1
|
Zheng D, Li X, Wang P, Zhu Q, Huang Z, Zhao T. Exploring the shared mechanism of fatigue between systemic lupus erythematosus and myalgic encephalomyelitis/chronic fatigue syndrome: monocytic dysregulation and drug repurposing. Front Immunol 2025; 15:1440922. [PMID: 39845969 PMCID: PMC11752880 DOI: 10.3389/fimmu.2024.1440922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 12/13/2024] [Indexed: 01/24/2025] Open
Abstract
Background SLE and ME/CFS both present significant fatigue and share immune dysregulation. The mechanisms underlying fatigue in these disorders remain unclear, and there are no standardized treatments. This study aims to explore shared mechanisms and predict potential therapeutic drugs for fatigue in SLE and ME/CFS. Methods Genes associated with SLE and ME/CFS were collected from disease target and clinical sample databases to identify overlapping genes. Bioinformatics analyses, including GO, KEGG, PPI network construction, and key target identification, were performed. ROC curve and correlation analysis of key targets, along with single-cell clustering, were conducted to validate their expression in different cell types. Additionally, an inflammation model was established using THP-1 cells to simulate monocyte activation in both diseases in vitro, and RT-qPCR was used to validate the expression of the key targets. A TF-mRNA-miRNA co-regulatory network was constructed, followed by drug prediction and molecular docking. Results Fifty-eight overlapping genes were identified, mainly involved in innate immunity and inflammation. Five key targets were identified (IL1β, CCL2, TLR2, STAT1, IFIH1). Single-cell sequencing revealed that monocytes are enriched with these targets. RT-qPCR confirmed significant upregulation of these targets in the model group. A co-regulatory network was constructed, and ten potential drugs, including suloctidil, N-Acetyl-L-cysteine, simvastatin, ACMC-20mvek, and camptothecin, were predicted. Simvastatin and camptothecin showed high affinity for the key targets. Conclusion SLE and ME/CFS share immune and inflammatory pathways. The identified key targets are predominantly enriched in monocytes at the single-cell level, suggesting that classical monocytes may be crucial in linking inflammation and fatigue. RT-qPCR confirmed upregulation in activated monocytes. The TF-mRNA-miRNA network provides a foundation for future research, and drug prediction suggests N-Acetyl-L-cysteine and camptothecin as potential therapies.
Collapse
Affiliation(s)
- Daisi Zheng
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaolong Li
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Peicheng Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Qingmiao Zhu
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhiyan Huang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Ting Zhao
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
2
|
Iwaya C, Iwata J. Associations between metabolic disorders and Sjögren's disease. JAPANESE DENTAL SCIENCE REVIEW 2024; 60:232-238. [PMID: 39502167 PMCID: PMC11535258 DOI: 10.1016/j.jdsr.2024.06.002] [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: 04/17/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 11/08/2024] Open
Abstract
Sjögren's disease (SjD) is a systemic autoimmune disorder characterized by dry eyes and mouth caused by chronic inflammation and is often accompanied by various extra-glandular manifestations, including fatigue and diffuse pain. Although the pathogenesis of the disease remains elusive, several factors (e.g. environmental, genetic and hormonal factors, abnormal metabolic status) are associated with this condition. Accumulating evidence suggests a potential role of cholesterol metabolism in immune and non-immune modulation in various diseases. In this review, we summarize the current findings on the associations between cholesterol metabolism and SjD.
Collapse
Affiliation(s)
- Chihiro Iwaya
- Department of Diagnostic & Biomedical Sciences, The University of Texas Health Science Center at Houston (UTHealth), School of Dentistry, Houston, Texas 77054, USA
- Center for Craniofacial Research, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA
| | - Junichi Iwata
- Department of Diagnostic & Biomedical Sciences, The University of Texas Health Science Center at Houston (UTHealth), School of Dentistry, Houston, Texas 77054, USA
- Center for Craniofacial Research, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
3
|
Wilkinson MJ, Shapiro MD. Immune-Mediated Inflammatory Diseases, Dyslipidemia, and Cardiovascular Risk: A Complex Interplay. Arterioscler Thromb Vasc Biol 2024; 44:2396-2406. [PMID: 39479765 PMCID: PMC11602385 DOI: 10.1161/atvbaha.124.319983] [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] [Indexed: 11/29/2024]
Abstract
Individuals with autoimmune inflammatory diseases, such as systemic lupus erythematosus, rheumatoid arthritis, and psoriasis, are at increased risk for cardiovascular disease. While these diseases share common features of systemic inflammation, the impact of individual autoimmune inflammatory conditions on circulating lipids and lipoproteins varies by specific disease, disease activity, and the immune-suppressing medications used to treat these conditions. A common feature observed in many autoimmune inflammatory diseases is the development of a proatherogenic dyslipidemic state, characterized by dysfunctional HDLs (high-density lipoproteins) and increased oxidation of LDLs (low-density lipoproteins). Various disease-modifying antirheumatic drugs also have complex and variable effects on lipids, and it is critical to take this into consideration when evaluating lipid-related risk in individuals with immune-mediated inflammatory conditions. This review aims to critically evaluate the current understanding of the relationship between immune-mediated inflammatory diseases and dyslipidemia, the underlying mechanisms contributing to atherogenesis, and the impact of various pharmacotherapies on lipid profiles and cardiovascular risk. We also discuss the role of lipid-lowering therapies, particularly statins, in managing residual risk in this high-risk population and explore the potential of emerging therapies with complementary anti-inflammatory and lipid-lowering effects.
Collapse
Affiliation(s)
- Michael J. Wilkinson
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093, USA
| | - Michael D. Shapiro
- Center for Prevention of Cardiovascular Disease, Section on Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
4
|
Pan GH, Zhang JQ, Sun YY, Shi YH, Zhang FR. Saturation association between serum 25-hydroxyvitamin D levels and mortality in elderly people with hyperlipidemia: a population-based study from the NHANES (2001-2016). Front Endocrinol (Lausanne) 2024; 15:1382419. [PMID: 39415789 PMCID: PMC11479873 DOI: 10.3389/fendo.2024.1382419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 09/06/2024] [Indexed: 10/19/2024] Open
Abstract
Background 25-hydroxyvitamin D is the body's main storage form of vitamin D and is internationally recognized as the best indicator of vitamin D status in the human body. There is a scarcity of research investigating the interrelationship between serum 25-hydroxyvitamin D (25(OH)D) levels and mortality among elderly individuals with hyperlipidemia. To address this knowledge gap, we examined the association between serum 25(OH)D levels and mortality in an older hyperlipidemic population from NHANES, while controlling for other influential factors. The study sought to elucidate the correlation between serum 25(OH)D levels and mortality about all-cause mortality, cardiovascular disease (CVD), malignant neoplasms, and mortality from other causes. Methods The data from NHANES 2001-2016, including 9,271 participants were analyzed to examine the association between serum 25(OH)D levels and mortality. The interrelationship was illustrated using Kaplan-Meier curves and restricted cubic splines, while the Cox proportional hazards model was utilized to estimate the multifactor adjusted hazard ratio (HR). Results This study included 9,271 participants (43.28% male) with an average age of 69.58 years, and the average duration of participant follow-up was 88.37 months. Kaplan-Meier curves demonstrated that lower serum 25(OH)D levels were associated with increased risks of all-cause mortality, cardiovascular mortality, malignant neoplasm mortality, and mortality from other causes. This negative association was further confirmed by the Cox proportional hazards models. Additionally, restricted cubic splines not only revealed this negative association but also highlighted the saturated serum 25(OH)D levels. Moreover, subgroup analyses indicated that the inverse correlation between serum 25(OH)D levels and all-cause mortality was more pronounced in the non-obese and smoking population. And the inverse correlation with mortality from other causes was even stronger in the non-obese population. Conclusions In the elderly population with hyperlipidemia, 25(OH)D serum levels were negatively correlated with both cause-specific mortality and all-cause mortality. Moreover, there was a threshold effect in this negative association.
Collapse
Affiliation(s)
- Guang-hui Pan
- Department of Nephrology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jun-qing Zhang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yi-yan Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yue-hui Shi
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fa-rong Zhang
- Department of Nephrology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
5
|
Jury EC, Peng J, Van Vijfeijken A, Martin Gutierrez L, Woodridge L, Wincup C, Pineda-Torra I, Ciurtin C, Robinson GA. Systemic lupus erythematosus patients have unique changes in serum metabolic profiles across age associated with cardiometabolic risk. Rheumatology (Oxford) 2024; 63:2741-2753. [PMID: 38048621 PMCID: PMC11443078 DOI: 10.1093/rheumatology/kead646] [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: 08/18/2023] [Revised: 11/02/2023] [Accepted: 11/05/2023] [Indexed: 12/06/2023] Open
Abstract
OBJECTIVES Cardiovascular disease through accelerated atherosclerosis is a leading cause of mortality for patients with systemic lupus erythematosus (SLE), likely due to increased chronic inflammation and cardiometabolic defects over age. We investigated age-associated changes in metabolomic profiles of SLE patients and healthy controls (HCs). METHODS Serum NMR metabolomic profiles from female SLE patients (n = 164, age = 14-76) and HCs (n = 123, age = 13-72) were assessed across age by linear regression and by age group between patients/HCs (Group 1, age ≤ 25, n = 62/46; Group 2, age = 26-49, n = 50/46; Group 3, age ≥ 50, n = 52/31) using multiple t tests. The impact of inflammation, disease activity and treatments were assessed, and UK Biobank disease-wide association analysis of metabolites was performed. RESULTS Age-specific metabolomic profiles were identified in SLE patients vs HCs, including reduced amino acids (Group 1), increased very-low-density lipoproteins (Group 2), and increased low-density lipoproteins (Group 3). Twenty-five metabolites were significantly altered in all SLE age groups, dominated by decreased atheroprotective high-density lipoprotein (HDL) subsets, HDL-bound apolipoprotein (Apo)A1 and increased glycoprotein acetyls (GlycA). Furthermore, ApoA1 and GlycA were differentially associated with disease activity and serological measures, as well as atherosclerosis incidence and myocardial infarction mortality risk through disease-wide association. Separately, glycolysis pathway metabolites (acetone/citrate/creatinine/glycerol/lactate/pyruvate) uniquely increased with age in SLE, significantly influenced by prednisolone (increased pyruvate/lactate) and hydroxychloroquine (decreased citrate/creatinine) treatment and associated with type 1 and type 2 diabetes by disease-wide association. CONCLUSIONS Increasing HDL (ApoA1) levels through therapeutic/nutritional intervention, whilst maintaining low disease activity, in SLE patients from a young age could improve cardiometabolic disease outcomes. Biomarkers from the glycolytic pathway could indicate adverse metabolic effects of current therapies.
Collapse
Affiliation(s)
- Elizabeth C Jury
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
| | - Junjie Peng
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| | | | - Lucia Martin Gutierrez
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| | - Laurel Woodridge
- Centre for Experimental & Translational Medicine, Division of Medicine, University College London, London, UK
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
| | - Chris Wincup
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
| | - Ines Pineda-Torra
- Centre for Experimental & Translational Medicine, Division of Medicine, University College London, London, UK
| | - Coziana Ciurtin
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| | - George A Robinson
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| |
Collapse
|
6
|
Atzeni F, Rodríguez-Pintó I, Cervera R. Cardiovascular disease risk in systemic lupus erythematous: Certainties and controversies. Autoimmun Rev 2024; 23:103646. [PMID: 39321952 DOI: 10.1016/j.autrev.2024.103646] [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: 06/09/2024] [Revised: 09/19/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024]
Abstract
Patients with systemic lupus erythematosus (SLE) experience greater cardiovascular morbidity and mortality compared to the general population. It is known that endothelial dysfunction, an early indicator of atherosclerosis development, can arise even without the presence of conventional cardiovascular risk factors. In fact, the risk factors contributing to cardiovascular disease can be classified into traditional risk factors and those uniquely associated with SLE such as disease activity, autoantibodies, etc.Furthermore, the pathogenesis of cardiovascular disease in SLE is linked to the activation of both the innate and adaptive immune systems. Given these findings, it is essential for clinicians to acknowledge the heightened CVD risk in SLE patients, perform comprehensive screenings for cardiovascular risk factors, and implement aggressive treatment strategies for those who exhibit signs of clinical CVD. The aim of this review is to summarize the findings on cardiovascular disease in SLE and to examine potential screening and therapeutic strategies for clinical practice.
Collapse
Affiliation(s)
- Fabiola Atzeni
- Rheumatology Unit, Department of Experimental and Internal Medicine, University of Messina, Messina, Italy
| | - Ignasi Rodríguez-Pintó
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases (UEC/CSUR) of the Catalan and Spanish Health Systems-Member of ERNReCONNET, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.
| | - Ricard Cervera
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases (UEC/CSUR) of the Catalan and Spanish Health Systems-Member of ERNReCONNET, Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.
| |
Collapse
|
7
|
Calabrese E, Pressman P. Enhancing the human health and lifespan: a targeted strategy emphasizing statins. Biogerontology 2024; 25:883-890. [PMID: 38811414 DOI: 10.1007/s10522-024-10112-y] [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: 04/24/2024] [Accepted: 05/10/2024] [Indexed: 05/31/2024]
Abstract
There has been substantial research interest in finding activities/agents that slow the onset and reduce the severity of numerous age-related diseases/conditions. This assessment indicates that the most studied agent intended to promote health in human population investigations for a broad spectrum of diseases are the statins, with large-scale epidemiological studies addressing numerous health endpoints. The key findings are that statin treatment consistently reduces the occurrence and attenuates the course of numerous non-communicable and contagious pathologies and numerous types of cancer with high mortality rates by about 20-50%. That one agent could affect such a broad based and consistently positive trends in epidemiological studies is unexpected and impressive, along with consistent cell and animal model research. Underlying mechanisms have been proposed that significantly contribute to the spectrum of salutary effects of statins, especially the capacity to activate Nrf2 showing hormetic dose responses in multiple organs and cell types, due to its bioavailability and broad tissue distribution. The widespread use of statins, which has the capacity to enhance human health span, should be considered for experimental exploration as a novel public health strategy that includes practical approaches for reduction of the most common adverse effects of this class of drugs including myalgia/myopathy and transaminitis.
Collapse
Affiliation(s)
- Edward Calabrese
- Department of Environmental Health Sciences, University of Massachusetts, Morrill I, N344, Amherst, MA, 01003, USA.
| | - Peter Pressman
- University of Maine, 5728 Fernald Hall, Room 201, Orono, ME, 04469, USA
| |
Collapse
|
8
|
Bolla E, Semb AG, Kerola AM, Ikdahl E, Petri M, Pons-Estel GJ, Karpouzas GA, Sfikakis PP, Quintana R, Misra DP, Borba EF, Garcia-de la Torre I, Popkova TV, Artim-Esen B, Troldborg A, Fragoso-Loyo H, Ajeganova S, Yazici A, Aroca-Martinez G, Direskeneli H, Ugarte-Gil MF, Mosca M, Goyal M, Svenungsson E, Macieira C, Hoi A, Lerang K, Costedoat-Chalumeau N, Tincani A, Mirrakhimov E, Acosta Colman I, Danza A, Massardo L, Blagojevic J, Yılmaz N, Tegzová D, Yavuz S, Korkmaz C, Hachulla E, Moreno Alvarez MJ, Muñoz-Louis R, Pantazis N, Tektonidou MG. Prevalence and target attainment of traditional cardiovascular risk factors in patients with systemic lupus erythematosus: a cross-sectional study including 3401 individuals from 24 countries. THE LANCET. RHEUMATOLOGY 2024; 6:e447-e459. [PMID: 38878780 DOI: 10.1016/s2665-9913(24)00090-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is characterised by increased cardiovascular morbidity and mortality risk. We aimed to examine the prevalence of traditional cardiovascular risk factors and their control in an international survey of patients with systemic lupus erythematosus. METHODS In this multicentre, cross-sectional study, cardiovascular risk factor data from medical files of adult patients (aged ≥18) with SLE followed between Jan 1, 2015, and Jan 1, 2020, were collected from 24 countries, across five continents. We assessed the prevalence and target attainment of cardiovascular risk factors and examined potential differences by country income level and antiphospholipid syndrome coexistence. We used the Systemic Coronary Risk Evaluation algorithm for cardiovascular risk estimation, and the European Society of Cardiology guidelines for assessing cardiovascular risk factor target attainment. People with lived experience were not involved in the research or writing process. FINDINGS 3401 patients with SLE were included in the study. The median age was 43·0 years (IQR 33-54), 3047 (89·7%) of 3396 patients were women, 349 (10.3%) were men, and 1629 (48·1%) of 3390 were White. 556 (20·7%) of 2681 patients had concomitant antiphospholipid syndrome. We found a high cardiovascular risk factor prevalence (hypertension 1210 [35·6%] of 3398 patients, obesity 751 [23·7%] of 3169 patients, and hyperlipidaemia 650 [19·8%] of 3279 patients), and suboptimal control of modifiable cardiovascular risk factors (blood pressure [target of <130/80 mm Hg], BMI, and lipids) in the entire SLE group. Higher prevalence of cardiovascular risk factors but a better blood pressure (target of <130/80 mm Hg; 54·9% [1170 of 2132 patients] vs 46·8% [519 of 1109 patients]; p<0·0001), and lipid control (75·0% [895 of 1194 patients] vs 51·4% [386 of 751 patients], p<0·0001 for high-density lipoprotein [HDL]; 66·4% [769 of 1158 patients] vs 60·8% [453 of 745 patients], p=0·013 for non-HDL; 80·9% [1017 of 1257 patients] vs 61·4% [486 of 792 patients], p<0·0001 for triglycerides]) was observed in patients from high-income versus those from middle-income countries. Patients with SLE with antiphospholipid syndrome had a higher prevalence of modifiable cardiovascular risk factors, and significantly lower attainment of BMI and lipid targets (for low-density lipoprotein and non-HDL) than patients with SLE without antiphospholipid syndrome. INTERPRETATION High prevalence and inadequate cardiovascular risk factor control were observed in a large multicentre and multiethnic SLE cohort, especially among patients from middle-income compared with high-income countries and among those with coexistent antiphospholipid syndrome. Increased awareness of cardiovascular disease risk in SLE, especially in the above subgroups, is urgently warranted. FUNDING None.
Collapse
Affiliation(s)
- Eleana Bolla
- Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Joint Academic Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Anne Grete Semb
- Preventive Cardio-Rheuma Clinic, Division of Research and Innovation, REMEDY Centre, Diakonhjemmet Hospital, Oslo, Norway
| | - Anne M Kerola
- Department of Internal Medicine, Päijät-Häme Central Hospital, Lahti, Finland; Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Eirik Ikdahl
- Preventive Cardio-Rheuma Clinic, Division of Research and Innovation, REMEDY Centre, Diakonhjemmet Hospital, Oslo, Norway
| | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - George A Karpouzas
- Division of Rheumatology, Harbor-UCLA Medical Center, Torrance, CA, USA; The Lundquist Institute, Torrance, CA, USA
| | - Petros P Sfikakis
- Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Joint Academic Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Rosana Quintana
- Grupo Oroño, Centro Regional de Enfermedades Autoinmunes y Reumáticas, Rosario, Argentina
| | - Durga Prasanna Misra
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Eduardo Ferreira Borba
- Rheumatology Division, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Ignacio Garcia-de la Torre
- Departamento de Inmunología y Reumatología, Hospital General de Occidente, Jalisco, Mexico; Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | | | - Bahar Artim-Esen
- Department of Internal Medicine, Division of Rheumatology, Faculty of Medicine, Istanbul University, Istanbul, Türkiye
| | - Anne Troldborg
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Hilda Fragoso-Loyo
- Immunology and Rheumatology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Sofia Ajeganova
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Sciences, Rheumatology Division, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ayten Yazici
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Kocaeli University, Kocaeli, Türkiye
| | | | - Haner Direskeneli
- Division of Rheumatology, Department of Internal Medicine, Marmara University, School of Medicine, Istanbul, Türkiye
| | - Manuel F Ugarte-Gil
- Grupo Peruano de Estudio de Enfermedades Autoinmunes Sistémicas, Universidad Científica del Sur, Lima, Peru; Rheumatology Department, Hospital Nacional Guillermo Almenara Irigoyen, EsSalud, Lima, Peru
| | - Marta Mosca
- Rheumatology Unit, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Mohit Goyal
- Division of Rheumatology, CARE Pain and Arthritis Centre, Udaipur, India
| | - Elisabet Svenungsson
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Carla Macieira
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon Academic Medical Centre, Lisboa, Portugal
| | - Alberta Hoi
- Department of Rheumatology, Monash Health, Clayton, VIC, Australia; School of Clinical Science, Monash University, Clayton, VIC, Australia
| | - Karoline Lerang
- Department of Rheumatology, Oslo University Hospital, Oslo, Norway
| | - Nathalie Costedoat-Chalumeau
- National Referral Centre for Rare Autoimmune and Systemic Diseases, Department of Internal Medicine, Hôpital Cochin, AP-HP Centre, Université Paris Cité, Paris, France
| | - Angela Tincani
- Rheumatology and Clinical Immunology Unit-ASST Spedali Civili, Brescia, Italy
| | | | - Isabel Acosta Colman
- Department of Rheumatology, Universidad Nacional de Asunción, Asuncion, Paraguay
| | - Alvaro Danza
- Department of Internal Medicine, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Loreto Massardo
- Centro de Biología Celular y Biomedicina, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Jelena Blagojevic
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Department of Geriatric Medicine, Division of Rheumatology and Scleroderma Unit, AOUC, Florence, Italy
| | - Neslihan Yılmaz
- Rheumatology Department, Faculty of Medicine, Demiroglu Bilim University, Istanbul, Türkiye
| | - Dana Tegzová
- Institute of Rheumatology, Prague, Czech Republic
| | - Sule Yavuz
- Department of Internal Medicine, Division of Rheumatology, Istanbul Bilim University, Istanbul, Türkiye
| | - Cengiz Korkmaz
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Eskişehir Osmangazi University, Eskişehir, Türkiye
| | - Eric Hachulla
- Department of Internal Medicine and Clinical Immunology, National Referral Centre for Rare Systemic Auto-Immune and Auto-inflammatory Diseases, University of Lille, Inserm, CHU Lille, U1286 - INFINITE, University of Lille, Lille, France
| | - Mario J Moreno Alvarez
- Servicio Reumatología, Hospital Luis Vernaza, Universidad de Especialidades Espíritu Santo, Guayaquil, Ecuador
| | - Roberto Muñoz-Louis
- Servicio de Reumatología, Hospital Docente Padre Billini, Santo Domingo, Dominican Republic
| | - Nikos Pantazis
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria G Tektonidou
- Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Joint Academic Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece.
| |
Collapse
|
9
|
Cintron D, Chang JC, Sadun RE. Transition to Adult Rheumatology Care: A Disease-Specific Guide. J Clin Rheumatol 2024; 30:159-167. [PMID: 38527973 DOI: 10.1097/rhu.0000000000002062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
ABSTRACT Young adults with childhood-onset rheumatic diseases are more frequently establishing and continuing care with adult rheumatologists. The transfer of care can be challenging for both the young adult patients and their adult rheumatologists, in large part due to differences between pediatric-onset rheumatic diseases and their adult-onset counterparts, or due to the rarity of some pediatric-onset rheumatic conditions. Other challenges are due to cultural differences between pediatric and adult medical care and to the young adult needing to increasingly perform self-management skills that were previously managed by parents or other caregivers. In this review, we will provide a summary of strategies for working effectively with young adults as they transition to adult care. We will then discuss a subset of childhood-onset rheumatic diseases-including juvenile idiopathic arthritis, localized scleroderma, autoinflammatory diseases, pediatric-onset systemic lupus erythematosus, juvenile-onset dermatomyositis, and autoimmune encephalitis-for which clinical manifestations, management, and prognosis frequently differ between pediatric onset and adult onset. Our aim is to highlight differences that make caring for this population of transitioning young adults unique, providing tools and knowledge to empower the adult rheumatologist to care for these young adults in ways that are evidence-based, effective, efficient, and rewarding.
Collapse
Affiliation(s)
| | - Joyce C Chang
- Boston Children's Hospital and Harvard Medical School, Boston, MA
| | | |
Collapse
|
10
|
Yang L, Liang Y, Pu J, Cai L, Gao R, Han F, Chang K, Pan S, Wu Z, Zhang Y, Wang Y, Song J, Wu H, Tang J, Wang X. Dysregulated serum lipid profile is associated with inflammation and disease activity in primary Sjögren's syndrome: a retrospective study in China. Immunol Lett 2024; 267:106865. [PMID: 38705483 DOI: 10.1016/j.imlet.2024.106865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 04/23/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
PURPOSE To investigate the relationship between the lipid profiles of patients with primary Sjögren's syndrome (pSS) and other clinical characteristics, laboratory examination, disease activity, and inflammatory factors. In addition, the risk factors for hyperlipidemia-related complications of pSS and the effect of hydroxychloroquine (HCQ) usage on the lipid profile were incorporated into this study. METHODS This is a single-center, retrospective study that included 367 patients who were diagnosed with pSS at Tongji Hospital, School of Medicine, Tongji University, China from January 2010 to March 2022. Initially, demographic information, clinical characteristics, medication records, and complications of the patients were gathered. A case-control analysis compared the 12 systems involvement (ESSDAI domain), clinical symptoms, and laboratory tests between pSS patients with and without dyslipidemia. A simple linear regression model was employed to investigate the relationship between serum lipid profile and inflammatory factors. Logistics regression analysis was performed to assess variables for hyperlipidemia-related complications of pSS. The paired t-test was then used to evaluate the improvement in lipid profile among pSS patients. RESULTS 48.7 % of all pSS patients had dyslipidemia, and alterations in lipid levels were related to gender, age, and smoking status but not body mass index (BMI). Dyslipidemia is more prevalent in pSS patients who exhibit heightened autoimmunity and elevated levels of inflammation. Higher concentrations of multiple highly inflammatory factors correlate with a more severe form of dyslipidemia. Non-traditional cardiovascular risk factors may contribute to hyperlipidemia-related complications of pSS, such as increased, low complement 3 (C3) and low C4. According to our study, HCQ usage may protect against lipid-related disease in pSS. CONCLUSION Attention should be paid to the dyslipidemia of pSS. This research aims to clarify the population portrait of pSS patients with abnormal lipid profiles and provides insights into the correlation between metabolism and inflammation in individuals with pSS and the potential role they play in the advancement of the disease. These findings provide novel avenues for further understanding the underlying mechanisms of pSS pathogenesis.
Collapse
Affiliation(s)
- Lufei Yang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Yuanyuan Liang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Jincheng Pu
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Li Cai
- Department of Science and Research, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Ronglin Gao
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Fang Han
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Keni Chang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Shengnan Pan
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Zhenzhen Wu
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Youwei Zhang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Yanqing Wang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Jiamin Song
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Huihong Wu
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Jianping Tang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China..
| | - Xuan Wang
- Department of Rheumatology and Immunology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China..
| |
Collapse
|
11
|
Ambler WG, Kaplan MJ. Vascular damage in systemic lupus erythematosus. Nat Rev Nephrol 2024; 20:251-265. [PMID: 38172627 PMCID: PMC11391830 DOI: 10.1038/s41581-023-00797-8] [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] [Accepted: 11/29/2023] [Indexed: 01/05/2024]
Abstract
Vascular disease is a major cause of morbidity and mortality in patients with systemic autoimmune diseases, particularly systemic lupus erythematosus (SLE). Although comorbid cardiovascular risk factors are frequently present in patients with SLE, they do not explain the high burden of premature vascular disease. Profound innate and adaptive immune dysregulation seems to be the primary driver of accelerated vascular damage in SLE. In particular, evidence suggests that dysregulation of type 1 interferon (IFN-I) and aberrant neutrophils have key roles in the pathogenesis of vascular damage. IFN-I promotes endothelial dysfunction directly via effects on endothelial cells and indirectly via priming of immune cells that contribute to vascular damage. SLE neutrophils are vasculopathic in part because of their increased ability to form immunostimulatory neutrophil extracellular traps. Despite improvements in clinical care, cardiovascular disease remains the leading cause of mortality among patients with SLE, and treatments that improve vascular outcomes are urgently needed. Improved understanding of the mechanisms of vascular injury in inflammatory conditions such as SLE could also have implications for common cardiovascular diseases, such as atherosclerosis and hypertension, and may ultimately lead to personalized therapeutic approaches to the prevention and treatment of this potentially fatal complication.
Collapse
Affiliation(s)
- William G Ambler
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
12
|
Hsu UH, Lin YT, Chiang BL. The characteristics and risk factors of cerebrovascular events in young systemic lupus erythematosus patients: A case-control study. J Formos Med Assoc 2024; 123:478-486. [PMID: 37813767 DOI: 10.1016/j.jfma.2023.09.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/10/2023] [Accepted: 09/28/2023] [Indexed: 10/11/2023] Open
Abstract
OBJECTIVES We clarified the characteristics and risk factors of CVEs in young SLE patients. METHOD We retrospectively reviewed the medical records of patients younger than 50 years of age diagnosed with SLE and first CVEs from 1995 to 2020 in a tertiary medical center in Taiwan. We collected data on the patient characteristics before the CVE and reviewed the laboratory data obtained during the period. At a ratio of 1:3, cases and controls were matched with sex, SLE diagnosis age, diagnosis year, and SLE duration. RESULTS We enrolled 43 CVE SLE patients and matched 129 non-CVE SLE controls. The median age at the time of the CVE was 39 years. Around 70% of young-aged CVE involved the cerebral lobes of frontal (∼30%), parietal (∼20%), occipital (∼10%), and temporal (∼10%). The peak incidence period for hemorrhagic CVE was within 1st year of SLE diagnosis (37%); in contrast, during the 2nd to 5th year of SLE diagnosis (25%) for ischemia CVEs. Hyperlipidemia (odds ratio [OR] = 19.36, p = 0.002), anti-phospholipid syndrome (APS) (OR = 41.9, p = 0.0068), a lower hemoglobin level (OR = 0.66, p = 0.0192), and a higher SLE Disease Activity Index (SLEDAI-2k) score (OR = 1.22, p = 0.0019) were independent risk factors for CVEs in young SLE patients. CONCLUSION Hyperlipidemia, APS, low Hb level, and high SLEDAI-2k significantly increase the risk of young-aged SLE patients developing CVE.
Collapse
Affiliation(s)
- Uei-Hsiang Hsu
- Department of Pediatrics, National Taiwan University Hospital Hsin-Chu Branch, Taiwan
| | - Yu-Tsan Lin
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Bor-Luen Chiang
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
13
|
Yu Y, Lu C, Yu W, Lei Y, Sun S, Liu P, Bai F, Chen Y, Chen J. B Cells Dynamic in Aging and the Implications of Nutritional Regulation. Nutrients 2024; 16:487. [PMID: 38398810 PMCID: PMC10893126 DOI: 10.3390/nu16040487] [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: 01/07/2024] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Aging negatively affects B cell production, resulting in a decrease in B-1 and B-2 cells and impaired antibody responses. Age-related B cell subsets contribute to inflammation. Investigating age-related alterations in the B-cell pool and developing targeted therapies are crucial for combating autoimmune diseases in the elderly. Additionally, optimal nutrition, including carbohydrates, amino acids, vitamins, and especially lipids, play a vital role in supporting immune function and mitigating the age-related decline in B cell activity. Research on the influence of lipids on B cells shows promise for improving autoimmune diseases. Understanding the aging B-cell pool and considering nutritional interventions can inform strategies for promoting healthy aging and reducing the age-related disease burden.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Juan Chen
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100091, China; (Y.Y.)
| |
Collapse
|
14
|
van Leuven SI, Major AS. Accelerated atherosclerosis in systemic lupus erythematosus: don't forget the devil we know! Rheumatology (Oxford) 2024; 63:3-5. [PMID: 37421390 PMCID: PMC10765142 DOI: 10.1093/rheumatology/kead329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/10/2023] Open
Affiliation(s)
- Sander I van Leuven
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Amy S Major
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt Medical Center, Nashville, TN, USA
- U.S. Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
| |
Collapse
|
15
|
Lanzolla G, Comi S, Cosentino G, Pakdel F, Marinò M. Statins in Graves Orbitopathy: A New Therapeutic Tool. Ophthalmic Plast Reconstr Surg 2023; 39:S29-S39. [PMID: 38054983 DOI: 10.1097/iop.0000000000002525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
PURPOSE Graves orbitopathy (GO) is the most common extrathyroidal manifestation of Graves disease. Although its pathogenesis is not fully elucidated, GO is commonly considered an autoimmune disease due to loss of self-tolerance against autoantigens shared by thyroid epithelial cells and orbital fibroblasts. High-dose intravenous glucocorticoids (ivGCs) are the most used treatment for moderate-to-severe, active GO, but the addition of other immunomodulating treatments can improve the efficacy of ivGCs. Among the various risk factors that can affect the occurrence of GO, cholesterol may be worthy of interest. Since 2015 the role of cholesterol and cholesterol-lowering medications has been investigated. The purpose of this review is to discuss this topic, thereby offering new therapeutic opportunities for patients with GO. METHODS We searched PubMed for studies published between January 1, 1980 and June 1, 2023, using the search terms "Graves orbitopathy," "thyroid eye disease," "Graves ophthalmopathy," "thyroid ophthalmopathy," "thyroid-associated ophthalmopathy," "endocrine ophthalmopathy," "cholesterol," "lipids," "statins," "low-density lipoprotein," "atorvastatin," and "cholesterol-lowering drugs." Only English-language articles were included. RESULTS A correlation between low-density lipoprotein cholesterol and the risk of GO development has been reported. Furthermore, low-density lipoprotein cholesterol has been proposed as a risk factor that can affect the course of GO and the response to ivGCs. The protective role of cholesterol-lowering medications in preventing GO has been also investigated. Statin treatment was found to have potential benefits in reducing the risk of GO in patients with Graves disease. Given these findings, measurement of low-density lipoprotein cholesterol and treatment of hypercholesterolemia in patients with moderate-to-severe, active GO may be considered before starting ivGCs administration. Recently, a randomized clinical trial aimed at investigating the effects of statins in GO suggested that the addition of oral atorvastatin to ivGCs improves the overall outcome of moderate-to-severe, active GO in hypercholesterolemic patients given ivGCs. CONCLUSIONS Overall, statins seem to have a preventive and therapeutic role in moderate-to-severe active GO. Their efficacy can be related to cholesterol-lowering activity, pleiotropic actions, and interaction with methylprednisolone.
Collapse
Affiliation(s)
- Giulia Lanzolla
- Department of Clinical and Experimental Medicine, Endocrinology Unit II, University of Pisa and University Hospital of Pisa, Pisa, Italy
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, U.S.A
| | - Simone Comi
- Department of Clinical and Experimental Medicine, Endocrinology Unit II, University of Pisa and University Hospital of Pisa, Pisa, Italy
| | - Giada Cosentino
- Department of Clinical and Experimental Medicine, Endocrinology Unit II, University of Pisa and University Hospital of Pisa, Pisa, Italy
| | - Farzad Pakdel
- Department of Ophthalmic Plastic and Reconstructive Surgery, Farabi Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Michele Marinò
- Department of Clinical and Experimental Medicine, Endocrinology Unit II, University of Pisa and University Hospital of Pisa, Pisa, Italy
| |
Collapse
|
16
|
Karakasis P, Patoulias D, Stachteas P, Lefkou E, Dimitroulas T, Fragakis N. Accelerated Atherosclerosis and Management of Cardiovascular Risk in Autoimmune Rheumatic Diseases: An Updated Review. Curr Probl Cardiol 2023; 48:101999. [PMID: 37506959 DOI: 10.1016/j.cpcardiol.2023.101999] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 07/24/2023] [Indexed: 07/30/2023]
Abstract
Even though diagnosis and management pathways have been substantially improved over the last years, autoimmune rheumatic diseases (AIRDs) such as rheumatoid arthritis, systemic sclerosis, systemic lupus erythematosus, antiphospholipid syndrome, Sjögren's syndrome, and systemic vasculitides have been linked to elevated rates of cardiovascular morbidity and mortality, primarily secondary to accelerated atherosclerosis. This phenomenon can be partially attributed to the presence of established cardiovascular risk factors but may also be a result of other inflammatory and autoimmune mechanisms that are enhanced in AIRDs. According to the current guidelines, the recommendations regarding cardiovascular disease prevention in patients with AIRDs are not significantly different from those applied to the general population. Herein, we present a review of the current literature on the risk of accelerated atherosclerosis in AIRDs and provide a summary of available recommendations for the management of cardiovascular risk in rheumatic diseases.
Collapse
Affiliation(s)
- Paschalis Karakasis
- Second Department of Cardiology, Aristotle University of Thessaloniki, General Hospital "Hippokration," Thessaloniki, Greece.
| | - Dimitrios Patoulias
- Second Department of Cardiology, Aristotle University of Thessaloniki, General Hospital "Hippokration," Thessaloniki, Greece; Outpatient Department of Cardiometabolic Medicine, Second Department of Cardiology, Aristotle University of Thessaloniki, General Hospital "Hippokration," Thessaloniki, Greece; Second Department of Internal Medicine, European Interbalkan Medical Center, Thessaloniki, Greece
| | - Panagiotis Stachteas
- Second Department of Cardiology, Aristotle University of Thessaloniki, General Hospital "Hippokration," Thessaloniki, Greece
| | - Eleftheria Lefkou
- Second Department of Cardiology, Aristotle University of Thessaloniki, General Hospital "Hippokration," Thessaloniki, Greece; Perigenesis, Institute of Obstetric Haematology, Thessaloniki, Greece
| | - Theodoros Dimitroulas
- Second Department of Cardiology, Aristotle University of Thessaloniki, General Hospital "Hippokration," Thessaloniki, Greece; Fourth Department of Internal Medicine, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Fragakis
- Second Department of Cardiology, Aristotle University of Thessaloniki, General Hospital "Hippokration," Thessaloniki, Greece
| |
Collapse
|
17
|
Straub RH, Pongratz G, Buttgereit F, Gaber T. [Energy metabolism of the immune system : Consequences in chronic inflammation]. Z Rheumatol 2023:10.1007/s00393-023-01389-4. [PMID: 37488246 DOI: 10.1007/s00393-023-01389-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2023] [Indexed: 07/26/2023]
Abstract
BACKGROUND Energy is the currency of life. The systemic and intracellular energy metabolism plays an essential role for the energy supply of the resting and activated immune system and this also applies to chronic inflammatory diseases. OBJECTIVE This presentation examines both components of the systemic and cellular energy metabolism in health and chronic inflammation. MATERIAL AND METHODS A literature search was conducted using PubMed, Embase and the Cochrane Library. The information is presented in the form of a narrative review. RESULTS A chronically activated immune system acquires large amounts of energy-rich substrates that are lost for other functions of the body. In particular, the immune system and the brain are in competition. The consequences of this competition are many known diseases, such as fatigue, anxiety, depression, anorexia, sleep problems, sarcopenia, osteoporosis, insulin resistance, hypertension and others. The permanent change in the brain causes long-term alterations that stimulate disease sequelae even after disease remission. In the intracellular energy supply, chronic inflammation typically involves a conversion to glycolysis (to lactate, which has its own regulatory functions) and the pentose phosphate pathway in disorders of mitochondrial function. The chronic changes in immune cells of patients with rheumatoid arthritis (RA) lead to a disruption of the citric acid cycle (Krebs cycle). The hypoxic situation in the inflamed tissue stimulates many alterations. A differentiation is made between effector functions and regulatory functions of immune cells. CONCLUSION Based on the energy changes mentioned, novel treatment suggestions can be made in addition to those already known in energy metabolism.
Collapse
Affiliation(s)
- Rainer H Straub
- Labor für Experimentelle Rheumatologie und Neuroendokrin-Immunologie, Klinik und Poliklinik für Innere Medizin I, Universitätsklinikum Regensburg, 93042, Regensburg, Deutschland.
| | - Georg Pongratz
- Abteilung für Rheumatologie, Klinik für Gastroenterologie, Krankenhaus Barmherzige Brüder Regensburg, 93049, Regensburg, Deutschland
| | - Frank Buttgereit
- Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Charité - Universitätsmedizin Berlin, Freie Universität Berlin und Humboldt-Universität zu Berlin, Berlin, Deutschland
| | - Timo Gaber
- Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Charité - Universitätsmedizin Berlin, Freie Universität Berlin und Humboldt-Universität zu Berlin, Berlin, Deutschland
| |
Collapse
|
18
|
Hurst C, Soto M, Vina ER, Rodgers KE. Renin-Angiotensin System-Modifying Antihypertensive Drugs Can Reduce the Risk of Cardiovascular Complications in Lupus: A Retrospective Cohort Study. Am J Med 2023; 136:284-293.e4. [PMID: 36495935 PMCID: PMC9957968 DOI: 10.1016/j.amjmed.2022.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Patients with systemic lupus erythematosus have a higher incidence of cardiovascular disease than the general population. Antihypertensive drugs that modify the renin-angiotensin system (RAS) are used to protect renal function in lupus nephritis and may also have extrarenal effects that lower cardiovascular disease risk due to their anti-inflammatory properties. In this study, we compared the effects of RAS vs non-RAS antihypertensive drugs on cardiovascular disease incidence in patients with lupus. METHODS Using a medical insurance claims dataset, 220,168 patients with lupus were identified, of which 31,647 patients (4018 patients prescribed RAS drugs, 27,629 patients prescribed non-RAS drugs) were eligible for the study. Patients had a mean age of 46.1 years, were 93.0% female, and healthy (96.9% Charlson Comorbidity Index score 0-4). Patients in the 2 drug groups were propensity score matched using demographic data, risk factors, and comorbidities. RESULTS Use of RAS vs non-RAS drugs lowered the relative risk (RR) of diagnosis of cardiovascular disease (RR 0.80; 95% confidence interval [CI], 0.74-0.87), which was more pronounced after propensity score matching (RR 0.62; 95% CI, 0.57-0.68). The decreased risk in cardiovascular disease occurred regardless of lupus nephritis status (with lupus nephritis: RR 0.51; 95% CI, 0.39-0.65; without lupus nephritis: RR 0.65; 95% CI, 0.59-0.72). RAS-modifying therapies significantly increased cardiovascular disease-free survival probability over a 5-year period (86.0% vs 78.3% probability). CONCLUSIONS RAS-modifying drugs reduced the risk of cardiovascular disease in patients with systemic lupus erythematosus in this dataset. These findings have the potential to impact clinical decision-making with regards to hypertension management in patients with lupus.
Collapse
Affiliation(s)
- Chelsie Hurst
- Department of Pharmacology, Center for Innovation in Brain Science, College of Medicine, University of Arizona, Tucson
| | - Maira Soto
- Department of Pharmacology, Center for Innovation in Brain Science, College of Medicine, University of Arizona, Tucson
| | - Ernest R Vina
- Section of Rheumatology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pa
| | - Kathleen E Rodgers
- Department of Pharmacology, Center for Innovation in Brain Science, College of Medicine, University of Arizona, Tucson.
| |
Collapse
|
19
|
Jiang L, Wei W, Kang S, Li XL, Luo Y. Insights into lipid metabolism and immune-inflammatory responses in the pathogenesis of coronary artery ectasia. Front Physiol 2023; 14:1096991. [PMID: 36760522 PMCID: PMC9905697 DOI: 10.3389/fphys.2023.1096991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 01/17/2023] [Indexed: 01/27/2023] Open
Abstract
Coronary artery ectasia (CAE) is a rare finding that is associated with poor clinical outcomes (Kawsara et al. 2018), and disorders in lipid metabolism have been reported in CAE. Lipids constitute one of the three metabolite types that regulate bodily functions and are also powerful signaling molecules (Han 2016; Zhu et al. 2021) that affect immunoregulation and inflammatory responses via a series of transcription factors and signaling pathways (Barrera et al. 2013). Although abnormal lipid metabolism and immunoinflammatory responses have been reported in CAE, their roles in the pathogenic mechanisms underlying CAE are currently unclear.
Collapse
Affiliation(s)
- Li Jiang
- Department of Cardiovascular Medicine, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wei Wei
- Department of Cardiovascular Medicine, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Sheng Kang
- Department of Cardiovascular Medicine, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiao-Lin Li
- Department of Cardiovascular Medicine, Jian East Hospital, Jinggangshan University School of Medicine, Jiangxi, China
| | - Yu Luo
- Department of Cardiovascular Medicine, East Hospital, Tongji University School of Medicine, Shanghai, China,*Correspondence: Yu Luo,
| |
Collapse
|
20
|
Lin GL, Lin HC, Lin HL, Keller JJ, Wang LH. Association between statin use and the risk of gout in patients with hyperlipidemia: A population-based cohort study. Front Pharmacol 2023; 14:1096999. [PMID: 36873987 PMCID: PMC9975165 DOI: 10.3389/fphar.2023.1096999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Objective: To investigate the association between statin use and risk of gout in patients with hyperlipidemia. Methods: In this population-based retrospective cohort study, patients ≥20 years and diagnosed as having incident hyperlipidemia between 2001 and 2012 were identified from the 2000 Longitudinal Generation Tracking Database in Taiwan. Regular statin users (incident statin use, having 2 times and ≥90 days of prescription for the first year) and two active comparators [irregular statin use and other lipid-lowering agent (OLLA) use] were compared; the patients were followed up until the end of 2017. Propensity score matching was applied to balance potential confounders. Time-to-event outcomes of gout and dose- and duration-related associations were estimated using marginal Cox proportional hazard models. Results: Regular statin use non-significantly reduced gout risk compared with irregular statin use (aHR, 0.95; 95% CI, 0.90-1.01) and OLLA use (aHR, 0.94; 95% CI, 0.84-1.04). However, a protective effect was noted for a cumulative defined daily dose (cDDD) of >720 (aHR, 0.57; 95% CI, 0.47-0.69 compared with irregular statin use and aHR, 0.48; 95% CI, 0.34-0.67 compared with OLLA use) or a therapy duration of >3 years (aHR, 0.76; 95% CI, 0.64-0.90 compared with irregular statin use and aHR, 0.50; 95% CI, 0.37-0.68 compared with OLLA use). Dose- and duration-dependent associations were consistent in the 5-year sensitivity analyses. Conclusion: Although statin use was not associated with a reduction in gout risk, the protective benefit was observed in those receiving higher cumulative doses or with a longer therapy duration.
Collapse
Affiliation(s)
- Guan-Ling Lin
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Hsiu-Chen Lin
- Department of Pediatrics, School of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Clinical Pathology, Taipei Medical University Hospital, Taipei, Taiwan
| | - Hsiu-Li Lin
- Department of Neurology, General Cathay Hospital, Sijhih Branch, New Taipei City, Taiwan
| | - Joseph Jordan Keller
- Department of Psychiatry, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Li-Hsuan Wang
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.,Department of Pharmacy, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
21
|
Kim HI, Kim TH, Kim H, Kim SW, Hahm JR, Chung JH. Dyslipidemia Is a Risk Factor for Hypothyroidism in Women: A Longitudinal Cohort Study from South Korea. Thyroid 2023; 33:100-108. [PMID: 36602760 DOI: 10.1089/thy.2022.0216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Background: Hypothyroidism is a risk factor for dyslipidemia. We explored whether dyslipidemia is a risk factor for hypothyroidism. Methods: We performed a retrospective analysis of data from a longitudinal cohort study of South Korean adults who underwent medical examination and ≥4 biochemical assessments of thyroid function. The primary outcome was hypothyroidism (thyrotropin [TSH] >4.2 mU/L), and the secondary outcome was severe subclinical hypothyroidism (SCH; TSH ≥10.0 mU/L and normal free thyroxine [fT4] level) or overt hypothyroidism (OH; total triiodothyronine <80 ng/dL and/or fT4 < 0.93 ng/dL and high TSH values). The association of baseline dyslipidemia status with subsequent hypothyroidism was evaluated using Kaplan-Meier curves with the log-rank test and Cox proportional hazards regression models (for the entire population and respective genders). Subgroup analyses according to age (<40 and ≥40 years) and body-mass index (BMI; <23, 23-25, and ≥25 kg/m2) were performed according to gender. Results: We included 1665 participants. During a median follow-up period of 61.0 months, 24.3% (404/1665) individuals developed hypothyroidism. Among these, 36 participants (2.1%) had severe SCH or OH. Excluding patients with a first abnormal TSH level at last follow-up, 44.5% (126/283) of the patients with hypothyroidism had spontaneous TSH normalization. In respective multivariate analyses, dyslipidemia at baseline was independently associated with development of hypothyroidism in women (adjusted hazard ratio [HR] = 2.05 [1.31-3.19], p = 0.002), but not in men (adjusted HR = 1.00 [0.77-1.30], p = 0.991). In women, the presence of dyslipidemia at baseline was associated with development of severe SCH or OH (adjusted HR = 5.33 [1.41-20.12], p = 0.014). In women, respective associations according to age and BMI were as follows: age <40 years, adjusted HR = 2.90 (1.34-6.26, p = 0.007); age ≥40 years, adjusted HR = 1.85 (1.08-3.14, p = 0.023); BMI <23 kg/m2, adjusted HR = 1.68 (0.82-3.43, p = 0.151); BMI = 23-25 kg/m2, adjusted HR = 2.17 (0.93-5.07, p = 0.071); and BMI ≥25 kg/m2, adjusted HR = 2.82 (1.16-6.86, p = 0.022). Conclusions: In Korean adults, dyslipidemia was associated with development of hypothyroidism in women. Our findings require confirmation.
Collapse
Affiliation(s)
- Hye In Kim
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea
- Department of Medicine, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Tae Hyuk Kim
- Division of Endocrinology and Metabolism, Department of Medicine, Thyroid Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hosu Kim
- Division of Endocrinology, Department of Medicine, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon, Republic of Korea
| | - Sun Wook Kim
- Division of Endocrinology and Metabolism, Department of Medicine, Thyroid Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jong Ryeal Hahm
- College of Medicine, Gyeongsang National University, Institute of Health Sciences, Jinju, Republic of Korea
- Department of Internal Medicine, Gyeongsang National University Hospital, Jinju, Republic of Korea
| | - Jae Hoon Chung
- Division of Endocrinology and Metabolism, Department of Medicine, Thyroid Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
22
|
Richter P, Cardoneanu A, Rezus C, Burlui AM, Rezus E. Non-Traditional Pro-Inflammatory and Pro-Atherosclerotic Risk Factors Related to Systemic Lupus Erythematosus. Int J Mol Sci 2022; 23:ijms232012604. [PMID: 36293458 PMCID: PMC9604037 DOI: 10.3390/ijms232012604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/16/2022] [Accepted: 10/17/2022] [Indexed: 11/23/2022] Open
Abstract
Cardiovascular diseases (CVD) are one of the leading causes of high mortality in patients with systemic lupus erythematosus (SLE). The Framingham risk score and other traditional risk factors do not fully reflect the CVD risk in SLE patients. Therefore, in order to stratify these high-risk patients, additional biomarkers for subclinical CVD are needed. The mechanisms of atherogenesis in SLE are still being investigated. During the past decades, many reports recognized that inflammation plays a crucial role in the development of atherosclerosis. The aim of this report is to present novel proinflammatory and pro-atherosclerotic risk factors that are closely related to SLE inflammation and which determine an increased risk for the occurrence of early cardiovascular events.
Collapse
Affiliation(s)
- Patricia Richter
- Department of Rheumatology, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Anca Cardoneanu
- Department of Rheumatology, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
- Correspondence: (A.C.); (C.R.); Tel.: +40232301615 (A.C. & C.R.)
| | - Ciprian Rezus
- Department of Internal Medicine, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania
- “Sfantul Spiridon” Emergency Hospital, 700111 Iasi, Romania
- Correspondence: (A.C.); (C.R.); Tel.: +40232301615 (A.C. & C.R.)
| | - Alexandra Maria Burlui
- Department of Rheumatology, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Elena Rezus
- Department of Rheumatology, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| |
Collapse
|
23
|
Vyletelová V, Nováková M, Pašková Ľ. Alterations of HDL's to piHDL's Proteome in Patients with Chronic Inflammatory Diseases, and HDL-Targeted Therapies. Pharmaceuticals (Basel) 2022; 15:1278. [PMID: 36297390 PMCID: PMC9611871 DOI: 10.3390/ph15101278] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/03/2022] [Accepted: 10/14/2022] [Indexed: 09/10/2023] Open
Abstract
Chronic inflammatory diseases, such as rheumatoid arthritis, steatohepatitis, periodontitis, chronic kidney disease, and others are associated with an increased risk of atherosclerotic cardiovascular disease, which persists even after accounting for traditional cardiac risk factors. The common factor linking these diseases to accelerated atherosclerosis is chronic systemic low-grade inflammation triggering changes in lipoprotein structure and metabolism. HDL, an independent marker of cardiovascular risk, is a lipoprotein particle with numerous important anti-atherogenic properties. Besides the essential role in reverse cholesterol transport, HDL possesses antioxidative, anti-inflammatory, antiapoptotic, and antithrombotic properties. Inflammation and inflammation-associated pathologies can cause modifications in HDL's proteome and lipidome, transforming HDL from atheroprotective into a pro-atherosclerotic lipoprotein. Therefore, a simple increase in HDL concentration in patients with inflammatory diseases has not led to the desired anti-atherogenic outcome. In this review, the functions of individual protein components of HDL, rendering them either anti-inflammatory or pro-inflammatory are described in detail. Alterations of HDL proteome (such as replacing atheroprotective proteins by pro-inflammatory proteins, or posttranslational modifications) in patients with chronic inflammatory diseases and their impact on cardiovascular health are discussed. Finally, molecular, and clinical aspects of HDL-targeted therapies, including those used in therapeutical practice, drugs in clinical trials, and experimental drugs are comprehensively summarised.
Collapse
Affiliation(s)
| | | | - Ľudmila Pašková
- Department of Cell and Molecular Biology of Drugs, Faculty of Pharmacy, Comenius University, 83232 Bratislava, Slovakia
| |
Collapse
|
24
|
Nowak MM, Niemczyk M, Florczyk M, Kurzyna M, Pączek L. Effect of Statins on All-Cause Mortality in Adults: A Systematic Review and Meta-Analysis of Propensity Score-Matched Studies. J Clin Med 2022; 11:jcm11195643. [PMID: 36233511 PMCID: PMC9572734 DOI: 10.3390/jcm11195643] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/25/2022] Open
Abstract
Statins are lipid-lowering medications used for the prevention of cardiovascular disease (CVD), but the pleiotropic effects of statins might be beneficial in other chronic diseases. This meta-analysis investigated the association between statin use and mortality in different chronic conditions. Eligible studies were real-world studies that compared all-cause mortality over at least 12 months between propensity score-matched statin users and non-users. Overall, 54 studies were included: 21 in CVD, 6 in chronic kidney disease, 6 in chronic inflammatory diseases, 3 in cancer, and 18 in other diseases. The risk of all-cause mortality was significantly reduced in statin users (hazard ratio: 0.72, 95% confidence interval: 0.66−0.76). The reduction in mortality risk was similar in CVD studies (0.73, 0.66−0.76) and non-CVD studies (0.70, 0.67−0.79). There were no significant differences in the risk reduction between cohorts with different diseases (p = 0.179). The greatest mortality reduction was seen in studies from Asia (0.61, 0.61−0.73) and the lowest in studies from North America (0.78, 0.73−0.83) and Australia (0.78, 0.62−0.97). There was a significant heterogeneity (I2 = 95%, tau2 = 0.029, p < 0.01). In conclusion, statin use was associated with a significantly reduced risk of all-cause mortality in real-world cohorts with CVD and non-CVD.
Collapse
Affiliation(s)
- Marcin M. Nowak
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology at the European Health Center, 05-400 Otwock, Poland
- Correspondence:
| | - Mariusz Niemczyk
- Department of Immunology, Transplant Medicine and Internal Diseases, Medical University of Warsaw, 02-006 Warsaw, Poland
| | - Michał Florczyk
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology at the European Health Center, 05-400 Otwock, Poland
| | - Marcin Kurzyna
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology at the European Health Center, 05-400 Otwock, Poland
| | - Leszek Pączek
- Department of Immunology, Transplant Medicine and Internal Diseases, Medical University of Warsaw, 02-006 Warsaw, Poland
| |
Collapse
|
25
|
Liu Y, Yu X, Zhang W, Zhang X, Wang M, Ji F. Mechanistic insight into premature atherosclerosis and cardiovascular complications in systemic lupus erythematosus. J Autoimmun 2022; 132:102863. [PMID: 35853760 DOI: 10.1016/j.jaut.2022.102863] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/02/2022] [Indexed: 12/12/2022]
Abstract
Systemic lupus erythematosus (SLE) is associated with a significant risk of cardiovascular disease (CVD), which substantially increases disease mortality and morbidity. The overall mechanisms associated with the development of premature atherosclerosis and CVD in SLE remain unclear, but has been considered as a result of an intricate interplay between the profound immune dysregulation and traditional CVD risk factors. Aberrant systemic inflammation in SLE may lead to an abnormal lipid profile and dysfunction, which can further fuel the pro-atherosclerotic environment. The existence of a strong imbalance between endothelial damage and vascular repair/angiogenesis promotes vascular injury, which is the early step in the progression of atherosclerotic CVD. Profound innate and adaptive immune dysregulation, characterized by excessive type I interferon burden, aberrant macrophage, platelet and complements activation, neutrophil dysregulation and neutrophil extracellular traps formation, uncontrolled T cell activation, and excessive autoantibody production and immune complex formation, have been proposed to promote accelerated CVD in SLE. While designing targeted therapies to correct the dysregulated immune activation may be beneficial in the treatment of SLE-related CVD, much additional work is needed to determine how to translate these findings into clinical practice. Additionally, a number of biomarkers display diagnostic potentials in improving CVD risk stratification in SLE, further prospective studies will help understand which biomarker(s) will be the most impactful one(s) in assessing SLE-linked CVD. Continued efforts to identify novel mechanisms and to establish criteria for assessing CVD risk as well as predicting CVD progression are in great need to improve CVD outcomes in SLE.
Collapse
Affiliation(s)
- Yudong Liu
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, PR China; The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, PR China
| | - Xue Yu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, PR China
| | - Wenduo Zhang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, PR China
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, PR China
| | - Min Wang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, PR China
| | - Fusui Ji
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, PR China.
| |
Collapse
|
26
|
Alghareeb R, Hussain A, Maheshwari MV, Khalid N, Patel PD. Cardiovascular Complications in Systemic Lupus Erythematosus. Cureus 2022; 14:e26671. [PMID: 35949751 PMCID: PMC9358056 DOI: 10.7759/cureus.26671] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2022] [Indexed: 12/14/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an auto-immune disease of a relapsing-remitting nature that can cause multiorgan damage depending on several factors, mainly the disease activity. Young age women are the most likely to be affected by the disease and the female-to-male prevalence ratio is approximately 1:10. As the number of SLE patients has been increasing in the last few decades, the annual number of deaths due to the disease and its complications has increased as well, and one of the most important systems to which high mortality is attributed is the cardiovascular system, leading to premature atherosclerosis and other events such as endocarditis and valve disease. In addition to the classical cardiovascular risk factors, studies have found a positive correlation between SLE and other cardio-harmful diseases such as metabolic syndrome and dyslipidemia. Moreover, some of the medications used in the treatment of SLE place a heavy burden on the heart. The article reviews the shared pathophysiology of SLE and cardiovascular disease along with the most common SLE- associated cardiac risks, events, and management.
Collapse
|
27
|
Oliveira CB, Kaplan MJ. Cardiovascular disease risk and pathogenesis in systemic lupus erythematosus. Semin Immunopathol 2022; 44:309-324. [PMID: 35355124 PMCID: PMC9064999 DOI: 10.1007/s00281-022-00922-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/08/2022] [Indexed: 02/06/2023]
Abstract
Systemic lupus erythematosus (SLE) often features extensive cardiovascular (CV) comorbidity and patients with SLE are at significantly increased risk of CV event occurrence and CV-related mortality. While the specific mechanisms leading to this increased cardiovascular disease (CVD) risk remain to be fully characterized, this heightened risk cannot be fully explained by traditional CV risk factors and is likely driven by immunologic and inflammatory features of SLE. Widespread innate and adaptive immune dysregulation characterize SLE, and factors including excessive type I interferon burden, inappropriate formation and ineffective clearance of neutrophil extracellular traps, and autoantibody formation have been linked to clinical and metabolic features impacting CV risk in SLE and may represent pathogenic drivers of SLE-related CVD. Indeed, functional and phenotypic aberrations in almost every immune cell type are present in SLE and may impact CVD progression. As understanding of the contribution of SLE-specific factors to CVD in SLE improves, improved screening and monitoring of CV risk alongside development of therapeutic treatments aimed at prevention of CVD in SLE patients are required and remain the focus of several ongoing studies and lines of inquiry.
Collapse
Affiliation(s)
- Christopher B Oliveira
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 10 Center Drive, 12N248C, Bethesda, MD, 20892, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 10 Center Drive, 12N248C, Bethesda, MD, 20892, USA.
| |
Collapse
|
28
|
Oliveira CB, Kaplan MJ. Cardiovascular disease risk and pathogenesis in systemic lupus erythematosus. Semin Immunopathol 2022. [PMID: 35355124 DOI: 10.1007/s00281-02200922-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Systemic lupus erythematosus (SLE) often features extensive cardiovascular (CV) comorbidity and patients with SLE are at significantly increased risk of CV event occurrence and CV-related mortality. While the specific mechanisms leading to this increased cardiovascular disease (CVD) risk remain to be fully characterized, this heightened risk cannot be fully explained by traditional CV risk factors and is likely driven by immunologic and inflammatory features of SLE. Widespread innate and adaptive immune dysregulation characterize SLE, and factors including excessive type I interferon burden, inappropriate formation and ineffective clearance of neutrophil extracellular traps, and autoantibody formation have been linked to clinical and metabolic features impacting CV risk in SLE and may represent pathogenic drivers of SLE-related CVD. Indeed, functional and phenotypic aberrations in almost every immune cell type are present in SLE and may impact CVD progression. As understanding of the contribution of SLE-specific factors to CVD in SLE improves, improved screening and monitoring of CV risk alongside development of therapeutic treatments aimed at prevention of CVD in SLE patients are required and remain the focus of several ongoing studies and lines of inquiry.
Collapse
Affiliation(s)
- Christopher B Oliveira
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 10 Center Drive, 12N248C, Bethesda, MD, 20892, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 10 Center Drive, 12N248C, Bethesda, MD, 20892, USA.
| |
Collapse
|
29
|
Robinson GA, Wilkinson MGL, Wincup C. The Role of Immunometabolism in the Pathogenesis of Systemic Lupus Erythematosus. Front Immunol 2022; 12:806560. [PMID: 35154082 PMCID: PMC8826250 DOI: 10.3389/fimmu.2021.806560] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/27/2021] [Indexed: 12/15/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disorder in which pathogenic abnormalities within both the innate and adaptive immune response have been described. In order to activated, proliferate and maintain this immunological response a drastic upregulation in energy metabolism is required. Recently, a greater understanding of these changes in cellular bioenergetics have provided new insight into the links between immune response and the pathogenesis of a number of diseases, ranging from cancer to diabetes and multiple sclerosis. In this review, we highlight the latest understanding of the role of immunometabolism in SLE with particular focus on the role of abnormal mitochondrial function, lipid metabolism, and mTOR signaling in the immunological phenomenon observed in the SLE. We also consider what implications this has for future therapeutic options in the management of the disease in future.
Collapse
Affiliation(s)
- George Anthony Robinson
- Department of Rheumatology, Division of Medicine, University College London, London, United Kingdom.,Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH) and Great Ormond Street Hospital (GOSH), University College London, London, United Kingdom
| | - Meredyth G Ll Wilkinson
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH) and Great Ormond Street Hospital (GOSH), University College London, London, United Kingdom.,Department of Rheumatology, University College London Great Ormond Street Institute of Child Health, Infection, Immunity and Inflammation Research and Teaching Department, University College London, London, United Kingdom
| | - Chris Wincup
- Department of Rheumatology, Division of Medicine, University College London, London, United Kingdom.,Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH) and Great Ormond Street Hospital (GOSH), University College London, London, United Kingdom
| |
Collapse
|
30
|
Drosos GC, Vedder D, Houben E, Boekel L, Atzeni F, Badreh S, Boumpas DT, Brodin N, Bruce IN, González-Gay MÁ, Jacobsen S, Kerekes G, Marchiori F, Mukhtyar C, Ramos-Casals M, Sattar N, Schreiber K, Sciascia S, Svenungsson E, Szekanecz Z, Tausche AK, Tyndall A, van Halm V, Voskuyl A, Macfarlane GJ, Ward MM, Nurmohamed MT, Tektonidou MG. EULAR recommendations for cardiovascular risk management in rheumatic and musculoskeletal diseases, including systemic lupus erythematosus and antiphospholipid syndrome. Ann Rheum Dis 2022; 81:768-779. [PMID: 35110331 DOI: 10.1136/annrheumdis-2021-221733] [Citation(s) in RCA: 142] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 01/05/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To develop recommendations for cardiovascular risk (CVR) management in gout, vasculitis, systemic sclerosis (SSc), myositis, mixed connective tissue disease (MCTD), Sjögren's syndrome (SS), systemic lupus erythematosus (SLE) and antiphospholipid syndrome (APS). METHODS Following European League against Rheumatism (EULAR) standardised procedures, a multidisciplinary task force formulated recommendations for CVR prediction and management based on systematic literature reviews and expert opinion. RESULTS Four overarching principles emphasising the need of regular screening and management of modifiable CVR factors and patient education were endorsed. Nineteen recommendations (eleven for gout, vasculitis, SSc, MCTD, myositis, SS; eight for SLE, APS) were developed covering three topics: (1) CVR prediction tools; (2) interventions on traditional CVR factors and (3) interventions on disease-related CVR factors. Several statements relied on expert opinion because high-quality evidence was lacking. Use of generic CVR prediction tools is recommended due to lack of validated rheumatic diseases-specific tools. Diuretics should be avoided in gout and beta-blockers in SSc, and a blood pressure target <130/80 mm Hg should be considered in SLE. Lipid management should follow general population guidelines, and antiplatelet use in SLE, APS and large-vessel vasculitis should follow prior EULAR recommendations. A serum uric acid level <0.36 mmol/L (<6 mg/dL) in gout, and disease activity control and glucocorticoid dose minimisation in SLE and vasculitis, are recommended. Hydroxychloroquine is recommended in SLE because it may also reduce CVR, while no particular immunosuppressive treatment in SLE or urate-lowering therapy in gout has been associated with CVR lowering. CONCLUSION These recommendations can guide clinical practice and future research for improving CVR management in rheumatic and musculoskeletal diseases.
Collapse
Affiliation(s)
- George C Drosos
- First Department of Propaedeutic Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Daisy Vedder
- Reade, Amsterdam Rheumatology and Immunology Center, Amsterdam, The Netherlands
| | - Eline Houben
- Department of Internal Medicine, Northwest Clinics, Alkmaar, The Netherlands
| | - Laura Boekel
- Reade, Amsterdam Rheumatology and Immunology Center, Amsterdam, The Netherlands
| | - Fabiola Atzeni
- Rheumatology Unit, Department of Internal Medicine, University of Messina, Messina, Italy
| | - Sara Badreh
- EULAR Patient Research Partner, Brussels, Belgium
| | - Dimitrios T Boumpas
- 4th Department of Internal Medicine, "Attikon" University Hospital, Athens, Greece.,Joint Academic Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nina Brodin
- Division of Physiotherapy, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Huddinge, Sweden.,Department of Orthopaedics, Danderyd Hospital Corp, Stockholm, Sweden
| | - Ian N Bruce
- Centre for Epidemiology Versus Arthritis, Centre for Musculoskeletal Research, University of Manchester, Manchester, UK.,NIHR Manchester Biomedical Research Centre, Manchester University Hospitals NHS Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Miguel Ángel González-Gay
- Rheumatology Division, Hospital Universitario Marqués de Valdecilla and University of Cantabria, Santander, Spain
| | - Søren Jacobsen
- Copenhagen Lupus and Vasculitis Clinic, Rigshospitalet, Copenhagen, Denmark.,Department of Clinical Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - György Kerekes
- Intensive Care Unit, Department of Medicine, University of Debrecen, Debrecen, Hungary
| | | | - Chetan Mukhtyar
- Rheumatology Department, Norfolk and Norwich University Hospital, Colney Lane, UK
| | - Manuel Ramos-Casals
- Department of Autoimmune Diseases, ICMiD, University of Barcelona, Hospital Clínic, Barcelona, Spain
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Karen Schreiber
- EMEUNET member, Danish Hospital for Rheumatic Diseases, Sonderburg, Denmark
| | - Savino Sciascia
- EMEUNET member, CMID-Nephrology, San Giovanni Bosco Hospital, University of Torino, Torino, Italy
| | - Elisabet Svenungsson
- Department of Medicine, Rheumatology Unit, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Zoltan Szekanecz
- Department of Rheumatology, University of Debrecen Faculty of Medicine, Debrecen, Hungary
| | - Anne-Kathrin Tausche
- Department of Rheumatology, University Clinic Carl Gustav Carus at the TU Dresden, Dresden, Germany
| | - Alan Tyndall
- Department of Rheumatology, University of Basel, Basel, Switzerland
| | - Vokko van Halm
- Department of Cardiology, Amsterdam University Medical Center, location VU University medical center, Amsterdam, The Netherlands
| | - Alexandre Voskuyl
- Department of Rheumatology and Clinical Immunology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | | | - Michael M Ward
- Intramural Research Program, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Michael T Nurmohamed
- Reade, Amsterdam Rheumatology and Immunology Center, Amsterdam, The Netherlands.,Amsterdam University Medical Center, location VU University Medical Centre, Amsterdam, The Netherlands
| | - Maria G Tektonidou
- First Department of Propaedeutic Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Athens, Greece .,Joint Academic Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
31
|
Zhu L, Singh M, Lele S, Sahakian L, Grossman J, Hahn B, McMahon M. Assessing the validity of QRISK3 in predicting cardiovascular events in systemic lupus erythematosus. Lupus Sci Med 2022; 9:9/1/e000564. [PMID: 35193947 PMCID: PMC8867320 DOI: 10.1136/lupus-2021-000564] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/19/2021] [Indexed: 11/27/2022]
Abstract
Objectives Traditional cardiovascular risk calculators such as the Framingham Risk Score (FRS) have been shown to underestimate risk in patients with SLE. The QRISK3 calculator is unique in including SLE and corticosteroid use as risk factors. This study aims to assess the validity of QRISK3 compared with other cardiovascular risk models in a cohort of patients with SLE in the USA. Methods We studied a prospective cohort of 366 adult patients with SLE without history of any cardiovascular event and followed them for 10 years. We compared the diagnostic performance of QRISK3 with FRS, modified FRS, Atherosclerotic Cardiovascular Disease (ASCVD), and Predictors of Risk for Elevated Flares, Damage Progression and Increased Cardiovascular Disease in Patients with SLE (PREDICTS). Results Sixty-four of the 366 patients (17.4%) experienced at least one cardiovascular event during the 10-year follow-up period. Of these patients 45% had a QRISK3 score >10%, whereas 20.5% of patients who did not have an event had a QRISK3 score >10% (p<0.001). The corresponding numbers for FRS, modified FRS, ASCVD and PREDICTS were 11.0% vs 7.2% (p=ns), 40.6% vs 28.0% (p=0.05), 12.2% vs 5.9% (p=ns), and 77% vs 32.1% (p<0.001), respectively. The areas under the receiver operating characteristic curve using QRISK3 >10% and high-risk PREDICTS were both larger than those using ASCVD >10%, FRS >10% and modified FRS >10%. Conclusions Both QRISK3 and PREDICTS demonstrated better performance in predicting risk of cardiovascular disease in this cohort of patients with SLE compared with FRS, modified FRS and ASCVD.
Collapse
Affiliation(s)
- Lisa Zhu
- Medicine - Rheumatology, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California, USA
| | - Manpreet Singh
- Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Sonia Lele
- Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Lori Sahakian
- Medicine - Rheumatology, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California, USA
| | - Jennifer Grossman
- Medicine - Rheumatology, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California, USA
| | - Bevra Hahn
- Medicine - Rheumatology, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California, USA
| | - Maureen McMahon
- Medicine - Rheumatology, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California, USA
| |
Collapse
|
32
|
Robinson G, Pineda-Torra I, Ciurtin C, Jury EC. Lipid metabolism in autoimmune rheumatic disease: implications for modern and conventional therapies. J Clin Invest 2022; 132:e148552. [PMID: 35040437 PMCID: PMC8759788 DOI: 10.1172/jci148552] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Suppressing inflammation has been the primary focus of therapies in autoimmune rheumatic diseases (AIRDs), including rheumatoid arthritis and systemic lupus erythematosus. However, conventional therapies with low target specificity can have effects on cell metabolism that are less predictable. A key example is lipid metabolism; current therapies can improve or exacerbate dyslipidemia. Many conventional drugs also require in vivo metabolism for their conversion into therapeutically beneficial products; however, drug metabolism often involves the additional formation of toxic by-products, and rates of drug metabolism can be heterogeneous between patients. New therapeutic technologies and research have highlighted alternative metabolic pathways that can be more specifically targeted to reduce inflammation but also to prevent undesirable off-target metabolic consequences of conventional antiinflammatory therapies. This Review highlights the role of lipid metabolism in inflammation and in the mechanisms of action of AIRD therapeutics. Opportunities for cotherapies targeting lipid metabolism that could reduce immunometabolic complications and potential increased cardiovascular disease risk in patients with AIRDs are discussed.
Collapse
Affiliation(s)
- George Robinson
- Centre for Rheumatology Research
- Centre for Adolescent Rheumatology Research, and
| | - Ines Pineda-Torra
- Centre for Cardiometabolic and Vascular Science, Division of Medicine, University College London, London, United Kingdom
| | - Coziana Ciurtin
- Centre for Rheumatology Research
- Centre for Adolescent Rheumatology Research, and
| | | |
Collapse
|
33
|
Liu J, Song W, Cui D. Relationship between Blood Lipid Profiles and Risk of Lupus Nephritis in Children. Int J Clin Pract 2022; 2022:6130774. [PMID: 36349053 PMCID: PMC9629941 DOI: 10.1155/2022/6130774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is a relatively common rheumatic disease in children. The characteristics of blood lipid metabolism in children with LN are little reported. This study aimed to explore the relationship between blood lipid profiles and the risk of lupus nephritis (LN) in children. METHODS A total of 134 children with newly diagnosed SLE were divided into LN and non-LN groups according to pathological renal biopsy results. Clinical manifestations and blood lipid profiles were analyzed and compared between the two groups, and the relationships between blood lipid profiles and risk of LN were evaluated. RESULTS The positivity rate of an anti-dsDNA antibody and an SLE disease activity index (SLEDAI) were significantly increased, and C3 and C4 levels were significantly reduced in the LN compared with the non-LN group. The overall incidence of dyslipidemia was 79.9%, with a significantly high incidence in the LN group compared with the non-LN group. Total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), and very LDLC (VLDL-C) were all higher in the LN group than those in the non-LN group. However, there was no significant difference in high-density lipoprotein cholesterol (HDL-C) between the two groups. The blood lipid levels were positively correlated with 24-hour urine protein quantification, urea, creatinine, uric acid, urinary IgG, urinary microalbumin, urinary transferrin, urinary α1 microglobulin, and urinary N-acetyl glucosidase, respectively. Receiver-operating characteristic curves showed that combined detection of TC, TG, LDL-C, and VLDL-C had higher discrimination capacity than that in individual measures. Additionally, increased TC was independently associated with the occurrence of LN. CONCLUSIONS Children with LN have significant dyslipidemia. High levels of TC, TG, LDL-C, and VLDL-C are closely related to the occurrence of pLN. Clinical attention should be paid to monitoring and managing blood lipid profiles in children with LN.
Collapse
Affiliation(s)
- Jiajia Liu
- Department of Clinical Laboratory Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Wenqi Song
- Department of Clinical Laboratory Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Dawei Cui
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
34
|
Mazurek A, Gryga K, Bugala K, Iwaniec T, Musial J, Podolec P, Plazak W. Influence of statin therapy on antiphospholipid antibodies and endothelial dysfunction in young patients with coronary artery disease and systemic lupus erythematosus. Arch Med Sci 2022; 18:18-24. [PMID: 35154521 PMCID: PMC8826879 DOI: 10.5114/aoms.2019.90271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 02/10/2019] [Indexed: 01/20/2023] Open
Abstract
INTRODUCTION Antiphospholipid antibodies (aPL) affect atherogenesis and may cause thromboembolism in systemic lupus erythematosus (SLE) and coronary artery disease (CAD). Intensive treatment with statins may reduce inflammation and decrease the number of thrombotic events. That may explain the beneficial effect of statin therapy in SLE and CAD. This study was established to investigate the influence of statin treatment on aPL antibody levels and selected endothelial dysfunction markers in CAD and SLE patients. MATERIAL AND METHODS Fifty-eight patients - 40 after coronary revascularization (age 38.9 (27-46), 35 males) and 18 with clinically stable SLE (age 38.8 (18-62), 1 male) - were enrolled in the study. In both groups intensive atorvastatin treatment was administered. At baseline and after 1 year of follow-up serology tests were performed: anticardiolipin antibodies (aCL), anti-β2 glycoprotein I (aβ2GPI), lupus anticoagulant (LA), C-reactive protein (CRP), soluble form of intracellular adhesion molecule-1 (sICAM-1), vWF:Ag. RESULTS Coronary artery disease patients in 1 year follow-up revealed a decrease of aβ2GPI IgG and CRP. There was a significant increase in aCL IgG, sICAM-1 and vWF:Ag. In SLE patients aPL levels showed no significant reduction after treatment. CONCLUSIONS In clinically stable patients IgM and IgG class aβ2GPI levels are higher in CAD than in SLE, whereas IgG class aCL levels are lower. Statin treatment decreases the CRP level in both SLE and CAD patients, while decreasing the aβ2GPI IgG level only in CAD patients.
Collapse
Affiliation(s)
- Adam Mazurek
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Jagiellonian University Medical College, Krakow, Poland
| | - Krzysztof Gryga
- Department of Internal Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Kamil Bugala
- Department of Diagnostic Medicine, John Paul II Hospital, Jagiellonian University Medical College, Krakow, Poland
| | - Teresa Iwaniec
- Department of Internal Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Jacek Musial
- Department of Internal Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Piotr Podolec
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Jagiellonian University Medical College, Krakow, Poland
| | - Wojciech Plazak
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
35
|
Metabolomics Defines Complex Patterns of Dyslipidaemia in Juvenile-SLE Patients Associated with Inflammation and Potential Cardiovascular Disease Risk. Metabolites 2021; 12:metabo12010003. [PMID: 35050125 PMCID: PMC8779263 DOI: 10.3390/metabo12010003] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 01/21/2023] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of mortality in patients with juvenile-onset systemic lupus erythematosus (JSLE) associated with atherosclerosis. The interplay between dyslipidaemia and inflammation—mechanisms that drive atherosclerosis—were investigated retrospectively in adolescent JSLE patients using lipoprotein-based serum metabolomics in patients with active and inactive disease, compared to healthy controls (HCs). Data was analysed using machine learning, logistic regression, and linear regression. Dyslipidaemia in JSLE patients was characterised by lower levels of small atheroprotective high-density lipoprotein subsets compared to HCs. These changes were exacerbated by active disease and additionally associated with significantly higher atherogenic very-low-density lipoproteins (VLDL) compared to patients with low disease activity. Atherogenic lipoprotein subset expression correlated positively with clinical and serological markers of JSLE disease activity/inflammation and was associated with disturbed liver function, and elevated expression of T-cell and B-cell lipid rafts (cell signalling platforms mediating immune cell activation). Finally, exposing VLDL/LDL from patients with active disease to HC lymphocytes induced a significant increase in lymphocyte lipid raft activation compared to VLDL/LDL from inactive patients. Thus, metabolomic analysis identified complex patterns of atherogenic dyslipidaemia in JSLE patients associated with inflammation. This could inform lipid-targeted therapies in JSLE to improve cardiovascular outcomes.
Collapse
|
36
|
Keyes E, Grinnell M, Jacoby D, Vazquez T, Diaz D, Werth VP, Williams KJ. Assessment and management of the heightened risk for atherosclerotic cardiovascular events in patients with lupus erythematosus or dermatomyositis. Int J Womens Dermatol 2021; 7:560-575. [PMID: 35024413 PMCID: PMC8721062 DOI: 10.1016/j.ijwd.2021.08.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 01/05/2023] Open
Abstract
For patients with lupus erythematosus (LE) or dermatomyositis (DM), there is an urgent need to address a heightened risk of clinical events, chiefly heart attacks and strokes, caused by atherosclerotic cardiovascular disease (ASCVD). Patients with LE or DM frequently exhibit high levels of conventional risk factors for ASCVD events, particularly dyslipoproteinemia and hypertension; an amplified burden of atherosclerotic plaques; and increased age- and sex-adjusted rates of ASCVD events compared with the general population. The rate of ASCVD events exceeds what would be expected from conventional risk factors, suggesting that disease-specific autoimmune processes exacerbate specific, known pathogenic steps in atherosclerosis. Importantly, despite their heightened risk, patients with LE or DM are often undertreated for known causative agents and exacerbators of ASCVD. Herein, we propose an approach to assess and manage the heightened risk of ASCVD events in patients with LE or DM. Our approach is modeled in large part on established approaches to patients with diabetes mellitus or stage 3 or 4 chronic kidney disease, which are well-studied conditions that also show heightened risk for ASCVD events and have been explicitly incorporated into standard clinical guidelines for ASCVD. Based on the available evidence, we conclude that patients with LE or DM require earlier and more aggressive screening and management of ASCVD. We suggest that physicians consider implementing multipliers of conventional risk calculators to trigger earlier initiation of lifestyle modifications and medical therapies in primary prevention of ASCVD events, employ vascular imaging to quantify the burden of subclinical plaques, and treat to lower lipid targets using statins and newer therapies, such as PCSK9 inhibitors, that decrease ASCVD events in nonautoimmune cohorts. More clinical vigilance is needed regarding surveillance, prevention, risk modification, and treatment of dyslipidemias, hypertension, and smoking in patients with LE or DM. All of these goals are achievable.
Collapse
Affiliation(s)
- Emily Keyes
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Corporal Michael J. Crescenz VAMC, Philadelphia, Pennsylvania
| | - Madison Grinnell
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Corporal Michael J. Crescenz VAMC, Philadelphia, Pennsylvania
| | - Douglas Jacoby
- Cardiovascular Division, Department of Medicine, University of Pennsylvania Health System, Philadelphia, Pennsylvania
| | - Thomas Vazquez
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Corporal Michael J. Crescenz VAMC, Philadelphia, Pennsylvania
| | - DeAnna Diaz
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Corporal Michael J. Crescenz VAMC, Philadelphia, Pennsylvania
| | - Victoria P. Werth
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Corporal Michael J. Crescenz VAMC, Philadelphia, Pennsylvania
| | - Kevin Jon Williams
- Department of Cardiovascular Sciences, Department of Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
37
|
Abstract
Cardiovascular disease risk is evident during childhood for patients with juvenile systemic lupus erythematosus, juvenile dermatomyositis, and juvenile idiopathic arthritis. The American Heart Association defines cardiovascular health as a positive health construct reflecting the sum of protective factors against cardiovascular disease. Disease-related factors such as chronic inflammation and endothelial dysfunction increase cardiovascular disease risk directly and through bidirectional relationships with poor cardiovascular health factors. Pharmacologic and nonpharmacologic interventions to improve cardiovascular health and long-term cardiovascular outcomes in children with rheumatic disease are needed.
Collapse
|
38
|
Cardiovascular Disease in Patients With Systemic Lupus Erythematosus: Potential for Improved Primary Prevention With Statins. Cardiol Rev 2021; 29:323-327. [PMID: 34609986 DOI: 10.1097/crd.0000000000000383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cardiovascular disease (CVD) is a significant cause of morbidity and mortality in patients with systemic lupus erythematosus (SLE). This is especially true in SLE patients with traditional CVD risk factors (eg, hypertension, hyperlipidemia, obesity) and disease-related risk factors (eg, increased SLE disease activity, elevated C-reactive protein levels, and antiphospholipid antibodies). The only guidelines in the primary prevention of CVD in SLE patients involve reducing traditional risk factors, but there are additional therapies that may be beneficial, including statin use. Current data on statin use for prevention of CVD in SLE patients are limited, but there have been some promising results. Statin use has been shown to be especially important in SLE patients for decreasing low-density lipoprotein levels and preventing CVD in hyperlipidemic patients. In addition, there is evidence suggesting that it may be beneficial to use statins in SLE patients with chronically elevated high-sensitivity C-reactive protein levels and antiphospholipid antibodies. It is important to continue to investigate the impact of statins on CVD in SLE patients, as they could significantly improve outcomes in patients with this disease.
Collapse
|
39
|
Greenan-Barrett J, Doolan G, Shah D, Virdee S, Robinson GA, Choida V, Gak N, de Gruijter N, Rosser E, Al-Obaidi M, Leandro M, Zandi MS, Pepper RJ, Salama A, Jury EC, Ciurtin C. Biomarkers Associated with Organ-Specific Involvement in Juvenile Systemic Lupus Erythematosus. Int J Mol Sci 2021; 22:7619. [PMID: 34299237 PMCID: PMC8306911 DOI: 10.3390/ijms22147619] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 12/16/2022] Open
Abstract
Juvenile systemic lupus erythematosus (JSLE) is characterised by onset before 18 years of age and more severe disease phenotype, increased morbidity and mortality compared to adult-onset SLE. Management strategies in JSLE rely heavily on evidence derived from adult-onset SLE studies; therefore, identifying biomarkers associated with the disease pathogenesis and reflecting particularities of JSLE clinical phenotype holds promise for better patient management and improved outcomes. This narrative review summarises the evidence related to various traditional and novel biomarkers that have shown a promising role in identifying and predicting specific organ involvement in JSLE and appraises the evidence regarding their clinical utility, focusing in particular on renal biomarkers, while also emphasising the research into cardiovascular, haematological, neurological, skin and joint disease-related JSLE biomarkers, as well as genetic biomarkers with potential clinical applications.
Collapse
Affiliation(s)
- James Greenan-Barrett
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
| | - Georgia Doolan
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
| | - Devina Shah
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
| | - Simrun Virdee
- Department of Ophthalmology, Royal Free Hospital, London NW3 2QG, UK;
| | - George A. Robinson
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
| | - Varvara Choida
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
| | - Nataliya Gak
- Department of Rheumatology, University College London Hospital NHS Foundation Trust, London NW1 2BU, UK; (N.G.); (M.L.)
| | - Nina de Gruijter
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
| | - Elizabeth Rosser
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
| | - Muthana Al-Obaidi
- Department of Paediatric Rheumatology, Great Ormond Street Hospital, London WC1N 3JH, UK;
- NIHR Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Maria Leandro
- Department of Rheumatology, University College London Hospital NHS Foundation Trust, London NW1 2BU, UK; (N.G.); (M.L.)
- Centre for Rheumatology, Division of Medicine, University College London, London WC1E 6DH, UK;
| | - Michael S. Zandi
- Department of Neurology, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London NW1 2BU, UK;
| | - Ruth J. Pepper
- Department of Renal Medicine, Royal Free Hospital, University College London, London NW3 2QG, UK; (R.J.P.); (A.S.)
| | - Alan Salama
- Department of Renal Medicine, Royal Free Hospital, University College London, London NW3 2QG, UK; (R.J.P.); (A.S.)
| | - Elizabeth C. Jury
- Centre for Rheumatology, Division of Medicine, University College London, London WC1E 6DH, UK;
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
- Department of Rheumatology, University College London Hospital NHS Foundation Trust, London NW1 2BU, UK; (N.G.); (M.L.)
| |
Collapse
|
40
|
Kim D, Chung H, Lee JE, Kim J, Hwang J, Chung Y. Immunologic Aspects of Dyslipidemia: a Critical Regulator of Adaptive Immunity and Immune Disorders. J Lipid Atheroscler 2021; 10:184-201. [PMID: 34095011 PMCID: PMC8159760 DOI: 10.12997/jla.2021.10.2.184] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/23/2021] [Accepted: 05/02/2021] [Indexed: 11/09/2022] Open
Abstract
Dyslipidemia is a major cause of cardiovascular diseases which represent a leading cause of death in humans. Diverse immune cells are known to be involved in the pathogenesis of cardiovascular diseases such as atherosclerosis. Conversely, dyslipidemia is known to be tightly associated with immune disorders in humans, as evidenced by a higher incidence of atherosclerosis in patients with autoimmune diseases including psoriasis, rheumatoid arthritis, and systemic lupus erythematosus. Given that the dyslipidemia-related autoimmune diseases are caused by autoreactive T cells and B cells, dyslipidemia seems to directly or indirectly regulate the adaptive immunity. Indeed, accumulating evidence has unveiled that proatherogenic factors can impact the differentiation and function of CD4+ T cells, CD8+ T cells, and B cells. This review discusses an updated overview on the regulation of adaptive immunity by dyslipidemia and proposes a potential therapeutic strategy for immune disorders by targeting lipid metabolism.
Collapse
Affiliation(s)
- Daehong Kim
- Laboratory of Immune Regulation, Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Hayeon Chung
- Laboratory of Immune Regulation, Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Jeong-Eun Lee
- Laboratory of Immune Regulation, Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Jiyeon Kim
- Laboratory of Immune Regulation, Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Junseok Hwang
- Laboratory of Immune Regulation, Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Yeonseok Chung
- Laboratory of Immune Regulation, Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| |
Collapse
|
41
|
Chen W, Wang Q, Zhou B, Zhang L, Zhu H. Lipid Metabolism Profiles in Rheumatic Diseases. Front Pharmacol 2021; 12:643520. [PMID: 33897433 PMCID: PMC8064727 DOI: 10.3389/fphar.2021.643520] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/18/2021] [Indexed: 12/25/2022] Open
Abstract
Rheumatic diseases are a group of chronic autoimmune disorders that involve multiple organs or systems and have high mortality. The mechanisms of these diseases are still ill-defined, and targeted therapeutic strategies are still challenging for physicians. Recent research indicates that cell metabolism plays important roles in the pathogenesis of rheumatic diseases. In this review, we mainly focus on lipid metabolism profiles (dyslipidaemia, fatty acid metabolism) and mechanisms in rheumatic diseases and discuss potential clinical applications based on lipid metabolism profiles.
Collapse
Affiliation(s)
- Weilin Chen
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.,Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, China
| | - Qi Wang
- Department of Radiology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Bin Zhou
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lihua Zhang
- Department of Rheumatology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Honglin Zhu
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.,Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, China
| |
Collapse
|
42
|
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: 18] [Impact Index Per Article: 4.5] [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
|
43
|
Qiu J, Wu B, Goodman SB, Berry GJ, Goronzy JJ, Weyand CM. Metabolic Control of Autoimmunity and Tissue Inflammation in Rheumatoid Arthritis. Front Immunol 2021; 12:652771. [PMID: 33868292 PMCID: PMC8050350 DOI: 10.3389/fimmu.2021.652771] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/15/2021] [Indexed: 12/19/2022] Open
Abstract
Like other autoimmune diseases, rheumatoid arthritis (RA) develops in distinct stages, with each phase of disease linked to immune cell dysfunction. HLA class II genes confer the strongest genetic risk to develop RA. They encode for molecules essential in the activation and differentiation of T cells, placing T cells upstream in the immunopathology. In Phase 1 of the RA disease process, T cells lose a fundamental function, their ability to be self-tolerant, and provide help for autoantibody-producing B cells. Phase 2 begins many years later, when mis-differentiated T cells gain tissue-invasive effector functions, enter the joint, promote non-resolving inflammation, and give rise to clinically relevant arthritis. In Phase 3 of the RA disease process, abnormal innate immune functions are added to adaptive autoimmunity, converting synovial inflammation into a tissue-destructive process that erodes cartilage and bone. Emerging data have implicated metabolic mis-regulation as a fundamental pathogenic pathway in all phases of RA. Early in their life cycle, RA T cells fail to repair mitochondrial DNA, resulting in a malfunctioning metabolic machinery. Mitochondrial insufficiency is aggravated by the mis-trafficking of the energy sensor AMPK away from the lysosomal surface. The metabolic signature of RA T cells is characterized by the shunting of glucose toward the pentose phosphate pathway and toward biosynthetic activity. During the intermediate and terminal phase of RA-imposed tissue inflammation, tissue-residing macrophages, T cells, B cells and stromal cells are chronically activated and under high metabolic stress, creating a microenvironment poor in oxygen and glucose, but rich in metabolic intermediates, such as lactate. By sensing tissue lactate, synovial T cells lose their mobility and are trapped in the tissue niche. The linkage of defective DNA repair, misbalanced metabolic pathways, autoimmunity, and tissue inflammation in RA encourages metabolic interference as a novel treatment strategy during both the early stages of tolerance breakdown and the late stages of tissue inflammation. Defining and targeting metabolic abnormalities provides a new paradigm to treat, or even prevent, the cellular defects underlying autoimmune disease.
Collapse
Affiliation(s)
- Jingtao Qiu
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Bowen Wu
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Stuart B Goodman
- Department of Orthopedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Gerald J Berry
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Jorg J Goronzy
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Cornelia M Weyand
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
44
|
Robinson GA, Waddington KE, Coelewij L, Peng J, Naja M, Wincup C, Radziszewska A, Peckham H, Isenberg DA, Ioannou Y, Ciurtin C, Pineda-Torra I, Jury EC. Increased apolipoprotein-B:A1 ratio predicts cardiometabolic risk in patients with juvenile onset SLE. EBioMedicine 2021; 65:103243. [PMID: 33640328 PMCID: PMC7992074 DOI: 10.1016/j.ebiom.2021.103243] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/29/2021] [Accepted: 01/29/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cardiovascular disease is a leading cause of mortality in patients with juvenile-onset systemic lupus erythematosus (JSLE). Traditional factors for cardiovascular risk (CVR) prediction are less robust in younger patients. More reliable CVR biomarkers are needed for JSLE patient stratification and to identify therapeutic approaches to reduce cardiovascular morbidity and mortality in JSLE. METHODS Serum metabolomic analysis (including >200 lipoprotein measures) was performed on a discovery (n=31, median age 19) and validation (n=31, median age 19) cohort of JSLE patients. Data was analysed using cluster, receiver operating characteristic analysis and logistic regression. RNA-sequencing assessed gene expression in matched patient samples. FINDINGS Hierarchical clustering of lipoprotein measures identified and validated two unique JSLE groups. Group-1 had an atherogenic and Group-2 had an atheroprotective lipoprotien profile. Apolipoprotein(Apo)B:ApoA1 distinguished the two groups with high specificity (96.2%) and sensitivity (96.7%). JSLE patients with high ApoB:ApoA1 ratio had increased CD8+ T-cell frequencies and a CD8+ T-cell transcriptomic profile enriched in genes associated with atherogenic processes including interferon signaling. These metabolic and immune signatures overlapped statistically significantly with lipid biomarkers associated with sub-clinical atherosclerosis in adult SLE patients and with genes overexpressed in T-cells from human atherosclerotic plaque respectively. Finally, baseline ApoB:ApoA1 ratio correlated positively with SLE disease activity index (r=0.43, p=0.0009) and negatively with Lupus Low Disease Activity State (r=-0.43, p=0.0009) over 5-year follow-up. INTERPRETATION Multi-omic analysis identified high ApoB:ApoA1 as a potential biomarker of increased cardiometabolic risk and worse clinical outcomes in JSLE. ApoB:ApoA1 could help identify patients that require increased disease monitoring, lipid modification or lifestyle changes. FUNDING Lupus UK, The Rosetrees Trust, British Heart Foundation, UCL & Birkbeck MRC Doctoral Training Programme and Versus Arthritis.
Collapse
Affiliation(s)
- George A Robinson
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK; Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK.
| | - Kirsty E Waddington
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK; Centre for Cardiometabolic and Vascular Science, Department of Medicine, University College London, London W1CE 6JF, UK
| | - Leda Coelewij
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK; Centre for Cardiometabolic and Vascular Science, Department of Medicine, University College London, London W1CE 6JF, UK
| | - Junjie Peng
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK; Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK
| | - Meena Naja
- Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK
| | - Chris Wincup
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK
| | - Anna Radziszewska
- Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK
| | - Hannah Peckham
- Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK
| | - David A Isenberg
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK; Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK
| | - Yiannis Ioannou
- Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK
| | - Coziana Ciurtin
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK; Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK.
| | - Ines Pineda-Torra
- Centre for Cardiometabolic and Vascular Science, Department of Medicine, University College London, London W1CE 6JF, UK.
| | - Elizabeth C Jury
- Centre for Rheumatology Research, Department of Medicine, University College London, Rayne Building, London W1CE 6JF, UK.
| |
Collapse
|
45
|
Skaggs BJ, Grossman J, Sahakian L, Perry L, FitzGerald J, Charles-Schoeman C, Gorn A, Taylor M, Moriarty J, Ragavendra N, Weisman M, Wallace DJ, Hahn BH, McMahon M. A Panel of Biomarkers Associates With Increased Risk for Cardiovascular Events in Women With Systemic Lupus Erythematosus. ACR Open Rheumatol 2021; 3:209-220. [PMID: 33605563 PMCID: PMC8063147 DOI: 10.1002/acr2.11223] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 12/21/2020] [Indexed: 01/14/2023] Open
Abstract
Objective The increase in cardiovascular events (CVEs) in systemic lupus erythematosus (SLE) is not fully explained by traditional risk factors. We previously identified four biomarkers (proinflammatory high‐density lipoprotein, leptin, soluble TNF‐like weak inducer of apoptosis (sTWEAK), and homocysteine) that we combined with age and diabetes to create the predictors of risk for elevated flares, damage progression, and increased cardiovascular diseasein patients with SLE (PREDICTS) risk profile. PREDICTS more accurately identified patients with SLE at risk for progression of subclinical atherosclerosis than any individual variable. We examined whether PREDICTS can also identify patients with SLE at risk for future CVEs. Methods A total of 342 patients with SLE and 155 matched control subjects participated in this longitudinal prospective study. A high PREDICTS score was defined as three or more predictors or diabetes + one or more predictor. The biomarkers were measured at baseline using published methods. All major adverse CVEs (MACEs) were confirmed by medical record review. Results During 116 months of follow‐up, 5% of patients with SLE died, 12% had a cerebrovascular event, and 5% had a cardiac event. Overall, 20% of patients with lupus experienced any new MACE compared with 5% of control subjects (P < 0.0001). More patients with SLE with a new MACE had high PREDICTS score at baseline (77%) versus patients with no new events (34%) (P < 0.0001). High baseline PREDICTS score also associated with cerebrovascular (P < 0.0001) and cardiac events (P < 0.0001) in SLE. Using Cox regression, a baseline high PREDICTS score associated with a 3.7‐fold increased hazard ratio (HR) for a new MACE (P < 0.0001) in SLE. Hypertension (HR = 2.1; P = 0.006) was also a risk. Conclusion A high PREDICTS score and hypertension confer increased risk for new MACEs in patients with SLE.
Collapse
Affiliation(s)
- Brian J Skaggs
- University of California, Los Angeles, David Geffen School of Medicine, Los Angeles, CA
| | - Jennifer Grossman
- University of California, Los Angeles, David Geffen School of Medicine, Los Angeles, CA
| | - Lori Sahakian
- University of California, Los Angeles, David Geffen School of Medicine, Los Angeles, CA
| | - Lucas Perry
- University of California, Los Angeles, David Geffen School of Medicine, Los Angeles, CA
| | - John FitzGerald
- University of California, Los Angeles, David Geffen School of Medicine, Los Angeles, CA
| | | | - Alan Gorn
- University of California, Los Angeles, David Geffen School of Medicine, Los Angeles, CA
| | - Mihaela Taylor
- University of California, Los Angeles, David Geffen School of Medicine, Los Angeles, CA
| | - John Moriarty
- University of California, Los Angeles, David Geffen School of Medicine, Los Angeles, CA
| | - Nagesh Ragavendra
- University of California, Los Angeles, David Geffen School of Medicine, Los Angeles, CA
| | | | - Daniel J Wallace
- University of California, Los Angeles, David Geffen School of Medicine, Los Angeles, CA.,Cedars Sinai Medical Center, Los Angeles, California
| | - Bevra H Hahn
- University of California, Los Angeles, David Geffen School of Medicine, Los Angeles, CA
| | - Maureen McMahon
- University of California, Los Angeles, David Geffen School of Medicine, Los Angeles, CA
| |
Collapse
|
46
|
Weng W, Wang Q, Wei C, Adu-Frimpong M, Toreniyazov E, Ji H, Yu J, Xu X. Mixed micelles for enhanced oral bioavailability and hypolipidemic effect of liquiritin: preparation, in vitro and in vivo evaluation. Drug Dev Ind Pharm 2021; 47:308-318. [PMID: 33494627 DOI: 10.1080/03639045.2021.1879839] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Liquiritin, as one of the main flavonoids in Glycyrrhiza, exhibits extensive pharmacological effects, such as the anti-oxidant, anti-inflammatory, anti-tumor and so on. Herein, the aqueous solubility and oral bioavailability of liquiritin was purposely enhanced via the preparation of the mixed micelles. METHODS The liquiritin-loaded micelles (LLM) were fabricated via thin-film dispersion method. The optimal LLM formulation was evaluated through physical properties including particle size (PS), encapsulation efficiency (EE) and drug loading (DL). In vitro accumulate release as well as in vivo pharmacokinetics were also evaluated. Moreover, the hypolipidemic activity of LLM was observed in the hyperlipidemia mice model. RESULTS The LLM exhibited a homogenous spherical shape with small mean PS, good stability and high encapsulation efficiency. The accumulate release rates in vitro of the LLM were obviously higher than free liquiritin. The oral bioavailability of the formulation was heightened by 3.98 times in comparison with the free liquiritin. More importantly, LLM increased the hypolipidemic and effect of alleviating lipid metabolism disorder in hepatocytes of liquiritin in hyperlipidemia mice model. CONCLUSIONS Collectively, the improved solubility of liquiritin in water coupled with its enhanced oral bioavailability and concomitant hypolipidemic activity could be attributed to the incorporation of the drug into the mixed micelles.
Collapse
Affiliation(s)
- Wen Weng
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
| | - Qilong Wang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
| | - Chunmei Wei
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
| | - Michael Adu-Frimpong
- Department of Biochemistry and Biotechnology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Elmurat Toreniyazov
- Ashkent State Agricultural University (Nukus branch), Nukus, The Republic of Uzbekistan.,Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, People's Republic of China
| | - Hao Ji
- Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, People's Republic of China.,Jiangsu Tian Sheng Pharmaceutical Co., Ltd, Zhenjiang, People's Republic of China
| | - Jiangnan Yu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China.,Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, People's Republic of China
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China.,Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, People's Republic of China
| |
Collapse
|
47
|
Abrahami D, Hudson M, Suissa S. Statins and lower mortality in rheumatic diseases: An effect of immortal time bias? Semin Arthritis Rheum 2020; 51:211-218. [PMID: 33385861 DOI: 10.1016/j.semarthrit.2020.11.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/26/2020] [Accepted: 11/12/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Randomized controlled trials of the effectiveness of statins on rheumatic disease outcomes have shown limited or no benefit. On the other hand, observational studies have reported remarkable effectiveness of statins at reducing mortality in patients with rheumatic diseases. We evaluated these observational studies for the presence of immortal time bias, which tends to exaggerate the effectiveness of drugs by creating a survival advantage for exposed patients. METHODS We searched PubMed for observational studies investigating the impact of statins in patients with common rheumatic diseases, including rheumatoid arthritis, osteoarthritis, lupus, ankylosing spondylitis, gout and systemic autoimmune diseases. Studies were included if estimates for all-cause mortality among statin users compared to non-users were reported. We evaluated each study for the presence of immortal time bias and estimated the impact of the bias on the published results. RESULTS We found eight observational studies investigating the impact of statins on mortality among patients with rheumatic diseases. Four studies were affected by the classical variant of immortal time bias, while the others introduced immortal time into the comparator via random-calendar date assignment. The studies with the classical form of immortal time bias, which tends to exaggerate drug effectiveness, reported protective effects of statins on mortality ranging from 13% to 57% reductions. In contrast, immortal time bias through random-calendar date assignment, which tends to play down the effectiveness by introducing immortal time in the comparator, reported 16% to 37% reductions in mortality. CONCLUSION Bias in observational studies may explain the discrepancy in findings with randomized controlled trials on the effectiveness of statins in patients with rheumatic diseases. Future observational studies will need to rely on incident and prevalent new-user designs that emulate randomized trials and avoid immortal time bias.
Collapse
Affiliation(s)
- Devin Abrahami
- Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Canada; Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Canada
| | - Marie Hudson
- Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Canada; Division of Rheumatology, Jewish General Hospital, Montreal, Canada; Department of Medicine, McGill University, Montréal, Canada
| | - Samy Suissa
- Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Canada; Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Canada; Department of Medicine, McGill University, Montréal, Canada.
| |
Collapse
|
48
|
Tay SH, Celhar T, Fairhurst A. Low-Density Neutrophils in Systemic Lupus Erythematosus. Arthritis Rheumatol 2020; 72:1587-1595. [PMID: 32524751 PMCID: PMC7590095 DOI: 10.1002/art.41395] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 06/04/2020] [Indexed: 12/13/2022]
Abstract
Patients with systemic lupus erythematosus (SLE) display increased numbers of immature neutrophils in the blood, but the exact role of these immature neutrophils is unclear. Neutrophils that sediment within the peripheral blood mononuclear cell fraction after density centrifugation of blood are generally defined as low-density neutrophils (LDNs). Far beyond antimicrobial functions, LDNs are emerging as decision-shapers during innate and adaptive immune responses. Traditionally, neutrophils have been viewed as a homogeneous population. However, the various LDN populations identified in SLE to date are heterogeneously composed of mixed populations of activated mature neutrophils and immature neutrophils at various stages of differentiation. Controversy also surrounds the role of LDNs in SLE in terms of whether they are proinflammatory or polymorphonuclear myeloid-derived suppressor cells. It is clear that LDNs in SLE can secrete increased levels of type I interferon (IFN) and that they contribute to the cycle of inflammation and tissue damage. They readily form neutrophil extracellular traps, exposing modified autoantigens and oxidized mitochondrial DNA, which contribute to autoantibody production and type I IFN signaling, respectively. Importantly, the ability of LDNs in SLE to perform canonical neutrophil functions is polarized, based on mature CD10+ and immature CD10- neutrophils. Although this field is still relatively new, multiomic approaches have advanced our understanding of the diverse origins, phenotype, and function of LDNs in SLE. This review updates the literature on the origin and nature of LDNs, their distinctive features, and their biologic roles in the immunopathogenesis and end-organ damage in SLE.
Collapse
Affiliation(s)
- Sen Hee Tay
- National University Hospital Yong Loo Lin School of MedicineInstitute for Molecular and Cellular Biology, Agency for Science, Technology and ResearchSingapore
| | - Teja Celhar
- Singapore Immunology NetworkAgency for Science, Technology and ResearchSingapore
| | - Anna‐Marie Fairhurst
- Institute for Molecular and Cellular BiologyAgency for Science, Technology and ResearchUniversity of Singapore Yong Loo Lin School of MedicineSingapore
| |
Collapse
|
49
|
Xie W, Huang H, Xiao S, Yang X, Zhang Z. Effect of statin use on cardiovascular events and all-cause mortality in immune-mediated inflammatory diseases: A systematic review and meta-analysis involving 148,722 participants. Pharmacol Res 2020; 160:105057. [DOI: 10.1016/j.phrs.2020.105057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/15/2020] [Accepted: 06/28/2020] [Indexed: 02/07/2023]
|
50
|
YKL-40 as a novel biomarker in cardio-metabolic disorders and inflammatory diseases. Clin Chim Acta 2020; 511:40-46. [PMID: 33002471 DOI: 10.1016/j.cca.2020.09.035] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 09/25/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023]
Abstract
Dyslipidaemia is associated with numerous health problems that include the combination of insulin resistance, hypertension and obesity, ie, metabolic syndrome. Although the use of statins to decrease serum low density lipoprotein cholesterol (LDL-C) has been an effective therapeutic in treating atherosclerosis, the persistence of high atherosclerotic risk, ie, residual risk, is notable and is not simply explained as a phenomenon of dyslipidaemia. As such, it is imperative that we identify new biomarkers to monitor treatment and more accurately predict future cardiovascular events. This athero-protective strategy includes the assessment of novel inflammatory biomarkers such as YKL-40. Recent evidence has implicated YKL-40 in patients with inflammatory diseases and cardio-metabolic disorders, making it potentially useful to evaluate disease severity, prognosis and survival. In this review, we summarize role of YKL-40 in the pathogenesis of cardio-metabolic disorders and explore its use as a novel biomarker for monitoring athero-protective therapy.
Collapse
|