1/M2 cells balance. METHODS SLE mice model was established by pristane induction. Flow cytometry was employed to measure the abundance of M1 and M2 cells within the peripheral blood of individuals diagnosed with SLE. The concentrations of various cytokines, namely TNF-α, IL-1β, IL-4, IL-6, and IL-10, within the serum of SLE patients or SLE mice were assessed via ELISA. Immunofluorescence staining was utilized to detect the deposition of IgG and complement C3 in renal tissues of the mice. We conducted immunohistochemistry analysis to assess the expression levels of Collagen-I, a collagen protein, and α-SMA, a fibrosis marker protein, in the renal tissues of mice. Hematoxylin-eosin staining, Masson's trichrome staining, and Periodic acid Schiff staining were used to examine histological alterations. In this study, we employed qPCR and western blot techniques to assess the expression levels of key molecular markers, namely CD80 and CD86 for M1 cells, as well as CD206 and Arg-1 for M2 cells, within kidney tissue. Additionally, we investigated the involvement of the MAPK signaling pathway. The Venny 2.1 online software tool was employed to identify shared drug-disease targets, and subsequently, the Cytoscape 3.9.2 software was utilized to construct the "disease-target-ingredient" network diagram. Protein-protein interactions of the target proteins were analyzed using the String database, and the network proteins underwent enrichment analysis for Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathways. RESULTS The results showed that an increase in M1 cells and a decrease in M2 cells within the peripheral blood of individuals diagnosed with SLE. Further analysis revealed that prednisone (PDN) combined with DHA can alleviate kidney damage and regulate the balance of M1 and M2 cells in both glomerular mesangial cells (GMC) and kidney. The MAPK signaling pathway was found to be involved in SLE kidney damage and M1/M2 balance in the kidney. Furthermore, PDN and/or DHA were found to inhibit the MAPK signaling pathway in GMC and kidney. CONCLUSION We demonstrated that PDN combined with DHA attenuates SLE by regulating M1/M2 balance through MAPK signaling pathway. These findings propose that the combination of PDN and DHA could serve as a promising therapeutic strategy for SLE, as it has the potential to mitigate kidney damage and reinstate the equilibrium of M1 and M2 cells.
Collapse
Affiliation(s)
- Yan Chen
- Department of Dermatology, Yangjiang People's Hospital, 42 Dongshan Road, Jiangcheng District, Yangjiang 529500, Guangdong, China.
| | - Tingjun Tao
- Department of Dermatology, Yangjiang People's Hospital, 42 Dongshan Road, Jiangcheng District, Yangjiang 529500, Guangdong, China
| | - Zhaoxin Liang
- The First Clinical Medical School, Southern Medical University, 1838 North of Guangzhou Avenue, Baiyun, Guangzhou 510515, Guangdong, China
| | - Xiangnong Chen
- Department of hematopathology, The First Affiliated Hospital of Sun Yat-sen University, 58 ZhongshanEr Road, Yuexiu District, Guangzhou, China
| | - Ya'nan Xu
- Department of Dermatology, Yan'an People's Hospital, 16 Qilipu Street, Baota District, Yan'an, Shanxi, China
| | - Tangtang Zhang
- Department of Dermatology, The First Clinical Academy, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Dunrong Zhou
- Department of Intensive Care Unit, Yangjiang People's Hospital, 42 Dongshan Road, Jiangcheng District, Yangjiang 529500, Guangdong, China
| |
Collapse
|
7
|
Rojo-Sánchez A, Carmona-Martes A, Díaz-Olmos Y, Santamaría-Torres M, Cala MP, Orozco-Acosta E, Aroca-Martínez G, Pacheco-Londoño L, Navarro-Quiroz E, Pacheco-Lugo LA. Urinary metabolomic profiling of a cohort of Colombian patients with systemic lupus erythematosus. Sci Rep 2024; 14:9555. [PMID: 38664528 PMCID: PMC11045835 DOI: 10.1038/s41598-024-60217-0] [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: 12/22/2023] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune and multisystem disease with a high public health impact. Lupus nephritis (LN), commonly known as renal involvement in SLE, is associated with a poorer prognosis and increased rates of morbidity and mortality in patients with SLE. Identifying new urinary biomarkers that can be used for LN prognosis or diagnosis is essential and is part of current active research. In this study, we applied an untargeted metabolomics approach involving liquid and gas chromatography coupled with mass spectrometry to urine samples collected from 17 individuals with SLE and no kidney damage, 23 individuals with LN, and 10 clinically healthy controls (HCs) to identify differential metabolic profiles for SLE and LN. The data analysis revealed a differentially abundant metabolite expression profile for each study group, and those metabolites may act as potential differential biomarkers of SLE and LN. The differential metabolic pathways found between the LN and SLE patients with no kidney involvement included primary bile acid biosynthesis, branched-chain amino acid synthesis and degradation, pantothenate and coenzyme A biosynthesis, lysine degradation, and tryptophan metabolism. Receiver operating characteristic curve analysis revealed that monopalmitin, glycolic acid, and glutamic acid allowed for the differentiation of individuals with SLE and no kidney involvement and individuals with LN considering high confidence levels. While the results offer promise, it is important to recognize the significant influence of medications and other external factors on metabolomics studies. This impact has the potential to obscure differences in metabolic profiles, presenting a considerable challenge in the identification of disease biomarkers. Therefore, experimental validation should be conducted with a larger sample size to explore the diagnostic potential of the metabolites found as well as to examine how treatment and disease activity influence the identified chemical compounds. This will be crucial for refining the accuracy and effectiveness of using urine metabolomics for diagnosing and monitoring lupus and lupus nephritis.
Collapse
Affiliation(s)
- Alejandra Rojo-Sánchez
- Life Sciences Research Center, School of Basic and Biomedical Sciences, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Ada Carmona-Martes
- Life Sciences Research Center, School of Basic and Biomedical Sciences, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Yirys Díaz-Olmos
- Health Sciences Division, Medicine Program, Universidad del Norte, Barranquilla, Colombia
| | - Mary Santamaría-Torres
- Metabolomics Core Facility-MetCore, Vice-Presidency for Research, Universidad de los Andes, Bogotá, Colombia
| | - Mónica P Cala
- Metabolomics Core Facility-MetCore, Vice-Presidency for Research, Universidad de los Andes, Bogotá, Colombia
| | - Erick Orozco-Acosta
- Life Sciences Research Center, School of Basic and Biomedical Sciences, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Gustavo Aroca-Martínez
- Life Sciences Research Center, School of Basic and Biomedical Sciences, Universidad Simón Bolívar, Barranquilla, Colombia
- Clínica de la Costa, Barranquilla, Colombia
| | - Leonardo Pacheco-Londoño
- Life Sciences Research Center, School of Basic and Biomedical Sciences, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Elkin Navarro-Quiroz
- Life Sciences Research Center, School of Basic and Biomedical Sciences, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Lisandro A Pacheco-Lugo
- Life Sciences Research Center, School of Basic and Biomedical Sciences, Universidad Simón Bolívar, Barranquilla, Colombia.
| |
Collapse
|
8
|
Zhao H, Pan M, Liu Y, Cheng F, Shuai Z. Evaluation of early retinal changes in patients on long-term hydroxychloroquine using optical coherence tomography angiography. Ther Adv Drug Saf 2024; 15:20420986231225851. [PMID: 38300766 PMCID: PMC10823852 DOI: 10.1177/20420986231225851] [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: 01/07/2023] [Accepted: 12/21/2023] [Indexed: 02/03/2024] Open
Abstract
Background Connective tissue diseases (CTD), including systemic lupus erythematosus and rheumatoid arthritis (RA), have long been treated with hydroxychloroquine (HCQ). However, prolonged HCQ use poses a risk of adverse effects, particularly retinopathy. Objective To detect early retinal changes assessed by optical coherence tomography angiography (OCTA) in CTD patients with long-term HCQ treatment and to explore the relationship between OCTA parameters and the concentrations of HCQ and its metabolites. Design A cross-sectional study conducted from March 2020 to October 2021 at the First Affiliated Hospital of Anhui Medical University. Methods The area and perimeter of the foveal avascular zone (FAZ), the thickness of the fovea and parafovea, and the vascular density of the superficial capillary plexus (SCP) and deep capillary plexus (DCP) in each area of the macula were measured by OCTA in 43 CTD patients treated with HCQ for over 6 months. Meantime, blood concentrations of HCQ and its metabolites were determined by high-performance liquid chromatography-tandem mass spectrometry, and the clinical documents of all 43 involved patients were collected. Results There is no significant correlation between OCTA outcomes and the patient's age, disease duration, and weight-dependent dose. HCQ cumulative duration positively correlated with FAZ area and perimeter (r = 0.419, p = 0.005 and r = 0.407, p = 0.007, respectively) and negatively correlated with the foveal vessel density in DCP (r = -0.378, p = 0.012). HCQ cumulative dose had a positive correlation with FAZ area and perimeter (r = 0.445, p = 0.003 and r = 0.434, p = 0.004, respectively) and had a negative correlation with foveal vessel density in SCP and DCP (r = -0.383, p = 0.011 and r = -0.424, p = 0.005, respectively). OCTA outcomes did not correlate with HCQ and its metabolite concentrations. Conclusion OCTA could be used to detect microvascular changes in the macula of CTD patients with long-term HCQ therapy. It was not found the concentrations of HCQ and its metabolites were associated with retinal vascular changes.
Collapse
Affiliation(s)
- Huanhuan Zhao
- Department of Rheumatology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Public Health Clinical Center, Hefei, China
| | - Menglu Pan
- Department of Rheumatology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yaping Liu
- Department of Clinical Medicine, Anhui Medical University, Hefei, China
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Fangyue Cheng
- Department of Rheumatology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zongwen Shuai
- Department of Rheumatology, First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Hefei 230022, China
- Anhui Public Health Clinical Center, Hefei 230022, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei 230032, China
| |
Collapse
|
9
|
Tobin R, Patel N, Tobb K, Weber B, Mehta PK, Isiadinso I. Atherosclerosis in Systemic Lupus Erythematosus. Curr Atheroscler Rep 2023; 25:819-827. [PMID: 37768411 DOI: 10.1007/s11883-023-01149-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2023] [Indexed: 09/29/2023]
Abstract
PURPOSE OF THE REVIEW Systemic lupus erythematosus (SLE) patients are at increased risk of cardiovascular disease (CVD) compared to the general population, despite most patients being young females, who are not classically considered to be at high risk for cardiovascular disease using traditional risk assessment tools. The purpose of this review is to discuss the pathophysiology of atherosclerosis in SLE and raise awareness of the relationship between SLE and CVD. RECENT FINDINGS The increased risk of CVD in SLE patients is multifactorial, due to proatherogenic lipid profiles, immune dysregulation and inflammation, side effects of lupus treatment, and microvascular dysfunction. Conventional CV risk models often underperform in the identification of SLE patients at high risk of atherosclerosis. The use of non-invasive imaging serves as a strategy to identify patients with evidence of subclinical CVD and in the evaluation of symptomatic patients. Identification of subclinical atherosclerosis allows for aggressive management of CV risk factors. SLE patients experience an increased risk of atherosclerotic CVD, which is not solely explained by traditional CV risk factors. It is imperative that clinicians are aware of this association to implement prompt detection and treatment of atherosclerotic CVD in SLE patients.
Collapse
Affiliation(s)
- Rachel Tobin
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| | - Nidhi Patel
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Kardie Tobb
- Cone Health Medical Group, Greensboro, NC, USA
| | - Brittany Weber
- Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Puja K Mehta
- Division of Cardiology, Department of Medicine, Center for Heart Disease Prevention, Emory University School of Medicine, Atlanta, GA, USA
| | - Ijeoma Isiadinso
- Division of Cardiology, Department of Medicine, Center for Heart Disease Prevention, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
10
|
Tan Y, Yang S, Liu Q, Li Z, Mu R, Qiao J, Cui L. Pregnancy-related complications in systemic lupus erythematosus. J Autoimmun 2022; 132:102864. [PMID: 35872104 DOI: 10.1016/j.jaut.2022.102864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 07/02/2022] [Indexed: 10/17/2022]
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune inflammatory disease that predominantly affects women of childbearing age and results in various adverse pregnancy outcomes (APOs). Pregnancy was formerly discouraged in patients with SLE because of unstable disease activity during the gestation period, increased thrombosis risk, severe organ damage, and inevitable side effects of immunosuppressive agents. Currently, most patients with SLE have successful pregnancies due to preconception counselling, strict monitoring, and improved therapy with minimised complications for both the mother and foetus. Hydroxychloroquine (HCQ) is extensively used and is beneficial for improving pregnancy outcomes. However, pregnant women with SLE have a high-risk of APOs, such as disease flare, preterm birth, intrauterine growth restriction (IUGR), preeclampsia, and pregnancy loss. Better understanding of the changes in maternal immunity and serum biomarkers, as well as their relationships with SLE-related APOs progression, would facilitate the investigation of molecular mechanisms for triggering and ameliorating APOs. Furthermore, it would enable us to explore and develop novel and effective therapeutic strategies to prevent disease activation. Therefore, this review briefly introduces the interaction between pregnancy outcomes and SLE, elucidates pathophysiological and immunological changes during SLE pregnancy. Furthermore, this review systematically expounds on the effective predictors of APOs and the molecular mechanisms underlying the SLE-related APOs to provide a solid foundation for the advanced management of lupus pregnancy.
Collapse
Affiliation(s)
- Yuan Tan
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China; Core Unit of National Clinical Research Center for Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China; Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
| | - Shuo Yang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China; Core Unit of National Clinical Research Center for Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Qi Liu
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China; Core Unit of National Clinical Research Center for Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China; Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
| | - Zhongxin Li
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China; Core Unit of National Clinical Research Center for Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Rong Mu
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing, 100191, China.
| | - Jie Qiao
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Beijing, 100191, China; Ministry of Education Key Laboratory of Assisted Reproduction, Center for Reproductive Medicine, Beijing, 100191, China; Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China.
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China; Core Unit of National Clinical Research Center for Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
11
|
Wakiya R, Ueeda K, Shimada H, Nakashima S, Kameda T, Miyatake N, Kato M, Miyagi T, Sugihara K, Mizusaki M, Mino R, Kadowaki N, Dobashi H. Supplemental hydroxychloroquine therapy regulates adipokines in patients with systemic lupus erythematosus with stable disease. Clin Rheumatol 2022; 41:3345-3353. [PMID: 35849246 DOI: 10.1007/s10067-022-06282-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 06/19/2022] [Accepted: 06/28/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND In patients with systemic lupus erythematosus (SLE), a higher frequency of atherosclerotic lesions is associated with poor prognosis. Hydroxychloroquine (HCQ) has been reported to improve the lifespan and the prognosis of dyslipidaemia in patients with SLE, but the mechanism is unclear. We investigated the effect of supplemental HCQ treatment on the levels of serum cytokines associated with atherosclerosis in patients with stable SLE. METHODS Patients with SLE who received supplemental HCQ and maintained low disease activity between January 2016 and September 2020 were included in this study. Disease activity was assessed using Safety of Estrogens in Lupus National Assessment-SLE Disease Activity Index, Cutaneous Lupus Erythematous Disease Area and Severity Index, and Lupus Low Disease Activity State. Serum complement titres, anti-dsDNA antibodies, and serum cytokines (adiponectin, resistin, and leptin) were analyzed before and after HCQ treatment. RESULTS Forty-one patients (4 males and 37 females, mean age 41.3 ± 13.2 years) were included. Serum adiponectin levels were significantly increased after 3 months of HCQ treatment compared to baseline, and serum resistin levels were significantly reduced. The change in serum resistin level after HCQ administration was correlated with a significant reduction in serum TNF-α, interleukin (IL)-6, IL-8, and IL-1RA levels. CONCLUSIONS Supplemental HCQ treatment in patients with SLE improved adipokine levels. HCQ may improve prognosis by controlling disease activity in SLE and reducing risk factors for atherosclerosis. Key Points • Hydroxychloroquine has been reported to improve the prognosis of dyslipidaemia in patients with SLE, but the underlying mechanism is unclear. • In this study, hydroxychloroquine improved adipokine levels in patients with SLE, implicating adipokines as a potential mechanism underlying the benefit of hydroxychloroquine on dyslipidaemia. • Supplemental hydroxychloroquine should be considered in patients with SLE harboring lipid abnormalities and risk factors for atherosclerosis.
Collapse
Affiliation(s)
- Risa Wakiya
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan.
| | - Kiyo Ueeda
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Hiromi Shimada
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Shusaku Nakashima
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Tomohiro Kameda
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Nobuyuki Miyatake
- Department of Hygiene, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Mikiya Kato
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Taichi Miyagi
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Koichi Sugihara
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Mao Mizusaki
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Rina Mino
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Norimitsu Kadowaki
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Hiroaki Dobashi
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| |
Collapse
|
12
|
Connolly CM, Li J, Goldman D, Fava A, Magder L, Petri M. Lipoprotein(a) in systemic lupus erythematosus is associated with history of proteinuria and reduced renal function. Lupus 2022; 31:1367-1372. [PMID: 35775881 DOI: 10.1177/09612033221111958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Proteinuria is the clinical expression of lupus nephritis and despite recent advances in the therapeutic armamentarium for lupus nephritis, morbidity and mortality rates remain high. Therefore, the identification of factors that predict lupus nephritis is paramount in preventing damage accrual and disease progression. Lipoprotein (a) (Lp[a]) is a primarily genetically inherited plasma lipoprotein with pro-thrombotic and pro-atherosclerotic effects. Elevated Lp(a) has been observed at early stages of renal impairment in the general population and is associated with the development of chronic kidney disease. However, little is known about renal implications of Lp(a) in SLE. Thus, we evaluated Lp(a) and atherosclerotic events, thrombotic events, renal disease, and disease activity in patients with SLE. METHODS SLE patients fulfilling the revised American College of Rheumatology (ACR) or SLICC classification criteria with a measurement of Lp(a) were included in the analysis. A cutoff of 125 nmol/L was chosen based on expert opinion. Chi-square test was used to compare the differences between patient characteristics and Lp(a) levels. Logistic regression or linear regression were used, where appropriate, to assess the association between Lp(a) values and the measured outcomes. RESULTS Lp(a) levels from 562 patients were analyzed. There was an association between elevated Lp(a) and a history of proteinuria (OR 1.58, p-value = 0.02). This association remained significant following adjustment for age, sex, race, low C3, and elevated anti-dsDNA (OR = 1.55, p-value = 0.04). There was also an association with eGFR < 60 (p = 0.02). Patients with elevated Lp(a) had higher physician global activity (p = 0.01) and erythrocyte sediment rate (p = 0.03). CONCLUSION Elevated Lp(a) was associated with proteinuria, independent of known factors associated with lupus proteinuria, as well as reduced eGFR and physician global activity. Our findings highlight the potential role of Lp(a) as a noninvasive biomarker for early renal disease in SLE.
Collapse
Affiliation(s)
- Caoilfhionn M Connolly
- Department of Medicine, Division of Rheumatology, 1500Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jessica Li
- Department of Medicine, Division of Rheumatology, 1500Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel Goldman
- Department of Medicine, Division of Rheumatology, 1500Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrea Fava
- Department of Medicine, Division of Rheumatology, 1500Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Laurence Magder
- Department of Epidemiology and Public Health, Division of Biostatistics and Bioinformatics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Michelle Petri
- Department of Medicine, Division of Rheumatology, 1500Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
13
|
Papachristos DA, Gladman DD, Su J, Urowitz MB. Outcomes following antimalarial withdrawal in patients with quiescent systemic lupus erythematosus. Semin Arthritis Rheum 2022; 55:152046. [PMID: 35716465 DOI: 10.1016/j.semarthrit.2022.152046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 11/18/2022]
Abstract
OBJECTIVE Antimalarial medications (AMs) are central to the management of SLE. We investigated the rate of flare in lupus patients who withdrew AM after achieving clinical remission for at least one year, compared to those who continued therapy and compared flare rates in cases who tapered AM versus abruptly withdrew. METHODS Cases achieved clinical remission for at least one year then ceased their AM. Index date was defined as the date of complete AM cessation. Controls achieved clinical remission for at least one year and continued AM. Controls were matched according to the duration of AM before remission and the duration of therapy from remission date to case index date. All patients had ≥ 2 years of follow up. RESULTS Of 1573 patients on AM, 165 achieved a one-year remission and then ceased their AM. 96 had adequate follow-up, 88 were successfully matched to one control and 85 to 2 controls for a total of 173 controls. Flare occurred in 61.4% of cases and 45.1% of controls (p = 0.002). 52.3% patients who withdrew AM later restarted it due to disease flare. 88% recaptured control or improved, while 12% did not. Patients who tapered had significantly fewer flares (45.9% vs. 72.6%; p = 0.01). CONCLUSION AMs aid in preventing disease flare even in patients who have achieved prolonged clinical remission. Tapering AM results in lower flare rates. Hence, except in the setting of toxicity, cessation of antimalarial therapy in patients with prolonged disease quiescence is feasible using a slow taper.
Collapse
Affiliation(s)
- Danaë A Papachristos
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases University Health Network, Toronto Western Hospital, 399 Bathurst St. 1E-410B, Toronto, Ontario M5T 2S8, Canada
| | - Dafna D Gladman
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases University Health Network, Toronto Western Hospital, 399 Bathurst St. 1E-410B, Toronto, Ontario M5T 2S8, Canada; Schroeder Arthritis Institute, Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Jiandong Su
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases University Health Network, Toronto Western Hospital, 399 Bathurst St. 1E-410B, Toronto, Ontario M5T 2S8, Canada
| | - Murray B Urowitz
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases University Health Network, Toronto Western Hospital, 399 Bathurst St. 1E-410B, Toronto, Ontario M5T 2S8, Canada; Schroeder Arthritis Institute, Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
14
|
Lang MG, Vinagre CG, Bonfa E, Freitas FR, Pasoto SG, Brito TS, Seguro LP, Maranhão RC, Borba EF. Hydroxychloroquine increased cholesterol transfer to high-density lipoprotein in systemic lupus erythematosus: A possible mechanism for the reversal of atherosclerosis in the disease. Lupus 2022; 31:659-665. [PMID: 35332823 DOI: 10.1177/09612033221090127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION The beneficial effect of hydroxychloroquine (HCQ) in decreasing LDL levels on Systemic Lupus Erythematosus (SLE) is well defined. The influence of this drug on HDL levels is still under debate and information about its effect on cholesterol reverse transport is lacking. OBJECTIVE To evaluate the effects of HCQ on HDL levels and the transfer of lipids to this lipoprotein in SLE. METHODS Nineteen SLE patients using only HCQ (SLE WITH HCQ), 19 SLE patients without any therapy (SLE WITHOUT THERAPY), and 19 healthy controls (CONTROL) were included. All three groups were premenopausal women age- and gender-matched. Serum lipids and apolipoproteins were determined by commercial kits. An in vitro transfer of four lipids (14C-Phospolipid, 3H-Cholesteryl ester, 3H-Triglyceride, and 14C-Unesterified cholesterol) from a radioactively labeled nanoemulsion donor to HDL was performed in all participants. RESULTS Groups had comparable mean age, weight, height, BMI(body mass index), and waist circumference (p > .05). Mean HDL levels were higher in SLE WITH HCQ group compared to SLE WITHOUT THERAPY(58.37 ± 14.04 vs 49.79 ± 8.0 mg/dL; p < .05) but lower than CONTROL (58.37 ± 14.04 vs 68.58 ± 9.99 mg/dL; p < .05). Total cholesterol (TC) and LDL levels were also significantly lower in SLE WITH HCQ compared SLE WITHOUT THERAPY(148.16 ± 16.43 vs 167.11 ± 30.18 mg/dL; p < .05, 75.05 ± 22.52 vs 96.05 ± 25.63 mg/dL; p < .05) and CONTROL (148.16 ± 16.43 vs 174.11 ± 23.70 mg/dL; p < .05, 75.05 ± 22.52 vs 88.53 ± 20.24 mg/dL; p < .05). The in vitro lipid transfer to HDL study revealed a significant difference among the three groups (p = .002) with a higher transfer of unesterified cholesterol(UC) in SLE WITH HCQ compared to SLE WITHOUT THERAPY(5.40 ± 1.05% vs. 4.44 ± 1.05%; p < .05). The latter was significantly decreased compared to CONTROL (5.40 ± 1.05% vs. 5.99 ± 1.71%; p < .05).The percentages of transfer of triacylglycerol (4.93 ± 0.69% vs. 4.50 ± 0.69% vs. 5.14 ± 1.01%; p = .054), esterified cholesterol (5.24 ± 0.70% vs. 4.96 ± 0.89% vs. 5.69 ± 1.27%; p = .079), and phospholipid (15.67 ± 1.03% vs. 15.34 ± 1.44% vs. 16.47 ± 1.89%; p = .066) were similar among groups. CONCLUSION The present study is the first to demonstrate that HCQ promoted a higher transfer of unesterified cholesterol which may account for the increased HDL levels in lupus patients under HCQ. This desirable effect may underlie the reported reduced atherosclerosis in SLE.
Collapse
Affiliation(s)
- Maria G Lang
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, 117265Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Carmen Gc Vinagre
- Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, 117265Universidade de Sao Paulo, Sao Paulo, Brazil.,Universidade Santo Amaro (UNISA), Sao Paulo, Brazil
| | - Eloisa Bonfa
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, 117265Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Fatima R Freitas
- Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, 117265Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Sandra G Pasoto
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, 117265Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Tatiane S Brito
- Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, 117265Universidade de Sao Paulo, Sao Paulo, Brazil.,Universidade Santo Amaro (UNISA), Sao Paulo, Brazil
| | - Luciana Pc Seguro
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, 117265Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Raul C Maranhão
- Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, 117265Universidade de Sao Paulo, Sao Paulo, Brazil.,Faculdade de Ciencias Farmaceuticas, 117265Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Eduardo F Borba
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, 117265Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
15
|
Dima A, Jurcut C, Chasset F, Felten R, Arnaud L. Hydroxychloroquine in systemic lupus erythematosus: overview of current knowledge. Ther Adv Musculoskelet Dis 2022; 14:1759720X211073001. [PMID: 35186126 PMCID: PMC8848057 DOI: 10.1177/1759720x211073001] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 12/17/2021] [Indexed: 12/22/2022] Open
Abstract
The antimalarial hydroxychloroquine (HCQ) has demonstrated several crucial properties for the treatment of systemic lupus erythematosus (SLE). Herein, we reviewed the main HCQ pharmacologic features, detailed its mechanism of action, and summarized the existing guidelines and recommendations for HCQ use in rheumatology with a systematic literature search for the randomized controlled trials focused on lupus. HCQ has been shown to decrease SLE activity, especially in mild and moderate disease, to prevent disease flare and to lower the long-term glucocorticoid need. The numerous benefits of HCQ are extended to pregnancy and breastfeeding period. Based on cohort studies, antithrombotic and metabolic HCQ’s effects were shown, including lipid-lowering properties, which might contribute to an improved cardiovascular risk. Moreover, early HCQ use in antinuclear antibodies positive individuals might delay the progression to SLE. Finally, HCQ has a significant favorable impact on long-term outcomes such as damage accrual and mortality in SLE. Based on these multiple benefits, HCQ is now the mainstay long-term treatment in SLE, recommended by current guidelines in all patients unless contraindications or side effects. The daily dose associated with the best compromise between efficacy and safety is matter of debate. The concern regarding retinal toxicity rather than proper efficacy data is the one that dictated the daily dosage of ⩽5 mg/kg/day actual body weight currently agreed upon.
Collapse
Affiliation(s)
- Alina Dima
- Department of Rheumatology, Colentina Clinical Hospital, Bucharest, Romania
| | - Ciprian Jurcut
- Department of Internal Medicine, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest, Romania
| | - François Chasset
- Department of Dermatology and Allergology, Hôpital Tenon, Paris, France; Faculté de Médecine, Sorbonne Université, Paris, France
| | - Renaud Felten
- National Reference Center for Rare Auto-immune and Systemic Diseases Est Sud-Est (RESO), Strasbourg, France
- Department of Rheumatology, Les Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Laurent Arnaud
- National Reference Center for Rare Auto-immune and Systemic Diseases Est Sud-Est (RESO), Strasbourg, France
- Department of Rheumatology, Les Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- Université de Strasbourg, Inserm UMR-S 1109, Strasbourg, France
- Service de Rhumatologie, Hôpital de Hautepierre, 1, avenue Molière BP 83049, 67098 Strasbourg Cedex, France
| |
Collapse
|
16
|
Imoto AM, Gottems LB, Salomon AL, Silva HECE, Júnior IL, Peccin MS, Amorim FF, Santana LA. The impact of a low-calorie, low-glycemic diet on systemic lupus erythematosus: a systematic review. Adv Rheumatol 2021; 61:66. [PMID: 34742350 DOI: 10.1186/s42358-021-00224-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 10/17/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Diet plays a critical role in Systemic Lupus Erythematosus (SLE) patients, impacting on the microbiota composition and, consequently, on the immune response. The objective was to analyze and verify the diet effect on SLE patients. METHODS This is a systematic review performed at the Evidences-based Health Lab, Escola Superior em Ciências da Saúde, Brasília (DF), Brazil. In March, 2021, five databases, and grey literature, through JSTOR, Open Grey, and Google Scholar were searched. Randomized Clinical Trials in which SLE patients with calorie restricted, low glycemic index or other diet involving the joint adequacy of these aspects, compared with placebo or different types of diet, were included. RESULTS It was identified in the databases 758 articles; 132 were duplicated; 616 references were screened, and 604 were excluded. After reading the title and abstract, 12 articles were included for full-text reading. After the full-text reading, three studies were included for quantitative analysis. The diet improved the quality of life at 6 (MD 16.30; 5.91;26.69) and 12 weeks (MD 14.60; 0.88;28.32). The GRADE was used to evaluate the quality of evidence. CONCLUSION There is low evidence that the diet has a positive impact on the quality of life of SLE patients. Trial registration PROSPERO-CRD4202012208.
Collapse
Affiliation(s)
- Aline Mizusaki Imoto
- Evidences-Based Health Lab, Professional and Academic Master Program, Escola Superior em Ciências da Saúde, Brasília, DF, Brazil.
| | - Leila Bernarda Gottems
- Evidences-Based Health Lab, Professional and Academic Master Program, Escola Superior em Ciências da Saúde, Brasília, DF, Brazil
| | - Ana Lúcia Salomon
- Professional and Academic Master Program, Escola Superior em Ciências da Saúde, Brasília, DF, Brazil
| | | | - Império Lombardi Júnior
- Human Movement Sciences Department, Interdisciplinary Post-Graduation Program in Health Sciences, Universidade Federal de São Paulo (UNIFESP), Santos, SP, Brazil
| | - Maria Stella Peccin
- Human Movement Sciences Department, Interdisciplinary Post-Graduation Program in Health Sciences, Universidade Federal de São Paulo (UNIFESP), Santos, SP, Brazil
| | - Fábio Ferreira Amorim
- Medicine Graduation Course, Escola Superior em Ciências da Saúde, Brasília, DF, Brazil.,Family Health Master Program (ProfSaúde), Escola Superior em Ciências da Saúde, Brasília, DF, Brazil
| | - Levy Aniceto Santana
- Evidences-Based Health Lab, Professional and Academic Master Program, Escola Superior em Ciências da Saúde, Brasília, DF, Brazil
| |
Collapse
|
17
|
Borg A, Gomez A, Cederlund A, Cobar F, Qiu V, Lindblom J, Emamikia S, Enman Y, Pettersson S, Parodis I. Contribution of abnormal BMI to adverse health-related quality of life outcomes after a 52-week therapy in patients with SLE. Rheumatology (Oxford) 2021; 60:4205-4217. [PMID: 33404659 PMCID: PMC8410008 DOI: 10.1093/rheumatology/keaa909] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/30/2020] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVES To investigate whether abnormal BMI is associated with adverse health-related quality of life (HRQoL) outcome, including severe fatigue, after 52 weeks of standard therapy plus belimumab or placebo in patients with SLE. METHODS We analysed data from the BLISS-52 (NCT00424476) and BLISS-76 (NCT00410384) trials (n = 1684). Adverse HRQoL was defined as SF-36 scores ≤ the fifth percentile in age- and sex-matched US population-based subjects, and FACIT-F scores <30. We compared BMI groups using the Pearson's χ2 test, and assessed independence with multivariable logistic regression analysis. RESULTS Overweight (BMI ≥25 kg/m2) and obese (BMI ≥30 kg/m2) patients showed increased likelihood to exhibit adverse SF-36 physical component summary (OR: 1.8; 95% CI: 1.4, 2.3; P <0.001 and OR: 2.4; 95% CI: 1.8, 3.2; P <0.001, respectively) and FACIT-F (OR: 1.3; 95% CI: 1.1, 1.6; P = 0.010 and OR: 1.5; 95% CI: 1.2, 2.0; P = 0.002, respectively) scores at week 52. Underweight was associated with adverse SF-36 mental component summary scores, also after adjustment for sex, ancestry, age, disease duration, disease activity, organ damage and prednisone dose during the study period (OR: 2.1; 95% CI: 1.2, 3.6; P = 0.007). Addition of belimumab to standard therapy independently protected against adverse SF-36 general health (OR: 0.8; 95% CI: 0.6, 1.0; P = 0.025) and FACIT-F < 30 (OR: 0.8; 95% CI: 0.6, 1.0; P = 0.018). CONCLUSION Overweight and obesity contributed to adverse physical and mental HRQoL outcomes after therapeutic intervention in SLE patients, and underweight contributed to adverse mental HRQoL outcome. A protective effect of belimumab against adverse general health and severe fatigue was implicated.
Collapse
Affiliation(s)
- Alexander Borg
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital
| | - Alvaro Gomez
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital
| | - Arvid Cederlund
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital
| | - Flordelyn Cobar
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital
| | - Victor Qiu
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital
| | - Julius Lindblom
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital
| | - Sharzad Emamikia
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital
| | - Yvonne Enman
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital
| | - Susanne Pettersson
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital
- Division of Physiotherapy, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital
| |
Collapse
|
18
|
The impact of antimalarial agents on traditional and non-traditional subclinical atherosclerosis biomarkers in systemic lupus erythematosus: A systematic review and meta-analysis. Autoimmun Rev 2021; 20:102887. [PMID: 34237422 DOI: 10.1016/j.autrev.2021.102887] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Cardiovascular (CV) morbidity is a well-established problem in systemic lupus erythematosus (SLE). Antimalarial (AM) therapy has been seen as a potential atheroprotective agent. The aim was to assess the impact of AM therapy on traditional and novel atherosclerosis (AT) biomarkers in patients with SLE. METHODS A search of MEDLINE, EMbase, and Cochrane library for studies evaluating the impact of AM on AT biomarkers in SLE was conducted. Data extraction included serum, functional and structural traditional and novel biomarkers. A narrative synthesis of the findings and a meta-analysis with random effects was conducted estimating mean differences (MD), OR, HR and 95% CIs. RESULTS The search strategy produced 148 articles, of which 64 were extracted for analysis. The MD in VLDL-cholesterol (-10.29, 95% CI -15.35, 5.24), triglycerides (-15.68, 95% CI -27.51, -3.86), and diastolic BP (-3.42, 95% CI -5.62, -1.23) differed significantly in patients on AM therapy compared with those without AM therapy. Patients on AM had a lower prevalence and incidence of diabetes mellitus than patients not on AM (HR: 0.39, 95% CI 0.17, 0.88). HCQ use was associated with lower blood pressure (BP) variability. Structural markers like carotid intima-media thickness (IMT), carotid plaque (CP) and coronary artery calcification (CAC) were not influenced by AM. For functional markers like endothelial and arterial stiffness the benefit was unclear. The GRADE approach showed a very low-to-low quality of evidence (QoE) per outcome. CONCLUSIONS There is some evidence on the associations between AM therapy and some AT markers. However, the data on which this conclusion was based was of low to very low evidence.
Collapse
|
19
|
Understanding Accelerated Atherosclerosis in Systemic Lupus Erythematosus: Toward Better Treatment and Prevention. Inflammation 2021; 44:1663-1682. [PMID: 33821395 DOI: 10.1007/s10753-021-01455-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/01/2021] [Accepted: 03/18/2021] [Indexed: 12/13/2022]
Abstract
Systemic lupus erythematosus (SLE) carries a significant risk of cardiovascular disease (CVD). The prevalence of premature CVD is especially noteworthy because it occurs in premenopausal women with SLE who would otherwise have very low rates of CVD. While traditional risk factors likely play a role in development of CVD in the setting of SLE, they do not fully explain the excess risk. The pathogenesis of CVD in SLE is not fully understood, but the inflammatory nature of SLE is believed to be a key factor in accelerating atherosclerosis. Systemic inflammation may lead to an abnormal lipid profile with elevated triglycerides, total cholesterol, and low-density lipoprotein cholesterol and dysfunctional high-density lipoprotein cholesterol. Additionally, the inflammatory milieu of SLE plasma promotes endothelial dysfunction and vascular injury, early steps in the progression of atherosclerotic CVD. Despite the overall headway that has been achieved in treating lupus, innovative therapeutics specifically targeting the progression of atherosclerosis within the lupus population are currently lacking. However, there have been advancements in the development of promising modalities for diagnosis of subclinical atherosclerosis and detection of high CVD risk patients. Due to the significant impact of CVD on morbidity and mortality, research addressing prevention and treatment of CVD in SLE needs to be prioritized. This review explores the intricate interplay of SLE-specific properties that contribute to atherosclerosis and CVD within this population, as well as screening methods and possible therapies.
Collapse
|
20
|
Appleton BD, Major AS. The latest in systemic lupus erythematosus-accelerated atherosclerosis: related mechanisms inform assessment and therapy. Curr Opin Rheumatol 2021; 33:211-218. [PMID: 33394753 PMCID: PMC8049098 DOI: 10.1097/bor.0000000000000773] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Accelerated atherosclerosis is a significant comorbidity and the leading cause of death for patients with systemic lupus erythematosus (SLE). It is now apparent that SLE-accelerated atherosclerosis is not driven solely by traditional cardiovascular risk factors, adding complexity to disease characterization and mechanistic understanding. In this review, we will summarize new insights into SLE-accelerated atherosclerosis evaluation, treatment, and mechanism. RECENT FINDINGS Recent work highlights the need to incorporate inflammatory biomarkers into cardiovascular disease (CVD) risk assessments. This is especially true for SLE patients, in which mechanisms of immune dysfunction likely drive CVD progression. There is new evidence that commonly prescribed SLE therapeutics hinder atherosclerosis development. This effect is achieved both by reducing SLE-associated inflammation and by directly improving measures of atherosclerosis, emphasizing the interconnected mechanisms of the two conditions. SUMMARY SLE-accelerated atherosclerosis is most likely the consequence of chronic autoimmune inflammation. Therefore, diligent management of atherosclerosis requires assessment of SLE disease activity as well as traditional cardiovascular risk factors. This supports why many of the therapeutics classically used to control SLE also modulate atherosclerosis development. Greater understanding of the mechanisms underlying this condition will allow for the development of more targeted therapeutics and improved outcomes for SLE patients.
Collapse
Affiliation(s)
- Brenna D. Appleton
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Amy S. Major
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt Medical Center, Nashville, TN, 37232, USA
- Tennessee Valley Healthcare System, U.S. Department of Veterans Affairs, Nashville, TN, 37212, USA
| |
Collapse
|
21
|
Lima G, Treviño-Tello F, Atisha-Fregoso Y, Llorente L, Fragoso-Loyo H, Jakez-Ocampo J. Exhausted T cells in systemic lupus erythematosus patients in long-standing remission. Clin Exp Immunol 2021; 204:285-295. [PMID: 33475152 DOI: 10.1111/cei.13577] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/24/2022] Open
Abstract
The mechanisms that drive systemic lupus erythematosus (SLE) patients to achieve remission are unknown; one possible explanation might be T cell exhaustion. The aim of the present study was to measure CD4+ and CD8+ T cell exhaustion in SLE patients in prolonged remission (PR-SLE) and compared them with patients with active SLE (Act-SLE) and healthy subjects. We included 15 PR-SLE patients, 15 Act-SLE and 29 healthy subjects. T cell exhaustion was determined by flow cytometry according to the expression of programmed cell death 1 (PD)-1, T cell immunoglobulin and mucin 3 (Tim-3), natural killer cell receptor (2B4), eomesodermin (EOMES) and T-box transcription factor TBX21 (T-bet) in CD4+ and CD8+ T cells. Dimensionality reduction using the T-distributed stochastic neighbor-embedding algorithm and clustering analysis was used for the identification of relevant populations. Percentages of CD3+ , CD4+ and CD8+ T cells were similar among groups. We identified five subpopulations of CD8+ and seven of CD4+ cells. The CD4+ T-bet+ CD45RO+ cells identified in the unsupervised analysis were significantly increased in PR-SLE versus Act-SLE [median = 0·20, interquartile range (IQR) = 1·74-30·50 versus 1·68, IQR = 0·4-2·83; P < 0·01]. CD4+ EOMES+ cells were also increased in PR-SLE versus Act-SLE (5·24, IQR = 3·38-14·70 versus 1·39, IQR = 0·48-2·87; P < 0·001). CD8+ EOMES+ cells were increased in PR-SLE versus Act-SLE (37·6, IQR = 24·9-53·2 versus 8·13, IQR = 2·33-20·5; P < 0·001). Exhausted and activated T cells presented an increased frequency of PD-1, CD57 and EOMES in SLE patients versus healthy subjects. Some subpopulations of T cells expressing markers associated with exhaustion are increased in patients in remission, supporting T cell exhaustion as a tolerance mechanism in SLE. Exhaustion of specific populations of T cells might represent a potential therapeutic tool that will contribute to the goal of achieving sustained remission in these patients.
Collapse
Affiliation(s)
- G Lima
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - F Treviño-Tello
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Y Atisha-Fregoso
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, Manhasset, NY, USA.,Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - L Llorente
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - H Fragoso-Loyo
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - J Jakez-Ocampo
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
22
|
Dima A, Jurcut C, Arnaud L. Hydroxychloroquine in systemic and autoimmune diseases: Where are we now? Joint Bone Spine 2021; 88:105143. [PMID: 33515791 DOI: 10.1016/j.jbspin.2021.105143] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 12/23/2020] [Indexed: 12/28/2022]
Abstract
Hydroxychloroquine (HCQ), one of the oldest drugs used in rheumatology, came recently into attention as one of the potential therapies tested for the severe acute respiratory syndrome coronavirus-2 disease treatment. Used initially as an antimalarial, then translated to rheumatic diseases, HCQ has been used in a wide range of pathologies, including infectious diseases, immune disorders, diabetes, dyslipidemia, or neoplasia. Regarding systemic diseases, HCQ is the mainstay treatment for systemic lupus erythematosus (SLE), where, according to last European guidelines, it is proposed to all SLE patients unless contraindicated or with side effects. HCQ proved positive impact in SLE on robust outcomes, such as accrual damage, disease activity and survival, but also pleiomorphic effects, including decrease in the need for glucocorticoids, reduction in the risk of neonatal lupus, lower fasting glucose and protection against diabetes, thrombotic risk, dyslipidemia, infections, etc. Moreover, HCQ can be used during pregnancy and breast-feeding. Besides SLE, the role for HCQ in the anti-phospholipid syndrome and Sjögren's disease is still under debate. On the contrary, recent advances showed only limited interest for rheumatoid arthritis, especially due the lack of structural damage prevention. There are still no strong data to sustain the HCQ use in other systemic diseases. In this review, we summarised the utility and efficacy of HCQ in different clinical conditions relevant for rheumatology practice.
Collapse
Affiliation(s)
- Alina Dima
- Department of rheumatology, Colentina clinical hospital, 020125 Bucharest, Romania
| | - Ciprian Jurcut
- Department of internal medicine, Dr. Carol Davila Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Laurent Arnaud
- National reference centre for rare auto-immune and systemic diseases Est Sud-Est (RESO), 67000 Strasbourg, France; Department of rheumatology, hôpitaux universitaires de Strasbourg, 67098 Strasbourg, France; Université de Strasbourg, Inserm UMR-S 1109, 67000 Strasbourg, France.
| |
Collapse
|
23
|
GlycA measured by NMR spectroscopy is associated with disease activity and cardiovascular disease risk in chronic inflammatory diseases. Am J Prev Cardiol 2020; 4:100120. [PMID: 34327480 PMCID: PMC8315361 DOI: 10.1016/j.ajpc.2020.100120] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/26/2020] [Accepted: 11/03/2020] [Indexed: 02/08/2023] Open
Abstract
GlycA is a biomarker of systemic inflammation, quantifying both the protein concentrations and glycosylation states of several acute phase proteins. GlycA has been shown to be associated with both subclinical atherosclerosis and with cardiovascular disease (CVD). GlycA levels are higher in acute and chronic inflammation. During ongoing systemic inflammatory processes, GlycA specific acute phase reactants and proteins undergo circulating concentration and glycosylation pattern changes, and these alterations are reflected in the GlycA NMR signal. Additionally, levels associate with ongoing disease severity in individuals with rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and psoriasis thus capturing active inflammation. Furthermore, in these disease states, GlycA is associated with cardiovascular disease (CVD) independent of traditional risk factors including C-reactive protein (CRP). Finally, GlycA levels decrease with exercise, weight loss, and systemic anti-inflammatory agents. Therefore, GlycA appears to be a promising new composite biomarker of active systemic inflammation including assessing CVD risk in patients with inflammatory diseases. Patients with chronic inflammatory disorders are at increased risk for cardiovascular diseasenot captured by traditional risk factors. GlycA is a biomarker of acute phase reactants by NMR spectroscopy which captures disease activity in human inflammatory diseases. GlycA associates with cardiovascular disease and offers a tool to monitor primary disease activity and assess CVD risk in inflammatory diseases.
Collapse
|
24
|
Russell DA, Markiewicz M, Oates JC. Lupus serum induces inflammatory interaction with neutrophils in human glomerular endothelial cells. Lupus Sci Med 2020; 7:e000418. [PMID: 33037079 PMCID: PMC7549476 DOI: 10.1136/lupus-2020-000418] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/30/2020] [Accepted: 09/01/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVES SLE is associated with endothelial cell dysfunction (ECD). Understanding how ECD leads to neutrophil infiltration into glomeruli is essential to finding therapeutic targets for SLE. The aim of this study is to determine the effect of SLE serum from patients with active disease to induce neutrophil adhesion to and chemotaxis towards glomerular endothelial cells and factors induced by serum that associate with neutrophil chemotaxis. METHODS Patients with SLE had serum collected during paired longitudinal visits with lower and higher activity. 13 patients with SLE (5 SLE, 5 SLE with hypertension (HTN) and 3 SLE lupus nephritis (LN) and HTN), and 10 healthy controls (5 with and 5 without HTN) were examined. The adhesion of neutrophils to serum-treated human renal glomerular endothelial cells (HRGECs) or chemotaxis of neutrophils towards conditioned media from serum-treated HRGECs was determined, and levels of cytokines in this conditioned medium were quantified. Pathway analysis of cytokines induced by SLE and LN serum that associated with neutrophil migration was performed. RESULTS HRGECs treated with SLE serum induced significantly greater neutrophil chemotaxis and adhesion compared with control serum. When examining specific cohorts, SLE HTN and LN HTN promoted greater neutrophil chemotaxis than control serum, while SLE HTN and LN HTN promoted greater chemotaxis than SLE serum. Serum from active disease visits promoted neutrophil chemotaxis and adhesion over paired inactive visits. Levels of platelet-derived growth factor-BB, interleukin (IL)-15 and IL-8 secreted by SLE serum-treated HRGECs positively correlated with neutrophil chemotaxis. Pathway analysis suggested that LN serum induced pathways important in endoplasmic reticulum and oxidative stress. CONCLUSIONS SLE serum induces expression of mediators by HRGECs that promote neutrophil chemotaxis and adhesion, which increases during disease activity, and associates with factors common to pathways of endoplasmic reticulum and oxidative stress. These findings highlight the potential importance of serum factor-induced ECD in SLE and LN.
Collapse
Affiliation(s)
- Dayvia A Russell
- Medical Service, Ralph H Johnson VA Medical Center, Charleston, South Carolina, USA
| | - Margaret Markiewicz
- Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jim C Oates
- Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Medical Service, Ralph H Johnson VA Medical Center, Charleston, South Carolina, USA
| |
Collapse
|
25
|
Kostopoulou M, Nikolopoulos D, Parodis I, Bertsias G. Cardiovascular Disease in Systemic Lupus Erythematosus: Recent Data on Epidemiology, Risk Factors and Prevention. Curr Vasc Pharmacol 2020; 18:549-565. [DOI: 10.2174/1570161118666191227101636] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 11/20/2019] [Accepted: 12/12/2019] [Indexed: 12/22/2022]
Abstract
Systemic Lupus Erythematosus (SLE) is associated with increased risk for accelerated atherosclerosis
and cardiovascular (CV) events including coronary heart disease, cerebrovascular and peripheral
artery disease. CV events occur both early and late during the disease course, with younger
patients being at much higher risk than age-matched counterparts. The risk cannot be fully accounted for
by the increased prevalence of traditional atherosclerotic factors and may be due to pathophysiologic
intermediates such as type I interferons and other inflammatory cytokines, oxidative stress, activated
granulocytes and production of extracellular chromatin traps, antiphospholipid and other autoantibodies
causing dysfunction of lipoproteins, altogether resulting in endothelial injury and pro-atherogenic
dyslipidaemia. These mechanisms may be further aggravated by chronic intake of prednisone (even at
doses <7.5 mg/day), whereas immunomodulatory drugs, especially hydroxychloroquine, may exert antiatherogenic
properties. To date, there is a paucity of randomized studies regarding the effectiveness of
preventative strategies and pharmacological interventions specifically in patients with SLE. Nevertheless,
both the European League Against Rheumatism recommendations and extrapolated evidence from
the general population emphasize that SLE patients should undergo regular monitoring for atherosclerotic
risk factors and calculation of the 10-year CV risk. Risk stratification should include diseaserelated
factors and accordingly, general (lifestyle modifications/smoking cessation, antihypertensive and
statin treatment, low-dose aspirin in selected cases) and SLE-specific (control of disease activity, minimization
of glucocorticoids, use of hydroxychloroquine) preventive measures be applied as appropriate.
Further studies will be required regarding the use of non-invasive tools and biomarkers for CV assessment
and of risk-lowering strategies tailored to SLE.
Collapse
Affiliation(s)
- Myrto Kostopoulou
- 4th Department of Internal Medicine, Attikon University Hospital, Joint Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Dionysis Nikolopoulos
- 4th Department of Internal Medicine, Attikon University Hospital, Joint Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - George Bertsias
- Department of Rheumatology, Clinical Rheumatology and Allergy, University of Crete Medical School, Iraklio, Greece
| |
Collapse
|
26
|
Clinical characteristics of systemic lupus erythematosus patients in long-term remission without treatment. Clin Rheumatol 2020; 39:3365-3371. [PMID: 32870418 DOI: 10.1007/s10067-020-05379-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/25/2020] [Accepted: 08/27/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVE To describe the clinical and serological characteristics of patients with SLE who reached a state of sustained remission for more than 10 years in the absence of treatment. METHODS From a retrospective cohort of 2121 patients, 44 cases with sustained remission (PtRem) were identified and compared with 88 patients whose course has been chronically active (PtAct).The clinical and serological characteristics were analyzed, as well as the treatment of each group at the beginning of the disease and during its evolution. RESULTS Older age at disease onset was associated with a tendency to reach a state of prolonged remission. These patients also had a higher frequency of thrombocytopenia at the beginning of the disease 34.1% vs 10.2% (p < 0.001). PtAct had a significantly higher initial SLEDAI compared with cases (10.4 ± 5.6 vs 14.1 ± 5.8; p < 0.001). PtRem had a higher initial frequency of anti-β2 GP1 IgG antibodies. Also, 25% of these patients were serologically active. We did not find differences in the initial treatment between both groups. The accumulated damage measured by SLICC/ACR damage index at the end of the study was significantly less in the patients who remained in prolonged remission. CONCLUSIONS Although patients with SLE who achieve prolonged remission have some different characteristics at baseline compared with PtAct, it is not possible to identify a characteristic phenotype for the former. Achieving a state of prolonged remission should always be the goal in patients with SLE. Key Points • SLE patients can reach a very prolonged state of remission, free of treatment, including antimalarials, for at least 10 years. • Venous thromboembolism and thrombocytopenia are commonly present in patients that achieved remission. • The presence of serological markers of activity, even after 10 years in remission, is a risk factor for relapse.
Collapse
|
27
|
Zhang T, Mohan C. Caution in studying and interpreting the lupus metabolome. Arthritis Res Ther 2020; 22:172. [PMID: 32680552 PMCID: PMC7367412 DOI: 10.1186/s13075-020-02264-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 07/07/2020] [Indexed: 02/06/2023] Open
Abstract
Several metabolomics studies have shed substantial light on the pathophysiological pathways underlying multiple diseases including systemic lupus erythematosus (SLE). This review takes stock of our current understanding of this field. We compare, collate, and investigate the metabolites in SLE patients and healthy volunteers, as gleaned from published metabolomics studies on SLE. In the surveyed primary reports, serum or plasma samples from SLE patients and healthy controls were assayed using mass spectrometry or nuclear magnetic resonance spectroscopy, and metabolites differentiating SLE from controls were identified. Collectively, the circulating metabolome in SLE is characterized by reduced energy substrates from glycolysis, Krebs cycle, fatty acid β oxidation, and glucogenic and ketogenic amino acid metabolism; enhanced activity of the urea cycle; decreased long-chain fatty acids; increased medium-chain and free fatty acids; and augmented peroxidation and inflammation. However, these findings should be interpreted with caution because several of the same metabolic pathways are also significantly influenced by the medications commonly used in SLE patients, common co-morbidities, and other factors including smoking and diet. In particular, whereas the metabolic alterations relating to inflammation, oxidative stress, lipid peroxidation, and glutathione generation do not appear to be steroid-dependent, the other metabolic changes may in part be influenced by steroids. To conclude, metabolomics studies of SLE and other rheumatic diseases ought to factor in the potential contributions of confounders such as medications, co-morbidities, smoking, and diet.
Collapse
Affiliation(s)
- Ting Zhang
- Department of biomedical engineering, University of Houston, Houston, TX, 77204, USA
| | - Chandra Mohan
- Department of biomedical engineering, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
28
|
Dierckx T, Chiche L, Daniel L, Lauwerys B, Van Weyenbergh J, Jourde-Chiche N. Serum GlycA Level is Elevated in Active Systemic Lupus Erythematosus and Correlates to Disease Activity and Lupus Nephritis Severity. J Clin Med 2020; 9:jcm9040970. [PMID: 32244481 PMCID: PMC7230647 DOI: 10.3390/jcm9040970] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/27/2020] [Accepted: 03/28/2020] [Indexed: 12/16/2022] Open
Abstract
Objective: Reliable non-invasive biomarkers are needed to assess disease activity and prognosis in patients with systemic lupus erythematosus (SLE). Glycoprotein acetylation (GlycA), a novel biomarker for chronic inflammation, has been reported to be increased in several inflammatory diseases. We investigated the relevance of serum GlycA in SLE patients exhibiting various levels of activity and severity, especially with regards to renal involvement. Methods: Serum GlycA was measured by nuclear magnetic resonance spectroscopy in samples from well characterized SLE patients and from both healthy controls and patients with other kidney diseases (KD). Disease activity was evaluated using the Systemic Lupus Erythematosus Activity Index 2000 (SLEDAI-2K). Renal severity was assessed by kidney biopsy. Results: Serum GlycA was elevated in active (n = 105) compared to quiescent SLE patients (n = 39, p < 10−6), healthy controls (n = 20, p = 0.009) and KD controls (n = 21, p = 0.04), despite a more severely altered renal function in the latter. GlycA level was correlated to disease activity (SLEDAI-2K, ρ = 0.37, p < 10−4), C-reactive protein, neutrophil count, triglyceride levels, proteinuria and inversely to serum albumin. In patients with biopsy-proven lupus nephritis (LN), GlycA levels were higher in proliferative (n = 26) than non-proliferative LN (n = 10) in univariate analysis (p = 0.04), and was shown to predict proliferative LN independently of renal parameters, immunological activity, neutrophil count and daily corticosteroid dosage by multivariate analysis (p < 5 × 10−3 for all models). In LN patients with repeated longitudinal GlycA measurement (n = 11), GlycA varied over time and seemed to peak at the time of the flare. Conclusions: GlycA, as a summary measure for different inflammatory processes, could be a valuable biomarker of disease activity in patients with SLE, and a non-invasive biomarker of pathological severity in the context of LN.
Collapse
Affiliation(s)
- Tim Dierckx
- Rega Institute, Laboratory of Clinical and Epidemiological Virology, Department of Microbiology and Immunology, KU Leuven, 3000 Leuven, Belgium; (T.D.); (J.V.W.)
| | - Laurent Chiche
- Médecine Interne, Hôpital Européen, 13003 Marseille, France;
| | - Laurent Daniel
- Hôpital de la Timone, Marseille, Laboratoire d’Anatomie Pathologique, AP-HM, 13005 Marseille, France;
- C2VN, INRA 1260, INSERM 1263, Aix-Marseille Université, 13005 Marseille, France
| | - Bernard Lauwerys
- Institut de Recherches Expérimentales et Cliniques, Université catholique de Louvain, 1200 Brussels, Belgium;
- Department of Rheumatology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Johan Van Weyenbergh
- Rega Institute, Laboratory of Clinical and Epidemiological Virology, Department of Microbiology and Immunology, KU Leuven, 3000 Leuven, Belgium; (T.D.); (J.V.W.)
| | - Noémie Jourde-Chiche
- C2VN, INRA 1260, INSERM 1263, Aix-Marseille Université, 13005 Marseille, France
- Hôpital de la Conception, Centre de Néphrologie et Transplantation Rénale, AP-HM, 13005 Marseille, France
- Correspondence: ; Tel.: +33-4-9138-3042; Fax: +33-4-9138-3206
| |
Collapse
|
29
|
Fuertes-Martín R, Correig X, Vallvé JC, Amigó N. Title: Human Serum/Plasma Glycoprotein Analysis by 1H-NMR, an Emerging Method of Inflammatory Assessment. J Clin Med 2020; 9:E354. [PMID: 32012794 PMCID: PMC7073769 DOI: 10.3390/jcm9020354] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/13/2020] [Accepted: 01/17/2020] [Indexed: 12/17/2022] Open
Abstract
Several studies suggest that variations in the concentration of plasma glycoproteins can influence cellular changes in a large number of diseases. In recent years, proton nuclear magnetic resonance (1H-NMR) has played a major role as an analytical tool for serum and plasma samples. In recent years, there is an increasing interest in the characterization of glycoproteins through 1H-NMR in order to search for reliable and robust biomarkers of disease. The objective of this review was to examine the existing studies in the literature related to the study of glycoproteins from an analytical and clinical point of view. There are currently several techniques to characterize circulating glycoproteins in serum or plasma, but in this review, we focus on 1H-NMR due to its great robustness and recent interest in its translation to the clinical setting. In fact, there is already a marker in H-NMR representing the acetyl groups of the glycoproteins, GlycA, which has been increasingly studied in clinical studies. A broad search of the literature was performed showing a general consensus that GlycA is a robust marker of systemic inflammation. The results also suggested that GlycA better captures systemic inflammation even more than C-reactive protein (CRP), a widely used classical inflammatory marker. The applications reviewed here demonstrated that GlycA was potentially a key biomarker in a wide range of diseases such as cancer, metabolic diseases, cardiovascular risk, and chronic inflammatory diseases among others. The profiling of glycoproteins through 1H-NMR launches an encouraging new paradigm for its future incorporation in clinical diagnosis.
Collapse
Affiliation(s)
- Rocío Fuertes-Martín
- Biosfer Teslab SL, 43201 Reus, Spain; (R.F.-M.); (N.A.)
- Metabolomic s platform, IISPV, CIBERDEM, Rovira i Virgili University, 43007 Tarragona, Spain
| | - Xavier Correig
- Metabolomic s platform, IISPV, CIBERDEM, Rovira i Virgili University, 43007 Tarragona, Spain
| | - Joan-Carles Vallvé
- Metabolomic s platform, IISPV, CIBERDEM, Rovira i Virgili University, 43007 Tarragona, Spain
- Lipids and Arteriosclerosis Research Unit, Sant Joan de Reus University Hospital, 43201 Reus, Spain
| | - Núria Amigó
- Biosfer Teslab SL, 43201 Reus, Spain; (R.F.-M.); (N.A.)
- Metabolomic s platform, IISPV, CIBERDEM, Rovira i Virgili University, 43007 Tarragona, Spain
| |
Collapse
|
30
|
Petri M, Elkhalifa M, Li J, Magder LS, Goldman DW. Hydroxychloroquine Blood Levels Predict Hydroxychloroquine Retinopathy. Arthritis Rheumatol 2020; 72:448-453. [PMID: 31532077 DOI: 10.1002/art.41121] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 09/12/2019] [Indexed: 01/03/2023]
Abstract
OBJECTIVE In 2016, the American Academy of Ophthalmology (AAO) changed the recommended daily dose of hydroxychloroquine (HCQ) from 6.5 mg/kg to <5 mg/kg. However, it is not clear that the lower prescribed dose of HCQ will have the same efficacy for systemic lupus erythematosus (SLE) activity or the same role in protecting against cardiovascular risk factors and thrombosis. This study was undertaken to address the frequency of HCQ retinopathy and the role of HCQ blood levels in identifying those individuals who are at a greater future risk of retinopathy. METHODS HCQ blood levels in 537 patients with SLE from a large clinical cohort were repeatedly measured, and patients were tested for HCQ retinopathy. We assessed the risk of retinopathy according to clinical characteristics and blood levels of HCQ. RESULTS The overall frequency of retinopathy was 4.3% (23 of 537 patients). There was a 1% risk of retinopathy in the first 5 years of HCQ treatment, 1.8% from 6 to 10 years, 3.3% from 11 to 15 years, 11.5% from 16 to 20 years, and 8.0% after 21 years of use. We found that older age (P < 0.0001), higher body mass index (P for trend = 0.0160), and longer duration of HCQ intake (P = 0.0024 and P for trend = 0.0006) were associated with a higher risk of HCQ toxicity. Higher blood levels of HCQ predicted later HCQ retinopathy (P = 0.0124 and P = 0.0340 for mean and maximum HCQ blood levels, respectively). CONCLUSION Our data prove the utility of assessing blood levels of HCQ in the prediction of retinopathy. This would allow clinicians to either decrease the dose or increase monitoring in those patients with high HCQ blood levels.
Collapse
Affiliation(s)
- Michelle Petri
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Jessica Li
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Daniel W Goldman
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
31
|
Kim SY, Yu M, Morin EE, Kang J, Kaplan MJ, Schwendeman A. High-Density Lipoprotein in Lupus: Disease Biomarkers and Potential Therapeutic Strategy. Arthritis Rheumatol 2020; 72:20-30. [PMID: 31350818 PMCID: PMC6935404 DOI: 10.1002/art.41059] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/23/2019] [Indexed: 12/31/2022]
Abstract
Systemic lupus erythematosus (SLE) patients exhibit accelerated development of atherosclerosis and increased incidents of cardiovascular disease (CVD) that cannot be explained by traditional risk factors alone. Accumulating evidence suggests that reduced levels of high-density lipoproteins (HDLs), along with altered HDL composition and function, may contribute to the accelerated atherosclerosis in SLE patients. Normally, HDLs play various atheroprotective roles through facilitating cholesterol efflux, inhibiting vascular inflammation, and scavenging oxidative species. However, systemic inflammation, oxidative stress, and autoimmunity in SLE patients induce changes in HDL size distribution and proteomic and lipidomic signatures. These compositional changes in HDLs result in the formation of proinflammatory, dysfunctional HDL. These lupus-altered HDLs have impaired antiatherogenic function with reduced cholesterol efflux capacities, impaired antioxidation abilities, and diminished antiinflammatory properties. In fact, dysfunctional HDL may promote atherogenesis by inducing inflammation. Thus, dysfunctional HDLs could be an important biomarker of accelerated atherosclerosis in lupus. Additionally, HDL-targeted therapies, especially infusion of reconstituted HDLs, may serve as a potential therapeutic intervention for SLE patients with CVD.
Collapse
Affiliation(s)
- Sang Yeop Kim
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Minzhi Yu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Emily E. Morin
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jukyung Kang
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mariana J. Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
32
|
Smith EMD, Lythgoe H, Midgley A, Beresford MW, Hedrich CM. Juvenile-onset systemic lupus erythematosus: Update on clinical presentation, pathophysiology and treatment options. Clin Immunol 2019; 209:108274. [PMID: 31678365 DOI: 10.1016/j.clim.2019.108274] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/12/2019] [Accepted: 10/13/2019] [Indexed: 12/25/2022]
Abstract
Juvenile-onset systemic lupus erythematosus (jSLE) accounts for up to 20% of all SLE patients. Key differences between juvenile- and adult-onset (aSLE) disease include higher disease activity, earlier development of damage, and increased use of immunosuppressive treatment in jSLE suggesting (at least partial) infectivity secondary to variable pathomechanisms. While the exact pathophysiology of jSLE remains unclear, genetic factors, immune complex deposition, complement activation, hormonal factors and immune cell dysregulation are involved to variable extents, promising future patient stratification based on immune phenotypes. Though less effective and potentially toxic, jSLE patients are treated based upon evidence from studies in aSLE cohorts. Here, age-specific clinical features of jSLE, underlying pathomechanisms, treatment options and disease outcomes will be addressed. Future directions to improve the care of jSLE patients, including implementation of the Single Hub and Access point for pediatric Rheumatology in Europe (SHARE) recommendations, biomarkers, treat to target and personalized medicine approaches are discussed.
Collapse
Affiliation(s)
- Eve Mary Dorothy Smith
- Department of Women's & Children's Health, Institution of Translational Medicine, University of Liverpool, UK; Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust, Eaton Rd, Liverpool L12 2AP, UK.
| | - Hanna Lythgoe
- Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust, Eaton Rd, Liverpool L12 2AP, UK
| | - Angela Midgley
- Department of Women's & Children's Health, Institution of Translational Medicine, University of Liverpool, UK
| | - Michael William Beresford
- Department of Women's & Children's Health, Institution of Translational Medicine, University of Liverpool, UK; Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust, Eaton Rd, Liverpool L12 2AP, UK
| | - Christian Michael Hedrich
- Department of Women's & Children's Health, Institution of Translational Medicine, University of Liverpool, UK; Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust, Eaton Rd, Liverpool L12 2AP, UK.
| |
Collapse
|
33
|
Mackey RH, Kuller LH, Moreland LW. Inflammatory joint diseases and atherosclerosis: time to look beyond the 'lipid paradox'. Curr Opin Lipidol 2019; 30:342-349. [PMID: 31145122 DOI: 10.1097/mol.0000000000000620] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Uncertainty persists about the contribution of lipids to the increased risk of cardiovascular disease (CVD) among rheumatoid arthritis and other inflammatory joint disease (IJD) patients. In reviewing recent research, we consider potential insights gained by quantifying lipoprotein particles directly, rather than by their lipid content. RECENT FINDINGS Although inflammation often decreases LDL cholesterol (LDL-C), and anti-inflammatory medications often increase LDL-C, both inflammation and anti-inflammatory medications can increase atherogenic Apolipoprotein B (ApoB)-containing lipoprotein particles, attenuated by statins. CVD risk factors, that is, smoking, obesity, ApoB, may increase years prior to IJD diagnosis. Increased risks of nonatherosclerotic myocardial and pulmonary disease, heart failure and mortality may be directly related to disease activity, inflammation, and possibly to HDL particles and function. SUMMARY For IJD patients, higher cumulative lifetime exposure to CVD risk factors accelerates atherosclerosis and subsequent CVD risk that is underestimated by current risk factor levels. CVD risk reduction in IJD requires aggressive and earlier reduction in CVD risk factors (ApoB lipoproteins, smoking, hypertension, diabetes, lack of physical activity), in addition to control of disease activity and inflammation. Lipid-lowering medications can attenuate anti-inflammatory medication-induced increases in ApoB and LDL-C, but can also reduce CVD risk due to cumulative lifetime exposure.
Collapse
Affiliation(s)
- Rachel H Mackey
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, University of Pittsburgh
| | - Lewis H Kuller
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, University of Pittsburgh
| | - Larry W Moreland
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW The mechanisms leading to the development of premature atherosclerosis and vascular injury in systemic lupus erythematosus (SLE) remain to be fully elucidated. This is a comprehensive review of recent research developments related to the understanding of cardiovascular disease (CVD) in lupus. RECENT FINDINGS SLE patients with lupus nephritis display significantly increased risk of myocardial infarction and CVD mortality than SLE patients without lupus nephritis. SLE disease-related parameters could be taken into consideration when calculating CVD risks. The type I interferon pathway is detrimental to the vasculature and may contribute to the development of insulin resistance. The level of low-density granulocytes, a distinct subset of proinflammatory neutrophils present in SLE, was independently associated with coronary plaque burden and endothelial dysfunction. Invariant natural killer T cells may promote an atheroprotective effect in SLE patients with asymptomatic atherosclerotic plaques. Oxidized lupus high-density lipoprotein promotes proinflammatory responses in macrophages. SUMMARY Recent discoveries have further strengthened the critical role of SLE-related immune dysregulation and metabolic disturbances in promoting accelerated CVD. Understanding how these pathogenic factors promote vascular injury may provide better molecular candidates for therapeutic targeting, and ultimately to improve CVD outcomes.
Collapse
|
35
|
Purmalek MM, Carlucci PM, Dey AK, Sampson M, Temesgen-Oyelakin Y, Sakhardande S, Lerman JB, Fike A, Davis M, Chung JH, Salahuddin T, Manna Z, Gupta S, Chen MY, Hasni S, Mehta NN, Remaley A, Kaplan MJ. Association of lipoprotein subfractions and glycoprotein acetylation with coronary plaque burden in SLE. Lupus Sci Med 2019; 6:e000332. [PMID: 31413851 PMCID: PMC6667837 DOI: 10.1136/lupus-2019-000332] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/09/2019] [Accepted: 06/18/2019] [Indexed: 11/05/2022]
Abstract
Objective Subjects with SLE display an enhanced risk of atherosclerotic cardiovascular disease (CVD) that is not explained by Framingham risk. This study sought to investigate the utility of nuclear MR (NMR) spectroscopy measurements of serum lipoprotein particle counts and size and glycoprotein acetylation (GlycA) burden to predict coronary atherosclerosis in SLE. Methods Coronary plaque burden was assessed in SLE subjects and healthy controls using coronary CT angiography. Lipoproteins and GlycA were quantified by NMR spectroscopy. Results SLE subjects displayed statistically significant decreases in high-density lipoprotein (HDL) particle counts and increased very low-density lipoprotein (VLDL) particle counts compared with controls. Non-calcified coronary plaque burden (NCB) negatively associated with HDL subsets whereas it positively associated with VLDL particle counts in multivariate adjusted models. GlycA was significantly increased in SLE sera compared with controls. In contrast to high-sensitivity C reactive protein, elevations in GlycA in SLE significantly associated with NCB and insulin resistance (IR), though the association with NCB was no longer significant after adjusting for prednisone use. Conclusions Patients with SLE display a proatherogenic lipoprotein profile that may significantly contribute to the development of premature CVD. The results demonstrate that NMR measures of GlycA and lipoprotein profiles, beyond what is captured in routine clinical labs, could be a useful tool in assessing CVD risk in patients with SLE.
Collapse
Affiliation(s)
- Monica M Purmalek
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Philip M Carlucci
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Amit K Dey
- National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland, USA
| | - Maureen Sampson
- National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Yenealem Temesgen-Oyelakin
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Simantini Sakhardande
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Joseph B Lerman
- National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland, USA
| | - Alice Fike
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Michael Davis
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Jonathan H Chung
- National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland, USA
| | - Taufiq Salahuddin
- National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland, USA
| | - Zerai Manna
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Sarthak Gupta
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Marcus Y Chen
- National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland, USA
| | - Sarfaraz Hasni
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Nehal N Mehta
- National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland, USA
| | - Alan Remaley
- National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland, USA
| | - Mariana J Kaplan
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
36
|
McMahon M, Skaggs B, Grossman J, Wong WK, Sahakian L, Chen W, Hahn B. Comparison of PREDICTS atherosclerosis biomarker changes after initiation of new treatments in patients with SLE. Lupus Sci Med 2019; 6:e000321. [PMID: 31321062 PMCID: PMC6606066 DOI: 10.1136/lupus-2019-000321] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 05/21/2019] [Accepted: 05/29/2019] [Indexed: 12/21/2022]
Abstract
Objective Patients with SLE have an increased risk of atherosclerosis (ATH) that is not adequately explained by traditional risk factors. We previously described the Predictors of Risk for Elevated Flares, Damage Progression, and Increased Cardiovascular disease in PaTients with SLE (PREDICTS) atherosclerosis-risk panel, which includes proinflammatory HDL (piHDL), leptin, soluble tumour necrosis factor-like weak inducer of apoptosis (sTWEAK) and homocysteine, as well as age and diabetes. A high PREDICTS score confers 28-fold increased odds for future atherosclerosis in SLE. The aim of this study is to determine whether PREDICTS biomarkers are modifiable by common lupus therapies. Methods This prospective observational study included SLE subjects started on new lupus treatments. Leptin, sTWEAK, homocysteine and antioxidant function of HDL were measured at baseline (prior to drug initiation), 6 weeks and 12 weeks. Results 16 subjects started mycophenolate (MMF), 18 azathioprine (AZA) and 25 hydroxychloroquine (HCQ). In MMF-treated subjects, HDL function progressively improved from 2.23 ± 1.32 at baseline to 1.37±0.81 at 6 weeks (p=0.02) and 0.93±0.54 at 12 weeks (p=0.009). sTWEAK levels also improved in MMF-treated subjects from 477.5±447.1 to 290.3±204.6 pg/mL after 12 weeks (p=0.04), but leptin and homocysteine levels were not significantly changed. In HCQ-treated subjects, only HDL function improved from 1.80±1.29 at baseline to 1.03±0.74 after 12 weeks (p=0.05). There were no changes in the AZA group. MMF treatment was still associated with significant improvements in HDL function after accounting for potential confounders such as total prednisone dose and changes in disease activity. Overall, the mean number of high-risk PREDICTS biomarkers at week 12 significantly decreased in the entire group of patients started on a new lupus therapy (2.1±0.9 to 1.8±0.9, p=0.02) and in the MMF-treated group (2.4±0.8 vs 1.8±0.9, p=0.003), but not in the AZA or HCQ groups. In multivariate analysis, the odds of having a high PREDICTS atherosclerosis risk score at 12 weeks were lower with MMF treatment (OR 0.002, 95% CI 0.000 to 0.55, p=0.03). Conclusions 12 weeks of MMF therapy improves the overall PREDICTS atherosclerosis biomarker profile. Further studies will determine whether biomarker changes reflect decreases in future cardiovascular events.
Collapse
Affiliation(s)
- Maureen McMahon
- Internal Medicine, Division Rheumatology, University of California, Los Angeles, Los Angeles, California, USA
| | - Brian Skaggs
- Internal Medicine, Division Rheumatology, University of California, Los Angeles, Los Angeles, California, USA
| | - Jennifer Grossman
- Internal Medicine, Division Rheumatology, University of California, Los Angeles, Los Angeles, California, USA
| | - Weng Kee Wong
- Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California, USA
| | - Lori Sahakian
- Internal Medicine, Division Rheumatology, University of California, Los Angeles, Los Angeles, California, USA
| | - Weiling Chen
- Internal Medicine, Division Rheumatology, University of California, Los Angeles, Los Angeles, California, USA
| | - Bevra Hahn
- Internal Medicine, Division Rheumatology, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
37
|
Fanouriakis A, Bertsias G. Changing paradigms in the treatment of systemic lupus erythematosus. Lupus Sci Med 2019; 6:e000310. [PMID: 31168398 PMCID: PMC6519431 DOI: 10.1136/lupus-2018-000310] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/09/2019] [Accepted: 01/13/2019] [Indexed: 12/12/2022]
Abstract
SLE poses formidable therapeutic challenges due to its heterogeneity and treatment decisions often cannot be guided by data of high quality. In this review, we attempt to provide insights regarding the treatment of SLE in everyday clinical practice, based on contemporary evidence and our own personal experience. We focus on common therapeutic issues and dilemmas arising in routine care, including monitoring for retinal toxicity associated with hydroxychloroquine, handling of glucocorticoid regimens in order to minimise their adverse events, choice of immunosuppressive medications based on prevailing disease manifestations and optimal use of available biological agents (belimumab and rituximab). We also provide our view on the position of calcineurin inhibitors in the management of lupus nephritis and conclude with remarks on the future perspectives for this challenging disease.
Collapse
Affiliation(s)
- Antonis Fanouriakis
- Rheumatology and Clinical Immunology Unit, 'Attikon' University Hospital, Athens, Greece
| | - George Bertsias
- Rheumatology, Clinical Immunology and Allergy, University Hospital of Heraklion, Heraklion, Greece
| |
Collapse
|
38
|
Worth C, Yusuf IH, Turner B, Gourier H, Brooks EE, Mort DO, Sharma S, Downes SM, Luqmani RA. An audit of the use of hydroxychloroquine in rheumatology clinics. Rheumatol Adv Pract 2018; 2:rky013. [PMID: 31431961 PMCID: PMC6649984 DOI: 10.1093/rap/rky013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 03/25/2018] [Indexed: 12/23/2022] Open
Abstract
Objective The aim was to audit the use, indications, complications and patient information regarding HCQ treatment in rheumatology clinics in a tertiary referral centre. Methods During a 9-month period, we identified all patients prescribed HCQ and attending rheumatology clinics in one hospital. We established: (i) the indication for HCQ; (ii) the prevalence of HCQ overdosing based on absolute body weight (ABW); (iii) documentation of warning of risk of retinal toxicity; (iv) systemic and ocular co-morbidities; (v) ocular symptoms during treatment; and (vi) reasons for stopping HCQ. Results We identified 427 patients (104 male and 323 female). The cumulative dose of HCQ was lower in RA (median 365 g; range 6–1752 g) compared with SLE (450 g; 66–1788 g) (P = 0.105). The median duration of HCQ therapy was 4 years (range 0.1–13 years); 28% of patients with RA and 29% with SLE continued HCQ beyond 5 years. After adjusting for ABW and renal function, 10% (31/312) had been prescribed doses exceeding recommendations. Formal documentation of counselling on ocular complications was found in only one-third of patients. Three cases of HCQ retinopathy were identified (all of whom had RA). Conclusion HCQ therapy is being used for >5 years in 29% of patients with rheumatic diseases, with higher than recommended doses in ∼10% of patients. We recommend more rigorous scrutiny of the use of HCQ to reduce the risk of retinopathy.
Collapse
Affiliation(s)
- Claudia Worth
- Department of Rheumatology, Oxford University Hospitals, Oxford
| | - Imran H Yusuf
- Oxford Eye Hospital, Oxford University Hospitals, Oxford
| | - Bethany Turner
- Department of Rheumatology, Oxford University Hospitals, Oxford
| | - Hanae Gourier
- Department of Emergency Medicine, Stoke Mandeville Hospital, Aylesbury
| | - Emma E Brooks
- School of Medicine, Rheumatology and Musculoskeletal Science, University of Oxford, Oxford, UK
| | - Daniel O Mort
- School of Medicine, Rheumatology and Musculoskeletal Science, University of Oxford, Oxford, UK
| | | | - Susan M Downes
- Oxford Eye Hospital, Oxford University Hospitals, Oxford
| | - Raashid A Luqmani
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Science, University of Oxford, Oxford, UK
| |
Collapse
|
39
|
|
40
|
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
|
41
|
Connelly MA, Otvos JD, Shalaurova I, Playford MP, Mehta NN. GlycA, a novel biomarker of systemic inflammation and cardiovascular disease risk. J Transl Med 2017; 15:219. [PMID: 29078787 PMCID: PMC5658936 DOI: 10.1186/s12967-017-1321-6] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 10/17/2017] [Indexed: 12/12/2022] Open
Abstract
Background GlycA is a novel spectroscopic marker of systemic inflammation with low intra-individual variability and other attributes favoring its clinical use in patients with chronic inflammatory and autoimmune diseases. GlycA is unique in its composite nature, reflecting both increased glycan complexity and circulating acute phase protein levels during local and systemic inflammation. Recent studies of GlycA from cross-sectional, observational and interventional studies have been highly informative, demonstrating that GlycA is elevated in acute and chronic inflammation, predicts death in healthy individuals and is associated with disease severity in patients with chronic inflammatory diseases such as rheumatoid arthritis, psoriasis and lupus. Moreover, following treatment with biological therapy in psoriasis, reduction in skin disease severity was accompanied by a decrease in GlycA levels and improvement in vascular inflammation. Conclusions Collectively, these findings suggest GlycA is a marker that tracks systemic inflammation and subclinical vascular inflammation. However, larger prospective studies and randomized trials are necessary in order to assess the impact of novel therapies on GlycA in patients with chronic inflammatory conditions, which may be concomitant with cardiovascular benefits.
Collapse
Affiliation(s)
- Margery A Connelly
- Laboratory Corporation of America Holdings (LabCorp), Morrisville, NC, 27560, USA
| | - James D Otvos
- Laboratory Corporation of America Holdings (LabCorp), Morrisville, NC, 27560, USA
| | - Irina Shalaurova
- Laboratory Corporation of America Holdings (LabCorp), Morrisville, NC, 27560, USA
| | - Martin P Playford
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Nehal N Mehta
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
42
|
Soh MC, Nelson-Piercy C, Westgren M, McCowan L, Pasupathy D. Do adverse pregnancy outcomes contribute to accelerated cardiovascular events seen in young women with systemic lupus erythematosus? Lupus 2017; 26:1351-1367. [PMID: 28728509 DOI: 10.1177/0961203317719146] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiovascular events (CVEs) are prevalent in patients with systemic lupus erythematosus (SLE), and it is the young women who are disproportionately at risk. The risk factors for accelerated cardiovascular disease remain unclear, with multiple studies producing conflicting results. In this paper, we aim to address both traditional and SLE-specific risk factors postulated to drive the accelerated vascular disease in this cohort. We also discuss the more recent hypothesis that adverse pregnancy outcomes in the form of maternal-placental syndrome and resultant preterm delivery could potentially contribute to the CVEs seen in young women with SLE who have fewer traditional cardiovascular risk factors. The pathophysiology of how placental-mediated vascular insufficiency and hypoxia (with the secretion of placenta-like growth factor (PlGF) and soluble fms-tyrosine-like kinase-1 (sFlt-1), soluble endoglin (sEng) and other placental factors) work synergistically to damage the vascular endothelium is discussed. Adverse pregnancy outcomes ultimately are a small contributing factor to the complex pathophysiological process of cardiovascular disease in patients with SLE. Future collaborative studies between cardiologists, obstetricians, obstetric physicians and rheumatologists may pave the way for a better understanding of a likely multifactorial aetiological process.
Collapse
Affiliation(s)
- M C Soh
- 1 Women's Health Academic Centre, King's College London, United Kingdom.,3 Faculty of Medical and Health Science, University of Auckland, New Zealand
| | - C Nelson-Piercy
- 1 Women's Health Academic Centre, King's College London, United Kingdom
| | - M Westgren
- 2 Department of Clinical Science, Karolinska Institutet, Sweden
| | - L McCowan
- 3 Faculty of Medical and Health Science, University of Auckland, New Zealand.,4 National Women's Health, South Auckland Clinical School of Medicine and Counties Manukau Health, Auckland, New Zealand
| | - D Pasupathy
- 1 Women's Health Academic Centre, King's College London, United Kingdom.,5 Biomedical Research Centre at Guy's & St Thomas's NHS Foundation Trust and King's College London, United Kingdom
| |
Collapse
|
43
|
Giannelou M, Mavragani CP. Cardiovascular disease in systemic lupus erythematosus: A comprehensive update. J Autoimmun 2017; 82:1-12. [PMID: 28606749 DOI: 10.1016/j.jaut.2017.05.008] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 05/31/2017] [Indexed: 12/21/2022]
Abstract
Heightened rates of both cardiovascular (CV) events and subclinical atherosclerosis, documented by imaging and vascular function techniques are well established in systemic lupus erythematosus (SLE). While traditional CV factors such as smoking, dyslipidemia, diabetes mellitus (DM), hypertension, central obesity and hyperhomocysteinemia have been reported to be prevalent in lupus patients, they do not fully explain the high rates of ischemic events so far reported, implying that other factors inherent to disease itself could account for the enhanced risk, including disease duration, activity and chronicity, psychosocial factors, medications, genetic variants and altered immunological mechanisms. Though the exact pathogenesis of atherosclerosis in the setting of lupus remains ill defined, an imbalance between endothelial damage and atheroprotection seems to be a central event. Insults leading to endothelial damage in the setting of lupus include oxidized low density lipoprotein (oxLDL), autoantibodies against endothelial cells and phospholipids, type I interferons (IFN) and neutrophil extracellular traps (NETs) directly or through activation of type I IFN pathway. Increased oxidative stress, reduced levels of the normally antioxidant high density lipoprotein (HDL), increased levels of proinflammatory HDL (piHDL) and reduced paraoxonase activity have been related to increased oxLDL levels. On the other hand, impaired atheroprotective mechanisms in lupus include decreased capacity of endothelial repair-partly mediated by type I IFN- and dampened production of atheroprotective autoantibodies. In the present review, traditional and disease related risk factors for CV disease (CVD) in the setting of chronic autoimmune disorders with special focus on SLE will be discussed.
Collapse
Affiliation(s)
- Mayra Giannelou
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece; Department of Rheumatology, General Hospital of Athens "G. Gennimatas", Greece
| | - Clio P Mavragani
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece; Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Greece; Joint Academic Rheumatology Program, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece.
| |
Collapse
|
44
|
Association of the Composite Inflammatory Biomarker GlycA, with Exercise-Induced Changes in Body Habitus in Men and Women with Prediabetes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017. [PMID: 28642810 PMCID: PMC5470023 DOI: 10.1155/2017/5608287] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
GlycA is a new composite measure of systemic inflammation and a predictor of many inflammatory diseases. GlycA is the nuclear magnetic resonance spectroscopy-derived signal arising from glucosamine residues on acute-phase proteins. This study aimed to evaluate how exercise-based lifestyle interventions modulate GlycA in persons at risk for type 2 diabetes. GlycA, fitness, and body habitus were measured in 169 sedentary adults (45–75 years) with prediabetes randomly assigned to one of four six-month exercise-based lifestyle interventions. Interventions included exercise prescription based on the amount (energy expenditure (kcal/kg weight/week (KKW)) and intensity (%VO2peak). The groups were (1) low-amount/moderate-intensity (10KKW/50%) exercise; (2) high-amount/moderate-intensity (16KKW/50%) exercise; (3) high-amount/vigorous-intensity (16KKW/75%) exercise; and (4) a Clinical Lifestyle (combined diet plus low-amount/moderate-intensity exercise) intervention. Six months of exercise training and/or diet-reduced GlycA (mean Δ: −6.8 ± 29.2 μmol/L; p = 0.006) and increased VO2peak (mean Δ: 1.98 ± 2.6 mL/kg/min; p < 0.001). Further, visceral (mean Δ: −21.1 ± 36.6 cm2) and subcutaneous fat (mean Δ: −24.3 ± 41.0 cm2) were reduced, while liver density (mean Δ: +2.3 ± 6.5HU) increased, all p < 0.001. When including individuals in all four interventions, GlycA reductions were associated with reductions in visceral adiposity (p < 0.03). Exercise-based lifestyle interventions reduced GlycA concentrations through mechanisms related to exercise-induced modulations of visceral adiposity. This trial is registered with Clinical Trial Registration Number NCT00962962.
Collapse
|
45
|
Abstract
Systemic lupus erythematosus (SLE) continues to have important morbidity and accelerated mortality despite therapeutic advances. Targeted therapies offer the possibility of improved efficacy with fewer side effects. Current management strategies rely heavily on nonspecific immunosuppressive agents. Prednisone, in particular, is responsible for a considerable burden of later organ damage. There are a multitude of diverse mechanisms of disease activity, immunogenic abnormalities and clinical manifestations to take into consideration in SLE. Many targeted agents with robust mechanistic preclinical data and promising early phase studies have ultimately been disappointing in phase III, randomized, controlled studies. Recent efforts have focused on B-cell therapies, in particular given the success of belimumab in clinical trials, with limited success. We remain optimistic regarding other specific therapies being evaluated, including interferon-alpha blockade. It is likely that in SLE, given the heterogeneity of the population involved, precision medicine is needed, rather than expecting that any single biologic will be universally effective.
Collapse
Affiliation(s)
- L Durcan
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - M Petri
- Division of Rheumatology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| |
Collapse
|
46
|
Pons-Estel GJ, Andreoli L, Scanzi F, Cervera R, Tincani A. The antiphospholipid syndrome in patients with systemic lupus erythematosus. J Autoimmun 2016; 76:10-20. [PMID: 27776934 DOI: 10.1016/j.jaut.2016.10.004] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 10/18/2016] [Indexed: 11/18/2022]
Abstract
The antiphospholipid syndrome (APS) is an autoimmune disease characterized by the occurrence of venous and/or arterial thrombosis and pregnancy morbidity in the presence of pathogenic autoantibodies known as antiphospholipid antibodies (aPL). APS may be associated with other diseases, mainly systemic lupus erythematosus (SLE). The presence or absence of SLE might modify the clinical or serological expression of APS. Apart from the classical manifestations, APS patients with associated SLE more frequently display a clinical profile with arthralgias, arthritis, autoimmune hemolytic anemia, livedo reticularis, epilepsy, glomerular thrombosis, and myocardial infarction. The management of patients with SLE and APS/aPL should include an accurate stratification of vascular risk factors. Low dose aspirin and hydroxychloroquine should be considered as primary prophylaxis. In high risk situations, such as surgery, prolonged immobilization, and puerperium, the prophylaxis should be potentiated with low molecular weight heparin. The challenge of treating patients with a previous vascular event (secondary prophylaxis) is the choice of treatment (anti-platelet agents, anticoagulation with vitamin K antagonists or combined therapy) and its duration, based on individual risk stratification and the site of vascular presentation. The role of novel anticoagulants in APS patients is still to be clearly defined. Novel approaches are needed since the prognosis of SLE patients with APS/aPL is still worse than that of SLE patients with negative aPL. The goal for the future is to improve the outcome of these patients by means of early recognition and optimal preventative treatment.
Collapse
Affiliation(s)
| | - Laura Andreoli
- Department of Clinical and Experimental Sciences, University of Brescia, Rheumatology and Clinical Immunology, Spedali Civili, Brescia, Italy
| | - Francesco Scanzi
- Department of Clinical and Experimental Sciences, University of Brescia, Rheumatology and Clinical Immunology, Spedali Civili, Brescia, Italy
| | - Ricard Cervera
- Department of Autoimmune Diseases, Hospital Clínic, Barcelona, Catalonia, Spain
| | - Angela Tincani
- Department of Clinical and Experimental Sciences, University of Brescia, Rheumatology and Clinical Immunology, Spedali Civili, Brescia, Italy.
| |
Collapse
|
47
|
Joshi AA, Lerman JB, Aberra TM, Afshar M, Teague HL, Rodante JA, Krishnamoorthy P, Ng Q, Aridi TZ, Salahuddin T, Natarajan B, Lockshin BN, Ahlman MA, Chen MY, Rader DJ, Reilly MP, Remaley AT, Bluemke DA, Playford MP, Gelfand JM, Mehta NN. GlycA Is a Novel Biomarker of Inflammation and Subclinical Cardiovascular Disease in Psoriasis. Circ Res 2016; 119:1242-1253. [PMID: 27654120 DOI: 10.1161/circresaha.116.309637] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 09/08/2016] [Accepted: 09/21/2016] [Indexed: 01/26/2023]
Abstract
RATIONALE GlycA, an emerging inflammatory biomarker, predicted cardiovascular events in population-based studies. Psoriasis, an inflammatory disease associated with increased cardiovascular risk, provides a model to study inflammatory biomarkers in cardiovascular disease (CVD). Whether GlycA associates with psoriasis and how it predicts subclinical CVD beyond high-sensitivity C-reactive protein in psoriasis is unknown. OBJECTIVE To investigate the relationships between GlycA and psoriasis and between GlycA and subclinical CVD. METHODS AND RESULTS Patients with psoriasis and controls (n=412) participated in a 2-stage study. We measured GlycA by nuclear magnetic resonance spectroscopy. National Institutes of Health (NIH) participants underwent 18-F Fluorodeoxyglucose Positron Emission Tomography Computed Tomography (18-FDG PET/CT) scans to assess vascular inflammation (VI) and coronary computed tomographic angiography to quantify coronary artery disease burden. Psoriasis cohorts were young (mean age=47.9), with low cardiovascular risk and moderate skin disease. high-sensitivity C-reactive protein and GlycA were increased in psoriasis compared with controls (GlycA: [PENN: 408.8±75.4 versus 289.4±60.2, P<0.0001; NIH: 415.8±63.2 versus 346.2±46, P<0.0001]) and demonstrated a dose-response with psoriasis severity. In stage 2, VI (β=0.36, P<0.001) and coronary artery disease (β=0.29, P=0.004) associated with GlycA beyond CV risk factors in psoriasis. In receiver operating characteristic analysis, GlycA added value in predicting VI (P=0.01) and coronary artery disease (P<0.01). Finally, initiating anti-tumor necrosis factor therapy (n=16) reduced psoriasis severity (P<0.001), GlycA (463.7±92.5 versus 370.1±78.5, P<0.001) and VI (1.93±0.36 versus 1.76±0.19, P<0.001), whereas GlycA remained associated with VI (β=0.56, P<0.001) post treatment. CONCLUSIONS GlycA associated with psoriasis severity and subclinical CVD beyond traditional CV risk and high-sensitivity C-reactive protein. Moreover, psoriasis treatment reduced GlycA and VI. These findings support the potential use of GlycA in subclinical CVD risk assessment in psoriasis and potentially other inflammatory diseases.
Collapse
Affiliation(s)
- Aditya A Joshi
- From the Cardiovascular and Pulmonary Branch (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., M.Y.C., A.T.R., D.A.B., M.P.P., N.N.M.), Section of Inflammation and Cardiometabolic Diseases (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., A.T.R., M.P.P., N.N.M.), National Heart, Lung, and Blood Institute, Bethesda, MD; Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA (A.A.J.); Department of Medicine, McGill University, Montreal, QC, Canada (M.A.); Department of Cardiology, Einstein Institute for Heart and Vascular Health, Albert Einstein Medical Center, Philadelphia, PA (P.K.); Department of Internal Medicine, University of Colorado, Denver (T.S.); Department of Internal Medicine, University of Arizona College of Medicine at South Campus, Tucson (B.N.); Derm Associates, Silver Spring, MD (B.N.L.); Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Research Center, Bethesda, MD (M.A.A.); Perelman School of Medicine (D.J.R.), Department of Dermatology (J.M.G.), and The Center for Clinical Epidemiology and Biostatistics (J.M.G.), University of Pennsylvania, Philadelphia; and Division of Cardiology, Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York (M.P.R.)
| | - Joseph B Lerman
- From the Cardiovascular and Pulmonary Branch (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., M.Y.C., A.T.R., D.A.B., M.P.P., N.N.M.), Section of Inflammation and Cardiometabolic Diseases (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., A.T.R., M.P.P., N.N.M.), National Heart, Lung, and Blood Institute, Bethesda, MD; Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA (A.A.J.); Department of Medicine, McGill University, Montreal, QC, Canada (M.A.); Department of Cardiology, Einstein Institute for Heart and Vascular Health, Albert Einstein Medical Center, Philadelphia, PA (P.K.); Department of Internal Medicine, University of Colorado, Denver (T.S.); Department of Internal Medicine, University of Arizona College of Medicine at South Campus, Tucson (B.N.); Derm Associates, Silver Spring, MD (B.N.L.); Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Research Center, Bethesda, MD (M.A.A.); Perelman School of Medicine (D.J.R.), Department of Dermatology (J.M.G.), and The Center for Clinical Epidemiology and Biostatistics (J.M.G.), University of Pennsylvania, Philadelphia; and Division of Cardiology, Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York (M.P.R.)
| | - Tsion M Aberra
- From the Cardiovascular and Pulmonary Branch (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., M.Y.C., A.T.R., D.A.B., M.P.P., N.N.M.), Section of Inflammation and Cardiometabolic Diseases (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., A.T.R., M.P.P., N.N.M.), National Heart, Lung, and Blood Institute, Bethesda, MD; Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA (A.A.J.); Department of Medicine, McGill University, Montreal, QC, Canada (M.A.); Department of Cardiology, Einstein Institute for Heart and Vascular Health, Albert Einstein Medical Center, Philadelphia, PA (P.K.); Department of Internal Medicine, University of Colorado, Denver (T.S.); Department of Internal Medicine, University of Arizona College of Medicine at South Campus, Tucson (B.N.); Derm Associates, Silver Spring, MD (B.N.L.); Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Research Center, Bethesda, MD (M.A.A.); Perelman School of Medicine (D.J.R.), Department of Dermatology (J.M.G.), and The Center for Clinical Epidemiology and Biostatistics (J.M.G.), University of Pennsylvania, Philadelphia; and Division of Cardiology, Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York (M.P.R.)
| | - Mehdi Afshar
- From the Cardiovascular and Pulmonary Branch (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., M.Y.C., A.T.R., D.A.B., M.P.P., N.N.M.), Section of Inflammation and Cardiometabolic Diseases (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., A.T.R., M.P.P., N.N.M.), National Heart, Lung, and Blood Institute, Bethesda, MD; Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA (A.A.J.); Department of Medicine, McGill University, Montreal, QC, Canada (M.A.); Department of Cardiology, Einstein Institute for Heart and Vascular Health, Albert Einstein Medical Center, Philadelphia, PA (P.K.); Department of Internal Medicine, University of Colorado, Denver (T.S.); Department of Internal Medicine, University of Arizona College of Medicine at South Campus, Tucson (B.N.); Derm Associates, Silver Spring, MD (B.N.L.); Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Research Center, Bethesda, MD (M.A.A.); Perelman School of Medicine (D.J.R.), Department of Dermatology (J.M.G.), and The Center for Clinical Epidemiology and Biostatistics (J.M.G.), University of Pennsylvania, Philadelphia; and Division of Cardiology, Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York (M.P.R.)
| | - Heather L Teague
- From the Cardiovascular and Pulmonary Branch (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., M.Y.C., A.T.R., D.A.B., M.P.P., N.N.M.), Section of Inflammation and Cardiometabolic Diseases (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., A.T.R., M.P.P., N.N.M.), National Heart, Lung, and Blood Institute, Bethesda, MD; Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA (A.A.J.); Department of Medicine, McGill University, Montreal, QC, Canada (M.A.); Department of Cardiology, Einstein Institute for Heart and Vascular Health, Albert Einstein Medical Center, Philadelphia, PA (P.K.); Department of Internal Medicine, University of Colorado, Denver (T.S.); Department of Internal Medicine, University of Arizona College of Medicine at South Campus, Tucson (B.N.); Derm Associates, Silver Spring, MD (B.N.L.); Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Research Center, Bethesda, MD (M.A.A.); Perelman School of Medicine (D.J.R.), Department of Dermatology (J.M.G.), and The Center for Clinical Epidemiology and Biostatistics (J.M.G.), University of Pennsylvania, Philadelphia; and Division of Cardiology, Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York (M.P.R.)
| | - Justin A Rodante
- From the Cardiovascular and Pulmonary Branch (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., M.Y.C., A.T.R., D.A.B., M.P.P., N.N.M.), Section of Inflammation and Cardiometabolic Diseases (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., A.T.R., M.P.P., N.N.M.), National Heart, Lung, and Blood Institute, Bethesda, MD; Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA (A.A.J.); Department of Medicine, McGill University, Montreal, QC, Canada (M.A.); Department of Cardiology, Einstein Institute for Heart and Vascular Health, Albert Einstein Medical Center, Philadelphia, PA (P.K.); Department of Internal Medicine, University of Colorado, Denver (T.S.); Department of Internal Medicine, University of Arizona College of Medicine at South Campus, Tucson (B.N.); Derm Associates, Silver Spring, MD (B.N.L.); Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Research Center, Bethesda, MD (M.A.A.); Perelman School of Medicine (D.J.R.), Department of Dermatology (J.M.G.), and The Center for Clinical Epidemiology and Biostatistics (J.M.G.), University of Pennsylvania, Philadelphia; and Division of Cardiology, Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York (M.P.R.)
| | - Parasuram Krishnamoorthy
- From the Cardiovascular and Pulmonary Branch (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., M.Y.C., A.T.R., D.A.B., M.P.P., N.N.M.), Section of Inflammation and Cardiometabolic Diseases (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., A.T.R., M.P.P., N.N.M.), National Heart, Lung, and Blood Institute, Bethesda, MD; Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA (A.A.J.); Department of Medicine, McGill University, Montreal, QC, Canada (M.A.); Department of Cardiology, Einstein Institute for Heart and Vascular Health, Albert Einstein Medical Center, Philadelphia, PA (P.K.); Department of Internal Medicine, University of Colorado, Denver (T.S.); Department of Internal Medicine, University of Arizona College of Medicine at South Campus, Tucson (B.N.); Derm Associates, Silver Spring, MD (B.N.L.); Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Research Center, Bethesda, MD (M.A.A.); Perelman School of Medicine (D.J.R.), Department of Dermatology (J.M.G.), and The Center for Clinical Epidemiology and Biostatistics (J.M.G.), University of Pennsylvania, Philadelphia; and Division of Cardiology, Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York (M.P.R.)
| | - Qimin Ng
- From the Cardiovascular and Pulmonary Branch (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., M.Y.C., A.T.R., D.A.B., M.P.P., N.N.M.), Section of Inflammation and Cardiometabolic Diseases (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., A.T.R., M.P.P., N.N.M.), National Heart, Lung, and Blood Institute, Bethesda, MD; Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA (A.A.J.); Department of Medicine, McGill University, Montreal, QC, Canada (M.A.); Department of Cardiology, Einstein Institute for Heart and Vascular Health, Albert Einstein Medical Center, Philadelphia, PA (P.K.); Department of Internal Medicine, University of Colorado, Denver (T.S.); Department of Internal Medicine, University of Arizona College of Medicine at South Campus, Tucson (B.N.); Derm Associates, Silver Spring, MD (B.N.L.); Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Research Center, Bethesda, MD (M.A.A.); Perelman School of Medicine (D.J.R.), Department of Dermatology (J.M.G.), and The Center for Clinical Epidemiology and Biostatistics (J.M.G.), University of Pennsylvania, Philadelphia; and Division of Cardiology, Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York (M.P.R.)
| | - Tarek Z Aridi
- From the Cardiovascular and Pulmonary Branch (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., M.Y.C., A.T.R., D.A.B., M.P.P., N.N.M.), Section of Inflammation and Cardiometabolic Diseases (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., A.T.R., M.P.P., N.N.M.), National Heart, Lung, and Blood Institute, Bethesda, MD; Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA (A.A.J.); Department of Medicine, McGill University, Montreal, QC, Canada (M.A.); Department of Cardiology, Einstein Institute for Heart and Vascular Health, Albert Einstein Medical Center, Philadelphia, PA (P.K.); Department of Internal Medicine, University of Colorado, Denver (T.S.); Department of Internal Medicine, University of Arizona College of Medicine at South Campus, Tucson (B.N.); Derm Associates, Silver Spring, MD (B.N.L.); Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Research Center, Bethesda, MD (M.A.A.); Perelman School of Medicine (D.J.R.), Department of Dermatology (J.M.G.), and The Center for Clinical Epidemiology and Biostatistics (J.M.G.), University of Pennsylvania, Philadelphia; and Division of Cardiology, Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York (M.P.R.)
| | - Taufiq Salahuddin
- From the Cardiovascular and Pulmonary Branch (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., M.Y.C., A.T.R., D.A.B., M.P.P., N.N.M.), Section of Inflammation and Cardiometabolic Diseases (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., A.T.R., M.P.P., N.N.M.), National Heart, Lung, and Blood Institute, Bethesda, MD; Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA (A.A.J.); Department of Medicine, McGill University, Montreal, QC, Canada (M.A.); Department of Cardiology, Einstein Institute for Heart and Vascular Health, Albert Einstein Medical Center, Philadelphia, PA (P.K.); Department of Internal Medicine, University of Colorado, Denver (T.S.); Department of Internal Medicine, University of Arizona College of Medicine at South Campus, Tucson (B.N.); Derm Associates, Silver Spring, MD (B.N.L.); Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Research Center, Bethesda, MD (M.A.A.); Perelman School of Medicine (D.J.R.), Department of Dermatology (J.M.G.), and The Center for Clinical Epidemiology and Biostatistics (J.M.G.), University of Pennsylvania, Philadelphia; and Division of Cardiology, Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York (M.P.R.)
| | - Balaji Natarajan
- From the Cardiovascular and Pulmonary Branch (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., M.Y.C., A.T.R., D.A.B., M.P.P., N.N.M.), Section of Inflammation and Cardiometabolic Diseases (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., A.T.R., M.P.P., N.N.M.), National Heart, Lung, and Blood Institute, Bethesda, MD; Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA (A.A.J.); Department of Medicine, McGill University, Montreal, QC, Canada (M.A.); Department of Cardiology, Einstein Institute for Heart and Vascular Health, Albert Einstein Medical Center, Philadelphia, PA (P.K.); Department of Internal Medicine, University of Colorado, Denver (T.S.); Department of Internal Medicine, University of Arizona College of Medicine at South Campus, Tucson (B.N.); Derm Associates, Silver Spring, MD (B.N.L.); Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Research Center, Bethesda, MD (M.A.A.); Perelman School of Medicine (D.J.R.), Department of Dermatology (J.M.G.), and The Center for Clinical Epidemiology and Biostatistics (J.M.G.), University of Pennsylvania, Philadelphia; and Division of Cardiology, Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York (M.P.R.)
| | - Benjamin N Lockshin
- From the Cardiovascular and Pulmonary Branch (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., M.Y.C., A.T.R., D.A.B., M.P.P., N.N.M.), Section of Inflammation and Cardiometabolic Diseases (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., A.T.R., M.P.P., N.N.M.), National Heart, Lung, and Blood Institute, Bethesda, MD; Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA (A.A.J.); Department of Medicine, McGill University, Montreal, QC, Canada (M.A.); Department of Cardiology, Einstein Institute for Heart and Vascular Health, Albert Einstein Medical Center, Philadelphia, PA (P.K.); Department of Internal Medicine, University of Colorado, Denver (T.S.); Department of Internal Medicine, University of Arizona College of Medicine at South Campus, Tucson (B.N.); Derm Associates, Silver Spring, MD (B.N.L.); Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Research Center, Bethesda, MD (M.A.A.); Perelman School of Medicine (D.J.R.), Department of Dermatology (J.M.G.), and The Center for Clinical Epidemiology and Biostatistics (J.M.G.), University of Pennsylvania, Philadelphia; and Division of Cardiology, Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York (M.P.R.)
| | - Mark A Ahlman
- From the Cardiovascular and Pulmonary Branch (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., M.Y.C., A.T.R., D.A.B., M.P.P., N.N.M.), Section of Inflammation and Cardiometabolic Diseases (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., A.T.R., M.P.P., N.N.M.), National Heart, Lung, and Blood Institute, Bethesda, MD; Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA (A.A.J.); Department of Medicine, McGill University, Montreal, QC, Canada (M.A.); Department of Cardiology, Einstein Institute for Heart and Vascular Health, Albert Einstein Medical Center, Philadelphia, PA (P.K.); Department of Internal Medicine, University of Colorado, Denver (T.S.); Department of Internal Medicine, University of Arizona College of Medicine at South Campus, Tucson (B.N.); Derm Associates, Silver Spring, MD (B.N.L.); Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Research Center, Bethesda, MD (M.A.A.); Perelman School of Medicine (D.J.R.), Department of Dermatology (J.M.G.), and The Center for Clinical Epidemiology and Biostatistics (J.M.G.), University of Pennsylvania, Philadelphia; and Division of Cardiology, Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York (M.P.R.)
| | - Marcus Y Chen
- From the Cardiovascular and Pulmonary Branch (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., M.Y.C., A.T.R., D.A.B., M.P.P., N.N.M.), Section of Inflammation and Cardiometabolic Diseases (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., A.T.R., M.P.P., N.N.M.), National Heart, Lung, and Blood Institute, Bethesda, MD; Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA (A.A.J.); Department of Medicine, McGill University, Montreal, QC, Canada (M.A.); Department of Cardiology, Einstein Institute for Heart and Vascular Health, Albert Einstein Medical Center, Philadelphia, PA (P.K.); Department of Internal Medicine, University of Colorado, Denver (T.S.); Department of Internal Medicine, University of Arizona College of Medicine at South Campus, Tucson (B.N.); Derm Associates, Silver Spring, MD (B.N.L.); Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Research Center, Bethesda, MD (M.A.A.); Perelman School of Medicine (D.J.R.), Department of Dermatology (J.M.G.), and The Center for Clinical Epidemiology and Biostatistics (J.M.G.), University of Pennsylvania, Philadelphia; and Division of Cardiology, Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York (M.P.R.)
| | - Daniel J Rader
- From the Cardiovascular and Pulmonary Branch (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., M.Y.C., A.T.R., D.A.B., M.P.P., N.N.M.), Section of Inflammation and Cardiometabolic Diseases (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., A.T.R., M.P.P., N.N.M.), National Heart, Lung, and Blood Institute, Bethesda, MD; Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA (A.A.J.); Department of Medicine, McGill University, Montreal, QC, Canada (M.A.); Department of Cardiology, Einstein Institute for Heart and Vascular Health, Albert Einstein Medical Center, Philadelphia, PA (P.K.); Department of Internal Medicine, University of Colorado, Denver (T.S.); Department of Internal Medicine, University of Arizona College of Medicine at South Campus, Tucson (B.N.); Derm Associates, Silver Spring, MD (B.N.L.); Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Research Center, Bethesda, MD (M.A.A.); Perelman School of Medicine (D.J.R.), Department of Dermatology (J.M.G.), and The Center for Clinical Epidemiology and Biostatistics (J.M.G.), University of Pennsylvania, Philadelphia; and Division of Cardiology, Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York (M.P.R.)
| | - Muredach P Reilly
- From the Cardiovascular and Pulmonary Branch (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., M.Y.C., A.T.R., D.A.B., M.P.P., N.N.M.), Section of Inflammation and Cardiometabolic Diseases (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., A.T.R., M.P.P., N.N.M.), National Heart, Lung, and Blood Institute, Bethesda, MD; Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA (A.A.J.); Department of Medicine, McGill University, Montreal, QC, Canada (M.A.); Department of Cardiology, Einstein Institute for Heart and Vascular Health, Albert Einstein Medical Center, Philadelphia, PA (P.K.); Department of Internal Medicine, University of Colorado, Denver (T.S.); Department of Internal Medicine, University of Arizona College of Medicine at South Campus, Tucson (B.N.); Derm Associates, Silver Spring, MD (B.N.L.); Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Research Center, Bethesda, MD (M.A.A.); Perelman School of Medicine (D.J.R.), Department of Dermatology (J.M.G.), and The Center for Clinical Epidemiology and Biostatistics (J.M.G.), University of Pennsylvania, Philadelphia; and Division of Cardiology, Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York (M.P.R.)
| | - Alan T Remaley
- From the Cardiovascular and Pulmonary Branch (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., M.Y.C., A.T.R., D.A.B., M.P.P., N.N.M.), Section of Inflammation and Cardiometabolic Diseases (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., A.T.R., M.P.P., N.N.M.), National Heart, Lung, and Blood Institute, Bethesda, MD; Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA (A.A.J.); Department of Medicine, McGill University, Montreal, QC, Canada (M.A.); Department of Cardiology, Einstein Institute for Heart and Vascular Health, Albert Einstein Medical Center, Philadelphia, PA (P.K.); Department of Internal Medicine, University of Colorado, Denver (T.S.); Department of Internal Medicine, University of Arizona College of Medicine at South Campus, Tucson (B.N.); Derm Associates, Silver Spring, MD (B.N.L.); Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Research Center, Bethesda, MD (M.A.A.); Perelman School of Medicine (D.J.R.), Department of Dermatology (J.M.G.), and The Center for Clinical Epidemiology and Biostatistics (J.M.G.), University of Pennsylvania, Philadelphia; and Division of Cardiology, Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York (M.P.R.)
| | - David A Bluemke
- From the Cardiovascular and Pulmonary Branch (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., M.Y.C., A.T.R., D.A.B., M.P.P., N.N.M.), Section of Inflammation and Cardiometabolic Diseases (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., A.T.R., M.P.P., N.N.M.), National Heart, Lung, and Blood Institute, Bethesda, MD; Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA (A.A.J.); Department of Medicine, McGill University, Montreal, QC, Canada (M.A.); Department of Cardiology, Einstein Institute for Heart and Vascular Health, Albert Einstein Medical Center, Philadelphia, PA (P.K.); Department of Internal Medicine, University of Colorado, Denver (T.S.); Department of Internal Medicine, University of Arizona College of Medicine at South Campus, Tucson (B.N.); Derm Associates, Silver Spring, MD (B.N.L.); Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Research Center, Bethesda, MD (M.A.A.); Perelman School of Medicine (D.J.R.), Department of Dermatology (J.M.G.), and The Center for Clinical Epidemiology and Biostatistics (J.M.G.), University of Pennsylvania, Philadelphia; and Division of Cardiology, Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York (M.P.R.)
| | - Martin P Playford
- From the Cardiovascular and Pulmonary Branch (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., M.Y.C., A.T.R., D.A.B., M.P.P., N.N.M.), Section of Inflammation and Cardiometabolic Diseases (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., A.T.R., M.P.P., N.N.M.), National Heart, Lung, and Blood Institute, Bethesda, MD; Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA (A.A.J.); Department of Medicine, McGill University, Montreal, QC, Canada (M.A.); Department of Cardiology, Einstein Institute for Heart and Vascular Health, Albert Einstein Medical Center, Philadelphia, PA (P.K.); Department of Internal Medicine, University of Colorado, Denver (T.S.); Department of Internal Medicine, University of Arizona College of Medicine at South Campus, Tucson (B.N.); Derm Associates, Silver Spring, MD (B.N.L.); Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Research Center, Bethesda, MD (M.A.A.); Perelman School of Medicine (D.J.R.), Department of Dermatology (J.M.G.), and The Center for Clinical Epidemiology and Biostatistics (J.M.G.), University of Pennsylvania, Philadelphia; and Division of Cardiology, Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York (M.P.R.)
| | - Joel M Gelfand
- From the Cardiovascular and Pulmonary Branch (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., M.Y.C., A.T.R., D.A.B., M.P.P., N.N.M.), Section of Inflammation and Cardiometabolic Diseases (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., A.T.R., M.P.P., N.N.M.), National Heart, Lung, and Blood Institute, Bethesda, MD; Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA (A.A.J.); Department of Medicine, McGill University, Montreal, QC, Canada (M.A.); Department of Cardiology, Einstein Institute for Heart and Vascular Health, Albert Einstein Medical Center, Philadelphia, PA (P.K.); Department of Internal Medicine, University of Colorado, Denver (T.S.); Department of Internal Medicine, University of Arizona College of Medicine at South Campus, Tucson (B.N.); Derm Associates, Silver Spring, MD (B.N.L.); Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Research Center, Bethesda, MD (M.A.A.); Perelman School of Medicine (D.J.R.), Department of Dermatology (J.M.G.), and The Center for Clinical Epidemiology and Biostatistics (J.M.G.), University of Pennsylvania, Philadelphia; and Division of Cardiology, Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York (M.P.R.)
| | - Nehal N Mehta
- From the Cardiovascular and Pulmonary Branch (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., M.Y.C., A.T.R., D.A.B., M.P.P., N.N.M.), Section of Inflammation and Cardiometabolic Diseases (A.A.J., J.B.L., T.M.A., H.L.T., J.A.R., Q.N., T.Z.A., T.S., B.N., A.T.R., M.P.P., N.N.M.), National Heart, Lung, and Blood Institute, Bethesda, MD; Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA (A.A.J.); Department of Medicine, McGill University, Montreal, QC, Canada (M.A.); Department of Cardiology, Einstein Institute for Heart and Vascular Health, Albert Einstein Medical Center, Philadelphia, PA (P.K.); Department of Internal Medicine, University of Colorado, Denver (T.S.); Department of Internal Medicine, University of Arizona College of Medicine at South Campus, Tucson (B.N.); Derm Associates, Silver Spring, MD (B.N.L.); Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Research Center, Bethesda, MD (M.A.A.); Perelman School of Medicine (D.J.R.), Department of Dermatology (J.M.G.), and The Center for Clinical Epidemiology and Biostatistics (J.M.G.), University of Pennsylvania, Philadelphia; and Division of Cardiology, Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York (M.P.R.).
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Atherosclerotic cardiovascular disease confers significant morbidity and mortality in patients with systemic lupus erythematosus (SLE) and cannot be fully explained by traditional cardiovascular risk factors. Recent immunologic discoveries have outlined putative pathways in SLE that may also accelerate the development of atherosclerosis. RECENT FINDINGS Aberrant innate and adaptive immune responses implicated in lupus pathogenesis may also contribute to the development of accelerated atherosclerosis in these patients. Defective apoptosis, abnormal lipoprotein function, autoantibodies, aberrant neutrophil responses, and a dysregulated type I interferon pathway likely contribute to endothelial dysfunction. SLE macrophages have an inflammatory phenotype that may drive progression of plaque. SUMMARY Recent discoveries have placed increased emphasis on the immunology of atherosclerotic cardiovascular disease. Understanding the factors that drive the increased risk for cardiovascular disease in SLE patients may provide selective therapeutic targets for reducing inflammation and improving outcomes in atherosclerosis.
Collapse
Affiliation(s)
- Laura B. Lewandowski
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Mariana J. Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
49
|
Durcan L, Petri M. Immunomodulators in SLE: Clinical evidence and immunologic actions. J Autoimmun 2016; 74:73-84. [PMID: 27371107 DOI: 10.1016/j.jaut.2016.06.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 06/16/2016] [Accepted: 06/21/2016] [Indexed: 01/08/2023]
Abstract
Systemic lupus erythematosus (SLE) is a potentially fatal autoimmune disease. Current treatment strategies rely heavily on corticosteroids, which are in turn responsible for a significant burden of morbidity, and immunosuppressives which are limited by suboptimal efficacy, increased infections and malignancies. There are significant deficiencies in our immunosuppressive armamentarium, making immunomodulatory therapies crucial, offering the opportunity to prevent disease flare and the subsequent accrual of damage. Currently available immunomodulators include prasterone (synthetic dehydroeipandrosterone), vitamin D, hydroxychloroquine and belimumab. These therapies, acting via numerous cellular and cytokine pathways, have been shown to modify the aberrant immune responses associated with SLE without overt immunosuppression. Vitamin D is important in SLE and supplementation appears to have a positive impact on disease activity particularly proteinuria. Belimumab has specific immunomodulatory properties and is an effective therapy in those with specific serological and clinical characteristics predictive of response. Hydroxychloroquine is a crucial background medication in SLE with actions in many molecular pathways. It has disease specific effects in reducing flare, treating cutaneous disease and inflammatory arthralgias in addition to other effects such as reduced thrombosis, increased longevity, improved lipids, better glycemic control and blood pressure. Dehydroeipandrosterone is also an immunomodulator in SLE which can have positive effects on disease activity and has bone protective properties. This review outlines the immunologic actions of these drugs and the clinical evidence supporting their use.
Collapse
Affiliation(s)
- L Durcan
- Division of Rheumatology, University of Washington, Seattle, USA.
| | - M Petri
- Division of Rheumatology, Johns Hopkins University, School of Medicine, Baltimore, USA
| |
Collapse
|
50
|
Inflammatory glycoproteins in cardiometabolic disorders, autoimmune diseases and cancer. Clin Chim Acta 2016; 459:177-186. [PMID: 27312321 DOI: 10.1016/j.cca.2016.06.012] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 06/08/2016] [Accepted: 06/11/2016] [Indexed: 12/25/2022]
Abstract
The physiological function initially attributed to the oligosaccharide moieties or glycans on inflammatory glycoproteins was to improve protein stability. However, it is now clear that glycans play a prominent role in glycoprotein structure and function and in some cases contribute to disease states. In fact, glycan processing contributes to pathogenicity not only in autoimmune disorders but also in atherosclerotic cardiovascular disease, diabetes and malignancy. While most clinical laboratory tests measure circulating levels of inflammatory proteins, newly developed diagnostic and prognostic tests are harvesting the information that can be gleaned by measuring the amount or structure of the attached glycans, which may be unique to individuals as well as various diseases. As such, these newer glycan-based tests may provide future means for more personalized approaches to patient stratification and improved patient care. Here we will discuss recent progress in high-throughput laboratory methods for glycomics (i.e. the study of glycan structures) and glycoprotein quantification by methods such as mass spectrometry and nuclear magnetic resonance spectroscopy. We will also review the clinical utility of glycoprotein and glycan measurements in the prediction of common low-grade inflammatory disorders including cardiovascular disease, diabetes and cancer, as well as for monitoring autoimmune disease activity.
Collapse
|