1
|
Frostegård J, Åkesson A, Helte E, Söderlund F, Su J, Hua X, Rautiainen S, Wolk A. Antibodies Against Phosphorylcholine in Prediction of Cardiovascular Disease Among Women: A Population-Based Prospective Cohort Study. JACC. ADVANCES 2024; 3:101298. [PMID: 39741640 PMCID: PMC11686053 DOI: 10.1016/j.jacadv.2024.101298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/29/2024] [Accepted: 08/12/2024] [Indexed: 01/03/2025]
Abstract
Background Antibodies against phosphorylcholine (anti-PC) have been reported as associated with protection against atherosclerosis, cardiovascular disease (CVD), and other chronic inflammatory diseases. Underlying potential mechanisms have been demonstrated and include anti-inflammatory, clearance of dead cells, and inhibition of oxidized low-density lipoprotein effects. Objectives This study examined the role of IgM anti-PC and incident CVD among women, where less is known than among men in the general population. Methods In a total of 932 women, age 66 ± 6 years at baseline, from the population-based Swedish Mammography Cohort, IgM anti-PC levels of sera were measured using Enzyme Linked Immunosorbent assay. Prospective associations with any first CVD, ischemic heart disease (IHD), myocardial infarction (MI), and ischemic stroke were assessed using Cox proportional hazard regression, generating HRs and 95% CIs. The model was adjusted for potential confounding factors. Results Over the course of 16 years (13,033 person-years), we identified 113 cases of composite CVD, 69 cases of IHD, 44 cases of MI, and 50 cases of ischemic stroke. IgM anti-PC was statistically significantly inversely associated with risk of CVD, IHD, and MI, but not with ischemic stroke. Comparing the highest tertile with lowest, we observed multivariable-adjusted HR of 0.27 (95% CI: 0.11-0.68; P trend <0.01) for MI. Conclusions IgM anti-PC may play an active role in inhibition of CVD development in women, especially MI. Furthermore, IgM anti-PC levels may play a role in identifying those at risk.
Collapse
Affiliation(s)
- Johan Frostegård
- Units of Immunology and chronic disease, and Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Agneta Åkesson
- Units of Immunology and chronic disease, and Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Emilie Helte
- Units of Immunology and chronic disease, and Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Söderlund
- Units of Immunology and chronic disease, and Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jun Su
- Units of Immunology and chronic disease, and Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Xiang Hua
- Units of Immunology and chronic disease, and Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Susanne Rautiainen
- Units of Immunology and chronic disease, and Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Alicja Wolk
- Units of Immunology and chronic disease, and Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
2
|
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
|
3
|
Maes M, Rachayon M, Jirakran K, Sughondhabirom A, Almulla AF, Sodsai P. Role of T and B lymphocyte cannabinoid type 1 and 2 receptors in major depression and suicidal behaviours. Acta Neuropsychiatr 2024; 36:287-298. [PMID: 37681553 DOI: 10.1017/neu.2023.35] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
Early flow cytometry studies revealed T cell activation in major depressive disorder (MDD). MDD is characterised by activation of the immune-inflammatory response system (IRS) and the compensatory immunoregulatory system (CIRS), including deficits in T regulatory (Treg) cells. This study examines the number of cannabinoid type 1 (CB1) and type 2 (CB2) receptor-bearing T/B lymphocytes in MDD, and the effects of in vitro cannabidiol (CBD) administration on CB1/CB2-bearing immunocytes. Using flow cytometry, we determined the percentage of CD20+CB2+, CD3+CB2+, CD4+CB2+, CD8+CB2+ and FoxP3+CB1+ cells in 19 healthy controls and 29 MDD patients in 5 conditions: baseline, stimulation with anti-CD3/CD28 with or without 0.1 µg/mL, 1.0 µg/mL, or 10.0 µg/mL CBD. CB2+ was significantly higher in CD20+ than CD3+ and CD4+ and CD 8+ cells. Stimulation with anti-CD3/CD8 increases the number of CB2-bearing CD3+, CD4+ and CD8+ cells, as well as CB1-bearing FoxP3+ cells. There was an inverse association between the number of reduced CD4+ CB2+ and IRS profiles, including M1 macrophage, T helper-(Th)-1 and Th-17 phenotypes. MDD is characterised by lowered basal FoxP3+ CB1+% and higher CD20+ CB2+%. 33.2% of the variance in the depression phenome (including severity of depression, anxiety and current suicidal behaviours) is explained by CD20+ CB2+ % (positively) and CD3+ CB2+% (inversely). All five immune cell populations were significantly increased by 10 µg/mL of CBD administration. Reductions in FoxP3+ CB1+% and CD3+ /CD4+ CB2+% contribute to deficits in immune homoeostasis in MDD, while increased CD20+CB2+% may contribute to the pathophysiology of MDD by activating T-independent humoral immunity.
Collapse
Affiliation(s)
- Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China,Chengdu610072, China
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
- Research Institute, Medical University Plovdiv, Plovdiv, Bulgaria
- Kyung Hee University, Seoul, Korea
| | - Muanpetch Rachayon
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Ketsupar Jirakran
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
- Department of Pediatrics, Faculty of Medicine, Maximizing Thai Children's Developmental Potential Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - Atapol Sughondhabirom
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Abbas F Almulla
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Pimpayao Sodsai
- Department of Immunology, Faculty of Medicine, Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| |
Collapse
|
4
|
Ghalali A, Alhamdan F, Upadhyay S, Ganguly K, Larsson K, Palmberg L, Rahman M. Contrasting effects of intracellular and extracellular human PCSK9 on inflammation, lipid alteration and cell death. Commun Biol 2024; 7:985. [PMID: 39138259 PMCID: PMC11322528 DOI: 10.1038/s42003-024-06674-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 08/02/2024] [Indexed: 08/15/2024] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is one of the major regulators of low-density lipoprotein receptor (LDLR). Information on role and regulation of PCSK9 in lung is very limited. Our study focuses on understanding the role and regulation of PCSK9 in the lung. PCSK9 levels are higher in Bronchoalveolar lavage fluid (BALF) of smokers with or without chronic obstructive pulmonary diseases (COPD) compared to BALF of nonsmokers. PCSK9-stimulated cells induce proinflammatory cytokines and activation of MAPKp38. PCSK9 transcripts are highly expressed in healthy individuals compared to COPD, pulmonary fibrosis or pulmonary systemic sclerosis. Cigarette smoke extract reduce PCSK9 levels in undifferentiated pulmonary bronchial epithelial cells (PBEC) but induce in differentiated PBEC. PCSK9 inhibition affect biological pathways, induces lipid peroxidation, and higher level of apoptosis in response to staurosporine. Our results suggest that higher levels of PCSK9 in BALF acts as an inflammatory marker. Furthermore, extracellular and intracellular PCSK9 play different roles.
Collapse
Affiliation(s)
- Aram Ghalali
- Vascular Biology Program, Boston Children Hospital, Harvard Medical school, Boston, MA, USA
| | - Fahd Alhamdan
- Department of Anesthesiology, Critical Care, and Pain Medicine, Cardiac Anesthesia Division, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Swapna Upadhyay
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Insitutet, Stockholm, Sweden
| | - Koustav Ganguly
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Insitutet, Stockholm, Sweden
| | - Kjell Larsson
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Insitutet, Stockholm, Sweden
| | - Lena Palmberg
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Insitutet, Stockholm, Sweden
| | - Mizanur Rahman
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Insitutet, Stockholm, Sweden.
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Cao S, Jiang J, Yin H, Wang L, Lu Q. Abnormal energy metabolism in the pathogenesis of systemic lupus erythematosus. Int Immunopharmacol 2024; 134:112149. [PMID: 38692019 DOI: 10.1016/j.intimp.2024.112149] [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: 03/18/2024] [Revised: 04/20/2024] [Accepted: 04/21/2024] [Indexed: 05/03/2024]
Abstract
Systemic lupus erythematosus (SLE) is a severe autoimmune disease with significant socioeconomic impact worldwide. Orderly energy metabolism is essential for normal immune function, and disordered energy metabolism is increasingly recognized as an important contributor to the pathogenesis of SLE. Disorders of energy metabolism are characterized by increased reactive oxygen species, ATP deficiency, and abnormal metabolic pathways. Oxygen and mitochondria are critical for the production of ATP, and both mitochondrial dysfunction and hypoxia affect the energy production processes. In addition, several signaling pathways, including mammalian target of rapamycin (mTOR)/adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) signaling and the hypoxia-inducible factor (HIF) pathway also play important regulatory roles in energy metabolism. Furthermore, drugs with clear clinical effects on SLE, such as sirolimus, metformin, and tacrolimus, have been proven to improve the disordered energy metabolism of immune cells, suggesting the potential of targeting energy metabolism for the treatment of SLE. Moreover, several metabolic modulators under investigation are expected to have potential therapeutic effects in SLE. This review aimed to gain insights into the role and mechanism of abnormal energy metabolism in the pathogenesis of SLE, and summarizes the progression of metabolic modulator in the treatment of SLE.
Collapse
Affiliation(s)
- Shumei Cao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, 210042, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China
| | - Jiao Jiang
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, 210042, China
| | - Haoyuan Yin
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, 210042, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China
| | - Lai Wang
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, 210042, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China.
| | - Qianjin Lu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, 210042, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China; Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
6
|
Qin R, Wu H, Guan H, Tang C, Zheng Z, Deng C, Chen C, Zou Q, Lu L, Ma K. Anti-phospholipid autoantibodies in human diseases. Clin Immunol 2023; 256:109803. [PMID: 37821073 DOI: 10.1016/j.clim.2023.109803] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/24/2023] [Accepted: 10/05/2023] [Indexed: 10/13/2023]
Abstract
Anti-phospholipid autoantibodies are a group of antibodies that can specifically bind to anionic phospholipids and phospholipid protein complexes. Recent studies have reported elevated serum anti-phospholipid autoantibody levels in patients with antiphospholipid syndrome, systemic lupus erythematosus, rheumatoid arthritis, metabolic disorders, malaria, SARS-CoV-2 infection, obstetric diseases and cardiovascular diseases. However, the underlying mechanisms of anti-phospholipid autoantibodies in disease pathogenesis remain largely unclear. Emerging evidence indicate that anti-phospholipid autoantibodies modulate NETs formation, monocyte activation, blockade of apoptotic cell phagocytosis in macrophages, complement activation, dendritic cell activation and vascular endothelial cell activation. Herein, we provide an update on recent advances in elucidating the effector mechanisms of anti-phospholipid autoantibodies in the pathogenesis of various diseases, which may facilitate the development of potential therapeutic targets for the treatment of anti-phospholipid autoantibody-related disorders.
Collapse
Affiliation(s)
- Rencai Qin
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Centre for Infection and Immunity Studies (CIIS), School of Medicine, The Seventh Affiliated Hospital, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Haiqi Wu
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Centre for Infection and Immunity Studies (CIIS), School of Medicine, The Seventh Affiliated Hospital, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Hui Guan
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Centre for Infection and Immunity Studies (CIIS), School of Medicine, The Seventh Affiliated Hospital, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Chun Tang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Centre for Infection and Immunity Studies (CIIS), School of Medicine, The Seventh Affiliated Hospital, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Zhihua Zheng
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Centre for Infection and Immunity Studies (CIIS), School of Medicine, The Seventh Affiliated Hospital, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Chong Deng
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong 999077, China
| | - Chengshun Chen
- Department of Rheumatology and Immunology, First Affiliated Hospital of Army Medical University, Chongqing, China
| | - Qinghua Zou
- Department of Rheumatology and Immunology, First Affiliated Hospital of Army Medical University, Chongqing, China.
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong 999077, China.
| | - Kongyang Ma
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Centre for Infection and Immunity Studies (CIIS), School of Medicine, The Seventh Affiliated Hospital, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| |
Collapse
|
7
|
Samal SK, Leander K, Vikström M, Griesbaum L, de Faire U, Frostegård J. Antibodies against malondialdehyde among 60-year-olds: prediction of cardiovascular disease. Sci Rep 2023; 13:15011. [PMID: 37697019 PMCID: PMC10495339 DOI: 10.1038/s41598-023-42264-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 09/07/2023] [Indexed: 09/13/2023] Open
Abstract
Malondialdehyde (MDA) is generated in oxidized LDL. It forms covalent protein adducts, and is recognized by antibodies (anti-MDA). We previously studied IgM anti-MDA, and here we focus on IgG, IgG1 and IgG2 anti-MDA in predicting cardiovascular disease (CVD). We determined, by ELISA, anti-MDA in a 7-year follow-up of 60-year-old men and women from Stockholm County (2039 men, 2193 women). We identified 210 incident CVD cases (defined as new events of myocardial infarction (MI), and hospitalization for angina pectoris) and ischemic stroke, and 620 age- and sex-matched controls. IgG anti-MDA was not associated with CVD. Median values only differed significantly for IgG1 anti-MDA among men, with lower levels among cases than controls (p = 0.039). High IgG1 anti-MDA (above 75th percentile) was inversely associated with CVD risk after adjustment for smoking, body mass index, type 2 diabetes, hyperlipidemia, and hypertension, (OR and 95% CI: 0.59; 0.40-0.89). After stratification by sex, this association emerged in men (OR and 95% CI: 0.46; 0.27-0.77), but not in women. IgG2 anti-MDA were associated with protection in the whole group and among men though weaker than IgG1 anti-MDA. IgG2 anti-MDA above the 75th percentile was associated with an increased risk of MI/angina in women (OR and 95% CI: 2.57; (1.08-6.16)). IgG1 and less so IgG2 anti-MDA are protection markers for CVD and MI/angina in the whole group and among men. However, IgG2 anti-MDA was a risk marker for MI/angina among women. These findings could have implications for both prediction and therapy.
Collapse
Affiliation(s)
- Shailesh Kumar Samal
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Nobels Väg 13, 17165, Stockholm, Sweden
| | - Karin Leander
- Division of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Max Vikström
- Division of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lena Griesbaum
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Nobels Väg 13, 17165, Stockholm, Sweden
| | - Ulf de Faire
- Division of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Johan Frostegård
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Nobels Väg 13, 17165, Stockholm, Sweden.
| |
Collapse
|
8
|
Wang Z, Wu Z, Tu J, Xu B. Muscle food and human health: A systematic review from the perspective of external and internal oxidation. Trends Food Sci Technol 2023; 138:85-99. [DOI: 10.1016/j.tifs.2023.06.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
9
|
Frostegård J. Antibodies against Phosphorylcholine-Implications for Chronic Inflammatory Diseases. Metabolites 2023; 13:720. [PMID: 37367878 DOI: 10.3390/metabo13060720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 06/28/2023] Open
Abstract
Atherosclerosis and its main consequence, cardiovascular disease (CVD) are nowadays regarded as chronic inflammatory disease conditions, and CVD is the main cause of death in the world. Other examples of chronic inflammation are rheumatic and other autoimmune conditions, but also diabetes, obesity, and even osteoarthritis among others. In addition, infectious diseases can have traits in common with these conditions. Systemic lupus erythematosus (SLE) is a prototypical autoimmune disease, where atherosclerosis is increased and the risk of CVD is very high. This is a clinical problem but could also shed light on the role of the immune system in atherosclerosis and CVD. Underlying mechanisms are of major interest and these are only partially known. Phosphorylcholine (PC) is a small lipid-related antigen, which is both a danger associated molecular pattern (DAMP), and a pathogen associated molecular pattern (PAMP). Antibodies against PC are ubiquitous and 5-10% of circulating IgM is IgM anti-PC. Anti-PC, especially IgM and IgG1 anti-PC, has been associated with protection in the chronic inflammatory conditions mentioned above, and develops during the first years of life, while being present at very low levels at birth. Animal experiments with immunization to raise anti-PC ameliorate atherosclerosis and other chronic inflammatory conditions. Potential mechanisms include anti-inflammatory, immune modulatory, clearance of dead cells and protection against infectious agents. An intriguing possibility is to raise anti-PC levels through immunization, to prevent and/or ameliorate chronic inflammation.
Collapse
Affiliation(s)
- Johan Frostegård
- IMM, Nobels Väg 13, Karolinska Institutet, 17165 Stockholm, Sweden
| |
Collapse
|
10
|
Abstract
The prognosis in systemic lupus erythematosus (SLE) has improved due to better treatment and care, but cardiovascular disease (CVD) still remains an important clinical problem, since the risk of CVD in SLE is much higher than among controls. Atherosclerosis is the main cause of CVD in the general population, and in SLE, increased atherosclerosis, especially the prevalence of atherosclerotic plaques, has been demonstrated. Atherosclerosis is an inflammatory condition, where immunity plays an important role. Interestingly, oxidized low-density lipoprotein, defective clearance of dead cells, and inflammation, with a pro-inflammatory T-cell profile are characteristics of both atherosclerosis and SLE. In addition to atherosclerosis as an underlying cause of CVD in SLE, there are also other non-mutually exclusive mechanisms, and the most important of these are antiphospholipid antibodies (aPL) leading to the antiphospholipid antibody syndrome with both arterial and venous thrombosis. aPL can cause direct pro-inflammatory and prothrombotic effects on endothelial and other cells and also interfere with the coagulation, for example, by inhibiting annexin A5 from its antithrombotic and protective effects. Antibodies against phosphorylcholine (anti-PC) and other small lipid-related epitopes, sometimes called natural antibodies, are negatively associated with CVD and atherosclerosis in SLE. Taken together, a combination of traditional risk factors such as hypertension and dyslipidemia, and nontraditional ones, especially aPL, inflammation, and low anti-PC are implicated in the increased risk of CVD in SLE. Close monitoring of both traditional risk factors and nontraditional ones, including treatment of disease manifestations, not lest renal disease in SLE, is warranted.
Collapse
Affiliation(s)
- Johan Frostegård
- Section of Immunology and Chronic Disease, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
11
|
Guzmán-Martínez G, Marañón C. Immune mechanisms associated with cardiovascular disease in systemic lupus erythematosus: A path to potential biomarkers. Front Immunol 2022; 13:974826. [PMID: 36420265 PMCID: PMC9677819 DOI: 10.3389/fimmu.2022.974826] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/13/2022] [Indexed: 10/29/2023] Open
Abstract
Systemic lupus erythematosus (SLE) patients display an increased risk of cardiovascular disease (CVD). With the improved clinical management of other classical severe manifestation of the disease, CVD is becoming one of the most relevant complications of SLE, and it is an important factor causing morbidity and mortality. Several immune constituents have been shown to be involved in the pathogenesis of atherosclerosis and endothelial damage in SLE patients, including specific circulating cell populations, autoantibodies, and inflammatory mediators. In this review, we summarize the presentation of CVD in SLE and the role of the autoimmune responses present in SLE patients in the induction of atherogenesis, endothelial impairment and cardiac disease. Additionally, we discuss the utility of these immune mediators as early CVD biomarkers and targets for clinical intervention in SLE patients.
Collapse
Affiliation(s)
- Gabriela Guzmán-Martínez
- Atrys Health, Madrid, Spain
- Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
- Department of Cardiology, La Paz University Hospital, IdiPaz, Madrid, Spain
| | - Concepción Marañón
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| |
Collapse
|
12
|
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
|
13
|
Frostegård J. Antibodies against phosphorylcholine and protection against atherosclerosis, cardiovascular disease and chronic inflammation. Expert Rev Clin Immunol 2022; 18:525-532. [PMID: 35471137 DOI: 10.1080/1744666x.2022.2070475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Chronic inflammatory diseases include cardiovascular disease (CVD) atherosclerosis, rheumatic and autoimmune diseases, and others, constitute a large part of the disease burden. It is therefore of major importance to improve understanding of underlying mechanisms, prediction and treatment. AREAS COVERED Broad fields including atherosclerosis, immunology and inflammation are covered, through searches on Pubmed and background knowledge. Phosphorylcholine (PC) is both a danger associated molecular pattern (DAMP), present on oxidized LDL (OxLDL) in atherosclerotic lesions and dead cells, and a pathogen associated molecular pattern (PAMP), present on microorganisms. IgM and IgG1 antibodies against PC (anti-PC) are associated with protection in several chronic inflammatory conditions, especially in CVD and atherosclerosis where most research has been done. PC-immunization ameliorates atherosclerosis in animal models and several potential underlying mechanisms have been proposed, including anti-inflammatory, decreased uptake of OxLDL in the artery wall, promotion of T regulatory cells. Anti-PC develops during the first years of life. Low levels of IgM and IgG1 anti-PC may be caused by lack of exposure to microorganisms, including nematodes and helminths among others. EXPERT OPINION anti-PC could improve prediction of clinical outcome and raising anti-PC could be developed into a novel therapy.
Collapse
Affiliation(s)
- Johan Frostegård
- Institute of Environmental Medicine, Karolinska Institutet, Nobels väg 15, 17165 Stockholm, Sweden,
| |
Collapse
|
14
|
She Z, Li C, Wu F, Mao J, Xie M, Hun M, Abdirahman AS, Luo S, Wan W, Tian J, Wen C. The Role of B1 Cells in Systemic Lupus Erythematosus. Front Immunol 2022; 13:814857. [PMID: 35418972 PMCID: PMC8995743 DOI: 10.3389/fimmu.2022.814857] [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: 11/14/2021] [Accepted: 03/03/2022] [Indexed: 11/13/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disease characterized by multisystemic and multi-organ involvement, recurrent relapses and remissions, and the presence of large amounts of autoantibodies in the body as the main clinical features. The mechanisms involved in this disease are complex and remain poorly understood; however, they are generally believed to be related to genetic susceptibility factors, external stimulation of the body’s immune dysfunction, and impaired immune regulation. The main immune disorders include the imbalance of T lymphocyte subsets, hyperfunction of B cells, production of large amounts of autoantibodies, and further deposition of immune complexes, which result in tissue damage. Among these, B cells play a major role as antibody-producing cells and have been studied extensively. B1 cells are a group of important innate-like immune cells, which participate in various innate and autoimmune processes. Yet the role of B1 cells in SLE remains unclear. In this review, we focus on the mechanism of B1 cells in SLE to provide new directions to explore the pathogenesis and treatment modalities of SLE.
Collapse
Affiliation(s)
- Zhou She
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Cuifang Li
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Feifeng Wu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jueyi Mao
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Min Xie
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Marady Hun
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Amin Sheikh Abdirahman
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Senlin Luo
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wuqing Wan
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jidong Tian
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chuan Wen
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
15
|
Samal SK, Panda PK, Vikström M, Leander K, de Faire U, Ahuja R, Frostegård J. Antibodies Against Phosphorylcholine Among 60-Year-Olds: Clinical Role and Simulated Interactions. Front Cardiovasc Med 2022; 9:809007. [PMID: 35479288 PMCID: PMC9035555 DOI: 10.3389/fcvm.2022.809007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/25/2022] [Indexed: 01/20/2023] Open
Abstract
AimsAntibodies against phosphorylcholine (anti-PC) are implicated as protection markers in atherosclerosis, cardiovascular disease (CVD), and other chronic inflammatory conditions. Mostly, these studies have been focused on IgM. In this study, we determined IgG, IgG1, and IgG2 anti-PC among 60-year-olds.MethodsBased on a 7-year follow-up of 60-year-olds (2,039 men and 2,193 women) from Stockholm County, we performed a nested case-control study of 209 incident CVD cases with 620 age- and sex-matched controls. Anti-PC was determined using ELISA. We predicted the binding affinity of PC with our fully human, in-house-produced IgG1 anti-PC clones (i.e., A01, D05, and E01) using the molecular docking and molecular dynamics simulation approach, to retrieve information regarding binding properties to PC.ResultsAfter adjustment for confounders, IgG and IgG2 anti-PC showed some significant associations, but IgG1 anti-PC was much stronger as a protection marker. IgG1 anti-PC was associated with an increased risk of CVD below 33rd, 25th, and 10th percentile and of stroke below 33rd and 25th, and of myocardial infarction (MI) below 10th percentile. Among men, a strong association with stroke was determined below the 33rd percentile [HR 9.20, CI (2.22–38.12); p = 0.0022]. D05 clone has higher binding affinity followed by E01 and A01 using molecular docking and further have been confirmed during the course of 100 ns simulation. The stability of the D05 clone with PC was substantially higher.ConclusionIgG1 anti-PC was a stronger protection marker than IgG anti-PC and IgG2 anti-PC and also separately for men. The molecular modeling approach helps in identifying the intrinsic properties of anti-PC clones and atomistic interactions with PC.
Collapse
Affiliation(s)
- Shailesh Kumar Samal
- Section of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Solna, Sweden
| | - Pritam Kumar Panda
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Uppsala, Sweden
| | - Max Vikström
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Solna, Sweden
| | - Karin Leander
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Solna, Sweden
| | - Ulf de Faire
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Solna, Sweden
| | - Rajeev Ahuja
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Uppsala, Sweden
- Department of Physics, Indian Institute of Technology Ropar, Rupnagar, India
| | - Johan Frostegård
- Section of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Solna, Sweden
- *Correspondence: Johan Frostegård,
| |
Collapse
|
16
|
Yuan C, Wu M, Chen X, Li C, Zhang A, Lu W. Growth Performance and Hematological Changes in Growing Sika Deers Fed with Spent Mushroom Substrate of Pleurotus ostreatus. Animals (Basel) 2022; 12:ani12060765. [PMID: 35327162 PMCID: PMC8944863 DOI: 10.3390/ani12060765] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/16/2022] [Accepted: 03/16/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary With the rapid development of the mushroom industry, a large number of spent mushroom substrate (SMS) has also been produced. SMS can be easily digested by ruminants and is suitable for feeding animals, such as cows, sheep, as well as deer. The results of this study show that the dietary spent mushroom substrate of Pleurotus ostreatus (SMS-MP) has no obvious effect on the physiological condition of growing sika deer, at the same time it can reduce the cost of feeding and avoid environmental pollution caused by improper disposal of SMS-MP. Abstract The purpose of this experiment is to expand the feed of growing sika deer and to explore the effects on growing sika deer of the spent mushroom substrate of Pleurotus ostreatus (SMS-MP). Twelve immature female growing sika deer were randomly assigned to four groups. The ratios of SMS-MP to replace concentration supplements were 0%, 10%, 20%, and 30%, respectively, and the growth performance, feed intake and apparent digestibility, serum biochemical indexes, blood physiological indexes, serum immune globulin and plasma amino acid of growing sika deer were measured. The results of the current study confirmed the applicability of SMS-MP as a feed ingredient in growing sika deer diets. There was no significant change in growth performance and hematology of growing sika deer when the concentrate supplement was replaced with 10–20% SMS-MP. However, replacing 30% of concentrate supplements with SMS-MP in the growing sika deer diet resulted in significantly decreased Hb and HCT levels. It can be concluded that, as a waste resource, adding a small amount of SMS-MP has no significant effect on the growth of sika deer, and at the same time can reduce the consumption of concentrate supplements, thereby improving the economic benefits of sika deer breeding.
Collapse
Affiliation(s)
| | | | | | | | - Aiwu Zhang
- Correspondence: (A.Z.); (W.L.); Tel.: +86-138-441-02196 (W.L.)
| | - Wenfa Lu
- Correspondence: (A.Z.); (W.L.); Tel.: +86-138-441-02196 (W.L.)
| |
Collapse
|
17
|
Wu H, Deng Y, Long D, Yang M, Li Q, Feng Y, Chen Y, Qiu H, Huang X, He Z, Hu L, Yin H, Li G, Guo Y, Du W, Zhao M, Lu L, Lu Q. The IL-21-TET2-AIM2-c-MAF pathway drives the T follicular helper cell response in lupus-like disease. Clin Transl Med 2022; 12:e781. [PMID: 35343082 PMCID: PMC8958352 DOI: 10.1002/ctm2.781] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 03/02/2022] [Accepted: 03/07/2022] [Indexed: 12/14/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease that involves T follicular helper (TFH ) cell-mediated humoral immune responses. Absent in melanoma 2 (human AIM2 and murine Aim2) is a well-known component of the inflammasome in the innate immune system. Surprisingly, we observed that in SLE patients, upregulated levels of AIM2 expression were found in peripheral blood and skin lesions, with the highest levels detected in TFH -like cells. In the CD4cre Aim2fl/fl conditional knockout mice, a markedly reduced TFH cell response was observed, with significantly lower levels of serum autoantibodies and proteinuria, as well as profoundly reduced renal IgG deposition in pristane-induced lupus mice. Mechanistically, IL-21 was found to recruit hydroxymethyltransferase ten-eleven translocation 2 (TET2) to the AIM2 promoter, resulting in DNA demethylation and increased transcription of AIM2. In addition, AIM2 could regulate c-MAF expression to enhance IL-21 production, which consequently promoted TFH cell differentiation. Our results have identified a role of AIM2 in promoting the TFH cell response and further revealed that the IL-21-TET2-AIM2-c-MAF signalling pathway is dysregulated in lupus pathogenesis, which provides a potential therapeutic target for SLE.
Collapse
Affiliation(s)
- Haijing Wu
- Department of DermatologySecond Xiangya HospitalHunan Key Laboratory of Medical EpigenomicsCentral South UniversityChangshaChina
| | - Yaxiong Deng
- Department of DermatologySecond Xiangya HospitalHunan Key Laboratory of Medical EpigenomicsCentral South UniversityChangshaChina
| | - Di Long
- Department of DermatologySecond Xiangya HospitalHunan Key Laboratory of Medical EpigenomicsCentral South UniversityChangshaChina
| | - Ming Yang
- Department of DermatologySecond Xiangya HospitalHunan Key Laboratory of Medical EpigenomicsCentral South UniversityChangshaChina
| | - Qianwen Li
- Department of DermatologySecond Xiangya HospitalHunan Key Laboratory of Medical EpigenomicsCentral South UniversityChangshaChina
| | - Yu Feng
- Department of DermatologySecond Xiangya HospitalHunan Key Laboratory of Medical EpigenomicsCentral South UniversityChangshaChina
| | - Yongjian Chen
- Department of DermatologySecond Xiangya HospitalHunan Key Laboratory of Medical EpigenomicsCentral South UniversityChangshaChina
| | - Hong Qiu
- Department of DermatologySecond Xiangya HospitalHunan Key Laboratory of Medical EpigenomicsCentral South UniversityChangshaChina
| | - Xin Huang
- Department of DermatologySecond Xiangya HospitalHunan Key Laboratory of Medical EpigenomicsCentral South UniversityChangshaChina
| | - Zhenghao He
- Department of DermatologySecond Xiangya HospitalHunan Key Laboratory of Medical EpigenomicsCentral South UniversityChangshaChina
| | - Longyuan Hu
- Department of DermatologySecond Xiangya HospitalHunan Key Laboratory of Medical EpigenomicsCentral South UniversityChangshaChina
| | - Heng Yin
- Department of DermatologySecond Xiangya HospitalHunan Key Laboratory of Medical EpigenomicsCentral South UniversityChangshaChina
| | - Guangdi Li
- Department of Public HealthCentral South UniversityChangshaChina
| | - Yunkai Guo
- Department of Otolaryngology Head and Neck SurgerySecond Xiangya HospitalCentral South UniversityChangshaChina
| | - Wenhan Du
- Department of Pathology and Center for Infection and ImmunologyThe University of Hong KongChongqing International Institute for ImmunologyHong KongChina
| | - Ming Zhao
- Department of DermatologySecond Xiangya HospitalHunan Key Laboratory of Medical EpigenomicsCentral South UniversityChangshaChina
| | - Liwei Lu
- Department of Pathology and Center for Infection and ImmunologyThe University of Hong KongChongqing International Institute for ImmunologyHong KongChina
| | - Qianjin Lu
- Department of DermatologySecond Xiangya HospitalHunan Key Laboratory of Medical EpigenomicsCentral South UniversityChangshaChina
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeInstitute of DermatologyNanjingChina
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesNanjingChina
- Chinese Academy of Medical SciencesJiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjingChina
| |
Collapse
|
18
|
Jha SB, Rivera AP, Flores Monar GV, Islam H, Puttagunta SM, Islam R, Kundu S, Sange I. Systemic Lupus Erythematosus and Cardiovascular Disease. Cureus 2022; 14:e22027. [PMID: 35282557 PMCID: PMC8910778 DOI: 10.7759/cureus.22027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2022] [Indexed: 12/14/2022] Open
|
19
|
Antibodies against phosphorylcholine in hospitalized versus non-hospitalized obese subjects. Sci Rep 2021; 11:20246. [PMID: 34642415 PMCID: PMC8511239 DOI: 10.1038/s41598-021-99615-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/24/2021] [Indexed: 12/18/2022] Open
Abstract
Obesity associates with reduced life expectancy, type 2 diabetes, hypertension and cardiovascular disease, and is characterized by chronic inflammation. Phosphorylcholine (PC) is an epitope on oxidized low-density lipoprotein, dead cells and some microorganisms. Antibodies against PC (anti-PC) have anti-inflammatory properties. Here, we explored the role of anti-PC in hospitalized versus non-hospitalized obese. One-hundred-and-twenty-eight obese (BMI ≥ 30 kg/m2) individuals (59.8 (± 5.5) years, 53.9% women) from the Malmö Diet and Cancer Cardiovascular Cohort were examined and IgM, IgG1 and IgG2 anti-PC were analyzed by ELISA. Individuals with at least one recorded history of hospitalization prior to study baseline were considered hospitalized obese (HO). Associations between IgM, IgG1 and IgG2 anti-PC and HO (n = 32)/non-hospitalized obese (NHO) (n = 96), but also with metabolic syndrome and diabetes were analysed using logistic regressions. Both IgM and IgG1 anti-PC were inversely associated with HO, also after controlling for age and sex. When further adjusted for waist circumference, systolic blood pressure, glucose levels and smoking status, only IgG1 anti-PC remained significantly associated with HO. In multivariate models, each 1 standard deviation of increment in anti-PC IgG1 levels was inversely associated with prevalence of HO (odds ratio 0.57; CI 95% 0.33–0.98; p = 0.044). IgG2 anti-PC did not show any associations with HO. Low levels of IgM and IgG1 anti-PC are associated with higher risk of being a HO individual independent of sex and age, IgG1 anti-PC also independently of diabetes and metabolic syndrome. The anti-inflammatory properties of these antibodies may be related to inflammation in obesity and its complications.
Collapse
|
20
|
Natural Antibodies and Alloreactive T Cells Long after Kidney Transplantation. J Transplant 2021; 2021:7005080. [PMID: 34631160 PMCID: PMC8497134 DOI: 10.1155/2021/7005080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 09/01/2021] [Accepted: 09/21/2021] [Indexed: 11/17/2022] Open
Abstract
Background The relationship between circulating effector memory T and B cells long after transplantation and their susceptibility to immunosuppression are unknown. To investigate the impact of antirejection therapy on T cell-B cell coordinated immune responses, we assessed IFN-γ-producing memory cells and natural antibodies (nAbs) that potentially bind to autoantigens on the graft. Methods Plasma levels of IgG nAbs to malondialdehyde (MDA) were measured in 145 kidney transplant recipients at 5-7 years after transplantation. In 54 of these patients, the number of donor-reactive IFN-γ-producing cells was determined. 35/145 patients experienced rejection, 18 of which occurred within 1 year after transplantation. Results The number of donor-reactive IFN-γ-producing cells and the levels of nAbs were comparable between rejectors and nonrejectors. The nAbs levels were positively correlated with the number of donor-reactive IFN-γ-producing cells (r s = 0.39, p=0.004). The positive correlation was only observed in rejectors (r s = 0.53, p=0.003; nonrejectors: r s = 0.24, p=0.23). Moreover, we observed that intravenous immune globulin treatment affected the level of nAbs and this effect was found in patients who experienced a late ca-ABMR compared to nonrejectors (p=0.008). Conclusion The positive correlation found between alloreactive T cells and nAbs in rejectors suggests an intricate role for both components of the immune response in the rejection process. Treatment with intravenous immune globulin impacted nAbs.
Collapse
|
21
|
Cardiovascular disease in systemic lupus erythematosus. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2021; 2:157-172. [PMID: 35880242 PMCID: PMC9242526 DOI: 10.2478/rir-2021-0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 09/08/2021] [Indexed: 11/21/2022]
Abstract
There is a well-known increased risk for cardiovascular disease that contributes to morbidity and mortality in systemic lupus erythematosus (SLE). Major adverse cardiovascular events and subclinical atherosclerosis are both increased in this patient population. While traditional cardiac risk factors do contribute to the increased risk that is seen, lupus disease-related factors, medications, and genetic factors also impact the overall risk. SLE-specific inflammation, including oxidized lipids, cytokines, and altered immune cell subtypes all are likely to play a role in the pathogenesis of atherosclerotic plaques. Research is ongoing to identify biomarkers that can help clinicians to predict which SLE patients are at the greatest risk for cardiovascular disease (CVD). While SLE-specific treatment regimens for the prevention of cardiovascular events have not been identified, current strategies include minimization of traditional cardiac risk factors and lowering of overall lupus disease activity.
Collapse
|
22
|
Porsch F, Mallat Z, Binder CJ. Humoral immunity in atherosclerosis and myocardial infarction: from B cells to antibodies. Cardiovasc Res 2021; 117:2544-2562. [PMID: 34450620 DOI: 10.1093/cvr/cvab285] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/30/2021] [Accepted: 08/24/2021] [Indexed: 02/06/2023] Open
Abstract
Immune mechanisms are critically involved in the pathogenesis of atherosclerosis and its clinical manifestations. Associations of specific antibody levels and defined B cell subsets with cardiovascular disease activity in humans as well as mounting evidence from preclinical models demonstrate a role of B cells and humoral immunity in atherosclerotic cardiovascular disease. These include all aspects of B cell immunity, the generation of antigen-specific antibodies, antigen presentation and co-stimulation of T cells, as well as production of cytokines. Through their impact on adaptive and innate immune responses and the regulation of many other immune cells, B cells mediate both protective and detrimental effects in cardiovascular disease. Several antigens derived from (oxidised) lipoproteins, the vascular wall and classical autoantigens have been identified. The unique antibody responses they trigger and their relationship with atherosclerotic cardiovascular disease are reviewed. In particular, we focus on the different effector functions of specific IgM, IgG, and IgE antibodies and the cellular responses they trigger and highlight potential strategies to target B cell functions for therapy.
Collapse
Affiliation(s)
- Florentina Porsch
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom.,INSERM U970, Paris Cardiovascular Research Centre, Paris, France.,Unversité Paris Descartes, Sorbonne Paris Cité, Paris France
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
23
|
Ajeganova S, Andersson MLE, Frostegård J, Hafström I. Higher levels of anti-phosphorylcholine autoantibodies in early rheumatoid arthritis indicate lower risk of incident cardiovascular events. Arthritis Res Ther 2021; 23:201. [PMID: 34311770 PMCID: PMC8314464 DOI: 10.1186/s13075-021-02581-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/15/2021] [Indexed: 12/21/2022] Open
Abstract
Background The increased risk of cardiovascular events (CVE) in rheumatoid arthritis (RA) is not fully explained by traditional risk factors. Immuno-inflammatory mechanisms and autoantibodies could be involved in the pathogenesis of atherosclerotic disease. It has been suggested that anti-phosphorylcholine antibodies (anti-PC) of the IgM subclass may have atheroprotective effects. Here, we aimed to investigate the association between levels of IgM anti-PC antibodies with CVE in patients with early RA. Methods The study population was derived from the BARFOT early RA cohort, recruited in 1994–1999. The outcome of incident CVE (AMI, angina pectoris, coronary intervention, ischemic stroke, TIA) was tracked through the Swedish Hospital Discharge and the National Cause of Death Registries. Sera collected at inclusion and the 2-year visit were analyzed with ELISA to determine levels of anti-PC IgM. The Kaplan-Meier estimates and Cox proportional hazards regression models were used to compare CV outcome in the groups categorized by baseline median level of IgM anti-PC. Results In all, 653 patients with early RA, 68% women, mean (SD) age 54.8 (14.7) years, DAS28 5.2 (1.3), 68% seropositive, and without prevalent CVD, were included. During the follow-up of mean 11.7 years, 141 incident CVE were recorded. Baseline IgM anti-PC above median was associated with a reduction in risk of incident CVE in patients aged below 55 years at inclusion, HR 0.360 (95% CI, 0.142–0.916); in males, HR 0.558 (0.325–0.958); in patients with BMI above 30 kg/m2, HR 0.235 (0.065–0.842); and in those who did not achieve DAS28 remission at 1 year, HR 0.592 (0.379–0.924). The pattern of associations was confirmed in the models with AUC IgM anti-PC over 2 years. Conclusion Protective effects of higher levels of innate IgM anti-PC autoantibodies on CVE were detected in younger patients with RA and those at high risk of CVE: males, presence of obesity, and non-remission at 1 year.
Collapse
Affiliation(s)
- Sofia Ajeganova
- Division of Gastroenterology and Rheumatology, Department of Medicine Huddinge, Karolinska Institutet, 171 77, Stockholm, Sweden. .,Department of Clinical Sciences, Rheumatology Division, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium.
| | - Maria L E Andersson
- Faculty of Medicine, Department of Rheumatology, Lund University, Lund and Spenshult Research and Development Centre, Halmstad, Sweden
| | - Johan Frostegård
- Section of Immunology and Chronic disease, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ingiäld Hafström
- Division of Gastroenterology and Rheumatology, Department of Medicine Huddinge, Karolinska Institutet, 171 77, Stockholm, Sweden.,Rheumatology Unit, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
24
|
de Vries MR, Ewing MM, de Jong RCM, MacArthur MR, Karper JC, Peters EAB, Nordzell M, Karabina SAP, Sexton D, Dahlbom I, Bergman A, Mitchell JR, Frostegård J, Kuiper J, Ninio E, Jukema JW, Pettersson K, Quax PHA. Identification of IgG1 isotype phosphorylcholine antibodies for the treatment of inflammatory cardiovascular diseases. J Intern Med 2021; 290:141-156. [PMID: 33342002 PMCID: PMC8359267 DOI: 10.1111/joim.13234] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/23/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Phosphorylcholine (PC) is an important pro-inflammatory damage-associated molecular pattern. Previous data have shown that natural IgM anti-PC protects against cardiovascular disease. We aimed to develop a monoclonal PC IgG antibody with anti-inflammatory and anti-atherosclerotic properties. METHODS Using various techniques PC antibodies were validated and optimized. In vivo testing was performed in a femoral artery cuff model in ApoE3*Leiden mice. Safety studies are performed in rats and cynomolgus monkeys. RESULTS A chimeric anti-PC (PC-mAb(T15), consisting of a human IgG1 Fc and a mouse T15/E06 Fab) was produced, and this was shown to bind specifically to epitopes in human atherosclerotic tissues. The cuff model results in rapid induction of inflammatory genes and altered expression of genes associated with ER stress and choline metabolism in the lesions. Treatment with PC-mAb(T15) reduced accelerated atherosclerosis via reduced expression of endoplasmic reticulum stress markers and CCL2 production. Recombinant anti-PC Fab fragments were identified by phage display and cloned into fully human IgG1 backbones creating a human monoclonal IgG1 anti-PC (PC-mAbs) that specifically bind PC, apoptotic cells and oxLDL. Based on preventing macrophage oxLDL uptake and CCL2 production, four monoclonal PC-mAbs were selected, which to various extent reduced vascular inflammation and lesion development. Additional optimization and validation of two PC-mAb antibodies resulted in selection of PC-mAb X19-A05, which inhibited accelerated atherosclerosis. Clinical grade production of this antibody (ATH3G10) significantly attenuated vascular inflammation and accelerated atherosclerosis and was tolerated in safety studies in rats and cynomolgus monkeys. CONCLUSIONS Chimeric anti-PCs can prevent accelerated atherosclerosis by inhibiting vascular inflammation directly and through reduced macrophage oxLDL uptake resulting in decreased lesions. PC-mAb represents a novel strategy for cardiovascular disease prevention.
Collapse
Affiliation(s)
- M. R. de Vries
- From theDeptartment of SurgeryLUMCLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLUMCLeidenThe Netherlands
| | - M. M. Ewing
- From theDeptartment of SurgeryLUMCLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLUMCLeidenThe Netherlands
- Deptartment of CardiologyLUMCLeidenThe Netherlands
| | - R. C. M. de Jong
- From theDeptartment of SurgeryLUMCLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLUMCLeidenThe Netherlands
| | - M. R. MacArthur
- Department of Molecular MetabolismHarvard T.H. Chan School of Public HealthBostonMAUSA
| | - J. C. Karper
- From theDeptartment of SurgeryLUMCLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLUMCLeidenThe Netherlands
| | - E. A. B. Peters
- From theDeptartment of SurgeryLUMCLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLUMCLeidenThe Netherlands
| | | | - S. A. P. Karabina
- INSERM UMR_S 933Hôpital Armand‐TrousseauSorbonne UniversitéParisFrance
| | | | - I. Dahlbom
- Dept. of MedicineKarolinska University Hospital Huddinge and Karolinska InstitutetStockholmSweden
| | | | - J. R. Mitchell
- Department of Molecular MetabolismHarvard T.H. Chan School of Public HealthBostonMAUSA
| | - J. Frostegård
- Dept. of MedicineKarolinska University Hospital Huddinge and Karolinska InstitutetStockholmSweden
| | - J. Kuiper
- Division of BioTherapeuticsLACDRLeidenThe Netherlands
| | - E. Ninio
- INSERM UMR_S 1166‐ICANGenomics and Pathophysiology of Cardiovascular DiseasesInstitute of Cardiometabolism and NutritionPitié‐Salpêtrière HôpitalSorbonne UniversitéParisFrance
| | - J. W. Jukema
- Einthoven Laboratory for Experimental Vascular MedicineLUMCLeidenThe Netherlands
- Deptartment of CardiologyLUMCLeidenThe Netherlands
| | | | - P. H. A. Quax
- From theDeptartment of SurgeryLUMCLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLUMCLeidenThe Netherlands
| |
Collapse
|
25
|
Pluijmert NJ, de Jong RCM, de Vries MR, Pettersson K, Atsma DE, Jukema JW, Quax PHA. Phosphorylcholine antibodies restrict infarct size and left ventricular remodelling by attenuating the unreperfused post-ischaemic inflammatory response. J Cell Mol Med 2021; 25:7772-7782. [PMID: 34190404 PMCID: PMC8358891 DOI: 10.1111/jcmm.16662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/30/2021] [Accepted: 05/08/2021] [Indexed: 12/19/2022] Open
Abstract
Phosphorylcholine is a pro‐inflammatory epitope exposed on apoptotic cells, and phosphorylcholine monoclonal immunoglobulin (Ig)G antibodies (PC‐mAb) have anti‐inflammatory properties. In this study, we hypothesize that PC‐mAb treatment reduces adverse cardiac remodelling and infarct size (IS) following unreperfused transmural myocardial infarction (MI). Unreperfused MI was induced by permanent ligation of the left anterior descending (LAD) coronary artery in hypercholesterolaemic APOE*3‐Leiden mice. Three weeks following MI, cardiac magnetic resonance (CMR) imaging showed a reduced LV end‐diastolic volume (EDV) by 21% and IS by 31% upon PC‐mAb treatment as compared to the vehicle control group. In addition, the LV fibrous content was decreased by 27% and LV wall thickness was better preserved by 47% as determined by histological analysis. Two days following MI, CCL2 concentrations, assessed by use of ELISA, were decreased by 81% and circulating monocytes by 64% as assessed by use of FACS analysis. Additionally, local leucocyte infiltration determined by immunohistological analysis showed a 62% decrease after three weeks. In conclusion, the local and systemic inflammatory responses are limited by PC‐mAb treatment resulting in restricted adverse cardiac remodelling and IS following unreperfused MI. This indicates that PC‐mAb holds promise as a therapeutic agent following MI limiting adverse cardiac remodelling.
Collapse
Affiliation(s)
- Niek J Pluijmert
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rob C M de Jong
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Margreet R de Vries
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Douwe E Atsma
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Paul H A Quax
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
26
|
Samal SK, Fröbert O, Kindberg J, Stenvinkel P, Frostegård J. Potential natural immunization against atherosclerosis in hibernating bears. Sci Rep 2021; 11:12120. [PMID: 34108551 PMCID: PMC8190116 DOI: 10.1038/s41598-021-91679-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 05/31/2021] [Indexed: 12/14/2022] Open
Abstract
Brown bears (Ursus arctos) hibernate for 5-6 months during winter, but despite kidney insufficiency, dyslipidemia and inactivity they do not seem to develop atherosclerosis or cardiovascular disease (CVD). IgM antibodies against phosphorylcholine (anti-PC) and malondialdehyde (anti-MDA) are associated with less atherosclerosis, CVD and mortality in uremia in humans and have anti-inflammatory and other potentially protective properties. PC but not MDA is exposed on different types of microorganisms. We determine anti-PC and anti-MDA in brown bears in summer and winter. Paired serum samples from 12 free ranging Swedish brown bears were collected during hibernation in winter and during active state in summer and analyzed for IgM, IgG, IgG1/2 and IgA anti-PC and anti-MDA by enzyme linked immunosorbent assay (ELISA). When determined as arbitrary units (median set at 100 for summer samples), significantly raised levels were observed in winter for anti-PC subclasses and isotypes, and for IgA anti-PC the difference was striking; 100 IQR (85.9-107.9) vs 782.3, IQR (422.8-1586.0; p < 0.001). In contrast, subclasses and isotypes of anti-MDA were significantly lower in winter except IgA anti-MDA, which was not detectable. Anti-PCs are significantly raised during hibernation in brown bears; especially IgA anti-PC was strikingly high. In contrast, anti-MDA titers was decreased during hibernation. Our observation may represent natural immunization with microorganisms during a vulnerable period and could have therapeutic implications for prevention of atherosclerosis.
Collapse
Affiliation(s)
- Shailesh Kumar Samal
- Division of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ole Fröbert
- Department of Cardiology, Faculty of Health, Örebro University, Örebro, Sweden
| | - Jonas Kindberg
- Norwegian Institute for Nature Research, 7485, Trondheim, Norway.,Department of Wildlife, Fish and Environmental Studies, Swedish University of Agricultural Sciences, 901 83, Umeå, Sweden
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Johan Frostegård
- Division of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
27
|
Bettacchioli E, Le Gaffric C, Mazeas M, Borghi MO, Frostegard J, Barturen G, Makowska Z, Babei S, Lesche R, Meroni PL, Alarcon-Riquelme ME, Renaudineau Y. An elevated polyclonal free light chain level reflects a strong interferon signature in patients with systemic autoimmune diseases. J Transl Autoimmun 2021; 4:100090. [PMID: 33817614 PMCID: PMC8010703 DOI: 10.1016/j.jtauto.2021.100090] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 02/21/2021] [Indexed: 12/17/2022] Open
Abstract
High amount of polyclonal free light chains (FLC) are reported in systemic autoimmune diseases (SAD) and we took advantage of the PRECISESADS study to better characterize them. Serum FLC levels were explored in 1979 patients with SAD (RA, SLE, SjS, Scl, APS, UCTD, MCTD) and 614 healthy controls. Information regarding clinical parameters, disease activity, medications, autoantibodies (Ab) and the interferon α and/or γ scores were recorded. Among SAD patients, 28.4% had raised total FLC (from 12% in RA to 30% in SLE and APS) with a normal kappa/lambda ratio. Total FLC levels were significantly higher in SAD with inflammation, active disease in SLE and SjS, and an impaired pulmonary functional capacity in SSc, while independent from kidney impairment, infection, cancer and treatment. Total FLC concentrations were positively correlated among the 10/17 (58.8%) autoantibodies (Ab) tested with anti-RNA binding protein Ab (SSB, SSA-52/60 kDa, Sm, U1-RNP), anti-dsDNA/nucleosome Ab, rheumatoid factor and negatively correlated with complement fractions C3/C4. Finally, examination of interferon (IFN) expression as a potential driver of FLC overexpression was tested showing an elevated level of total FLC among patients with a high IFNα and IFNγ Kirou's score, a strong IFN modular score, and the detection in the sera of B-cell IFN dependent factors, such as TNF-R1/TNFRSF1A and CXCL10/IP10. In conclusion, an elevated level of FLC, in association with a strong IFN signature, defines a subgroup of SAD patients, including those without renal affectation, characterized by increased disease activity, autoreactivity, and complement reduction.
Collapse
Key Words
- APS, primary antiphospholipid syndrome
- AUC, area under the curve
- Ab, autoantibody
- Autoantibodies
- Autoimmune diseases
- CCP, cyclic citrulinated peptide
- CXCL10, C-X-C motif chemokine 10
- F, female
- FLC, free light chains
- Free light chains
- HC, healthy controls
- IFN, interferon
- Interferon signature
- M, male
- MCTD, mixed connective tissue disease
- MDA, malondialdehyde
- NK, natural killer
- PC, phosphorylcholine
- RA, rheumatoid arthritis
- RF, rheumatoid factor
- RNP, ribonucleoprotein
- ROC, Receiver Operating Characteristics
- SAD, systemic autoimmune diseases
- SD, standard deviation
- SLE, systemic lupus erythematosus
- Scl, systemic sclerosis
- SjS, Sjögren's syndrome
- TH1, T helper type 1
- TNF-R1, tumor necrosis factor receptor 1
- UCTD, undetermined connective tissue disease
- VAS, visual analogical scale
- κ, kappa
- λ, lambda
Collapse
Affiliation(s)
| | | | - Margaux Mazeas
- Laboratory of Immunology and Immunotherapy, CHRU Morvan, Brest, France
| | - Maria Orietta Borghi
- Immunorheumatology Research Laboratory, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Johan Frostegard
- Section of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Guillermo Barturen
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
| | | | | | | | | | - Pier Luigi Meroni
- Immunorheumatology Research Laboratory, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Marta E. Alarcon-Riquelme
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
| | - Yves Renaudineau
- Laboratory of Immunology and Immunotherapy, CHRU Morvan, Brest, France
- Univ Brest, INSERM, LBAI, 29238, Brest Cedex 3, France
- Corresponding author. Laboratory of Immunology and Immunotherapy, CHRU Morvan, Brest, France.
| |
Collapse
|
28
|
Tmoyan NA, Afanasieva OI, Ezhov MV, Klesareva EA, Balakhonova TV, Pokrovsky SN. Lipoprotein(a), Immunity, and Inflammation in Polyvascular Atherosclerotic Disease. J Cardiovasc Dev Dis 2021; 8:jcdd8020011. [PMID: 33513851 PMCID: PMC7911372 DOI: 10.3390/jcdd8020011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 12/24/2022] Open
Abstract
Background and aims: lipoprotein(a) (Lp(a)) is a genetically determined risk factor for coronary artery disease and its complications, although data on the association with other vascular beds and the severity of atherosclerosis is limited. The aim of this study was to evaluate the association of atherosclerosis of various vascular beds with Lp(a), as well as its autoantibodies and generalized inflammatory markers. Material and methods: this study included 1288 adult patients with clinical and imaging examination of three vascular beds (coronary, carotid, and lower limb arteries). Patients were categorized according to the number of affected vascular beds (with at least one atherosclerotic stenosis ≥50%): 0 (n = 339), 1 (n = 470), 2 (n = 315), 3 (n = 164). We assessed blood cell count, lipid profile, C-reactive protein, circulating immune complexes, Lp(a), and its autoantibodies. Results: the number of affected vascular beds was associated with an increasing level of Lp(a) and a lower level of IgM autoantibodies to Lp(a). Hyperlipoproteinemia(a) (Lp(a) ≥ 30 mg/dL) was detected more frequently in patients with atherosclerosis. In logistic regression analysis adjusted for age, sex, hypertension, type 2 diabetes, and smoking, an elevated Lp(a) level was independently associated with stenotic atherosclerosis and lesion severity. There was a positive association of the number of affected vascular beds with C-reactive protein (r = 0.21, p < 0.01) and a negative association with circulating immune complexes (r = −0.29, p < 0.01). The neutrophil-to-lymphocyte ratio was significantly higher and the lymphocyte-to-monocyte ratio was significantly lower in patients with atherosclerosis compared to the controls (p < 0.01). Conclusion: Lp(a), C-reactive protein, circulating immune complexes, and neutrophil-to-lymphocyte ratio are associated with the stenotic atherosclerosis of different vascular beds. Lp(a) levels increase and IgM autoantibodies to Lp(a) decrease with the number of affected vascular beds.
Collapse
Affiliation(s)
- Narek A. Tmoyan
- A.L. Myasnikov Institute of Clinical Cardiology, National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, 121552 Moscow, Russia; (M.V.E.); (T.V.B.)
- Correspondence: ; Tel.: +7-(925)-077-07-70
| | - Olga I. Afanasieva
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, 121552 Moscow, Russia; (O.I.A.); (E.A.K.); (S.N.P.)
| | - Marat V. Ezhov
- A.L. Myasnikov Institute of Clinical Cardiology, National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, 121552 Moscow, Russia; (M.V.E.); (T.V.B.)
| | - Elena A. Klesareva
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, 121552 Moscow, Russia; (O.I.A.); (E.A.K.); (S.N.P.)
| | - Tatiana V. Balakhonova
- A.L. Myasnikov Institute of Clinical Cardiology, National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, 121552 Moscow, Russia; (M.V.E.); (T.V.B.)
- Department of Cardiology, Functional and Ultrasound Diagnostics, Sklifosovsky Institute of Clinical Medicine, Federal State Autonomus Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia
| | - Sergei N. Pokrovsky
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, 121552 Moscow, Russia; (O.I.A.); (E.A.K.); (S.N.P.)
| |
Collapse
|
29
|
Phosphorylcholine Antibodies Preserve Cardiac Function and Reduce Infarct Size by Attenuating the Post-Ischemic Inflammatory Response. JACC Basic Transl Sci 2020; 5:1228-1239. [PMID: 33426378 PMCID: PMC7775955 DOI: 10.1016/j.jacbts.2020.09.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 09/30/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022]
Abstract
Phosphorylcholine is a proinflammatory epitope exposed on the outer membrane of apoptotic cells. This study investigated the modulatory effects of a fully human IgG1 monoclonal antibody directed against phosphorylcholine (PC-mAb) on myocardial remodeling and cardiac function following myocardial ischemia-reperfusion injury. PC-mAb attenuates the immediate post-ischemic inflammatory response by reducing the proinflammatory CCL2 chemokine and circulating Ly-6Chi monocytes. This subsequently enhances the post-ischemic repair process resulting in limited adverse cardiac remodeling and preservation of cardiac function. PC-mAb therapy may be a valid therapeutic approach against myocardial ischemia-reperfusion injury.
Phosphorylcholine monoclonal immunoglobulin G antibody attenuates the immediate post-ischemic inflammatory response by reducing the proinflammatory chemokine (C-C motif) ligand 2 chemokine and circulating Ly-6Chi monocytes. This subsequently enhances the post-ischemic repair process, resulting in limited adverse cardiac remodeling and preservation of cardiac function. Therefore, phosphorylcholine monoclonal immunoglobulin G antibody therapy may be a valid therapeutic approach against myocardial ischemia-reperfusion injury.
Collapse
Key Words
- CCL2, chemokine (C-C motif) ligand 2
- CMR, cardiac magnetic resonance
- EDV, end-diastolic volume
- EF, ejection fraction
- ESV, end-systolic volume
- IS, infarct size
- Ig, immunoglobulin
- LV, left ventricular/ventricle
- MI, myocardial infarction
- MI-R, myocardial ischemia-reperfusion
- PC, phosphorylcholine
- PC-mAb, phosphorylcholine monoclonal immunoglobulin G antibody
- cardiac function
- infarct size
- inflammation
- myocardial infarction
- myocardial ischemia-reperfusion
Collapse
|
30
|
Gambino CM, Accardi G, Aiello A, Caruso C, Carru C, Gioia BG, Guggino G, Rizzo S, Zinellu A, Ciaccio M, Candore G. Uncoupling Protein 2 as genetic risk factor for systemic lupus erythematosus: association with malondialdehyde levels and intima media thickness. Minerva Cardioangiol 2020; 68:609-618. [PMID: 32492982 DOI: 10.23736/s0026-4725.20.05225-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
BACKGROUND Increased oxidative stress potentially leads to accelerated atherosclerosis and, consequently, cardiovascular diseases, the main cause of death in systemic lupus erythematous (SLE). To gain insight into these mechanisms, we studied the association of uncoupling protein (UCP) 2 genetic variants, gene involved in the mitochondrial production of reactive oxygen species, and oxidative stress with SLE and the presence of atherosclerosis. METHODS Genetic analysis of the UCP2 -866G/A and UCP2 Ins/Del polymorphisms was performed in 45 SLE patients and 36 healthy controls by RFLP-PCR. Oxidation status was determined by measuring malondialdehyde (MDA) levels. Presence of subclinical atherosclerosis was investigated by evaluation of intima-media thickness using echo-color-Doppler carotid ultrasound examination. RESULTS Allelic and genotypic frequencies of the SNPs analysed were evaluated by gene count. Significant association was found between UCP2-866A allele and susceptibility for SLE (P=0.001). Higher levels of MDA were found significantly increased in SLE patients (MDA, 5.05±3.36 µmol/L) compared to normal controls (MDA, 2.79±0.89 µmol/L) (P<0.0001). CONCLUSIONS Our results suggest that -866G/A UCP2 polymorphism is associated with SLE causing increased ROS production that, in turn, results in increased MDA levels responsible of accelerated atherosclerosis.
Collapse
Affiliation(s)
- Caterina M Gambino
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Giulia Accardi
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Anna Aiello
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Calogero Caruso
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Bruno G Gioia
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Giuliana Guggino
- Unit of Rheumatology, Paolo Giaccone University Hospital, Palermo, Italy
| | - Sergio Rizzo
- Unit of Transfusion Medicine, Paolo Giaccone University Hospital, Palermo, Italy
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Marcello Ciaccio
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Giuseppina Candore
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy -
| |
Collapse
|
31
|
Ding X, Xiang W, He X. IFN-I Mediates Dysfunction of Endothelial Progenitor Cells in Atherosclerosis of Systemic Lupus Erythematosus. Front Immunol 2020; 11:581385. [PMID: 33262760 PMCID: PMC7686511 DOI: 10.3389/fimmu.2020.581385] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease including the cardiovascular system. Atherosclerosis is the most common cardiovascular complication of SLE and a significant risk factor for morbidity and mortality. Vascular damage/protection mechanism in SLE patients is out of balance, caused by the cascade reaction among oxidative stress, proinflammatory cytokines, Neutrophil Extracellular Traps, activation of B cells and autoantibodies and abnormal T cells. As a precursor cell repairing vascular endothelium, endothelial progenitor cells (EPCs) belong to the protective mechanism and show the reduced number and impaired function in SLE. However, the pathological mechanism of EPCs dysfunction in SLE remains ill-defined. This paper reviews the latest SLE epidemiology and pathogenesis, discusses the changes in the number and function of EPCs in SLE, expounds the role of EPCs in SLE atherosclerosis, and provides new guidance and theoretical basis for exploring novel targets for SLE treatment.
Collapse
Affiliation(s)
- Xuewei Ding
- Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wei Xiang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, NHC Key Laboratory of Control of Tropical diseases (Hainan Medical University), Haikou, China
| | - Xiaojie He
- Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
32
|
The Effect of a 13-Valent Conjugate Pneumococcal Vaccine on Circulating Antibodies Against Oxidized LDL and Phosphorylcholine in Man, A Randomized Placebo-Controlled Clinical Trial. BIOLOGY 2020; 9:biology9110345. [PMID: 33105582 PMCID: PMC7716233 DOI: 10.3390/biology9110345] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/15/2020] [Accepted: 10/17/2020] [Indexed: 12/19/2022]
Abstract
Simple Summary Atherosclerosis is the main underlying mechanism for cardiovascular disease. The main cause for atherosclerosis development is oxidized low density lipoprotein (oxLDL) accumulation in the vessel wall and a subsequent immune response. It has been established that immunoglobulin M antibodies against oxLDL help protect against atherosclerosis. It has been found in mice that vaccination with Streptococcus pneumoniae results in an increase of these protective antibodies and thereby decreases the development of atherosclerosis. In this study, we investigated if this increase of antibodies can be found in human as well. Twenty-four healthy male volunteers were vaccinated with Prevenar-13, a pneumococcal vaccine, using different dosing regimens. An increase in anti-Prevenar antibodies was found, showing that the vaccination worked. However, no increase in protective anti-phosphorylcholine or anti-oxLDL antibodies was observed. This work shows that vaccination against pneumococcal does not seem to be a suitable treatment option to help prevent atherosclerosis development, although further research would be required to test alternative pneumococcal-based vaccines, vaccination regimens or study populations. Abstract In mice vaccination with Streptococcus pneumoniae results in an increase in anti-oxLDL IgM antibodies due to mimicry of anti-phosphorylcholine (present in the cell wall of S. pneumoniae) and anti-oxLDL IgM. In this study we investigated the human translation of this molecular mimicry by vaccination against S. pneumoniae using the Prevenar-13 vaccine. Twenty-four healthy male volunteers were vaccinated with Prevenar-13, either three times, twice or once in a double-blind, placebo-controlled, randomized single center clinical study. Anti-pneumococcal wall, oxLDL and phosphorycholine antibody levels were measured at a fixed serum dilution, as well as circulating lipid levels over the course of 68 weeks. A significant increase in anti-oxLDL IgG and IgM was seen in the group receiving two doses six months apart compared to the placebo. However, these differences were not observed in the groups receiving a single dose, two doses one month apart, or three doses. This study shows that vaccination with Prevenar-13 does not result in robust anti-oxLDL IgM levels in humans. Further research would be required to test alternative pneumococcal-based vaccines, vaccination regimens or study populations, such as cardiovascular disease patients.
Collapse
|
33
|
Kostopoulou M, Nikolopoulos D, Parodis I, Bertsias G. Cardiovascular Disease in Systemic Lupus Erythematosus: Recent Data on Epidemiology, Risk Factors and Prevention. Curr Vasc Pharmacol 2020; 18:549-565. [DOI: 10.2174/1570161118666191227101636] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 11/20/2019] [Accepted: 12/12/2019] [Indexed: 12/22/2022]
Abstract
Systemic Lupus Erythematosus (SLE) is associated with increased risk for accelerated atherosclerosis
and cardiovascular (CV) events including coronary heart disease, cerebrovascular and peripheral
artery disease. CV events occur both early and late during the disease course, with younger
patients being at much higher risk than age-matched counterparts. The risk cannot be fully accounted for
by the increased prevalence of traditional atherosclerotic factors and may be due to pathophysiologic
intermediates such as type I interferons and other inflammatory cytokines, oxidative stress, activated
granulocytes and production of extracellular chromatin traps, antiphospholipid and other autoantibodies
causing dysfunction of lipoproteins, altogether resulting in endothelial injury and pro-atherogenic
dyslipidaemia. These mechanisms may be further aggravated by chronic intake of prednisone (even at
doses <7.5 mg/day), whereas immunomodulatory drugs, especially hydroxychloroquine, may exert antiatherogenic
properties. To date, there is a paucity of randomized studies regarding the effectiveness of
preventative strategies and pharmacological interventions specifically in patients with SLE. Nevertheless,
both the European League Against Rheumatism recommendations and extrapolated evidence from
the general population emphasize that SLE patients should undergo regular monitoring for atherosclerotic
risk factors and calculation of the 10-year CV risk. Risk stratification should include diseaserelated
factors and accordingly, general (lifestyle modifications/smoking cessation, antihypertensive and
statin treatment, low-dose aspirin in selected cases) and SLE-specific (control of disease activity, minimization
of glucocorticoids, use of hydroxychloroquine) preventive measures be applied as appropriate.
Further studies will be required regarding the use of non-invasive tools and biomarkers for CV assessment
and of risk-lowering strategies tailored to SLE.
Collapse
Affiliation(s)
- Myrto Kostopoulou
- 4th Department of Internal Medicine, Attikon University Hospital, Joint Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Dionysis Nikolopoulos
- 4th Department of Internal Medicine, Attikon University Hospital, Joint Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - George Bertsias
- Department of Rheumatology, Clinical Rheumatology and Allergy, University of Crete Medical School, Iraklio, Greece
| |
Collapse
|
34
|
Thiagarajan D, Lundström SL, Pershagen G, Almqvist C, Andolf E, Hedman A, Berg O, Oparina N, Frostegård J. Antibodies against Phosphorylcholine and Malondialdehyde during the First Two Years of Life. THE JOURNAL OF IMMUNOLOGY 2020; 205:2109-2116. [PMID: 32887753 DOI: 10.4049/jimmunol.2000437] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/05/2020] [Indexed: 01/21/2023]
Abstract
Abs against phosphorylcholine (anti-PC) and Abs against malondialdehyde (anti-MDA) may be protective in chronic inflammation, like atherosclerosis and cardiovascular disease. It is not known how they develop early in life. Ab titers were measured using ELISA in healthy women (n = 105; born into life study) and their children. Plasma samples were collected from the mothers before conception and from the children at birth as well as at 1 and 2 y after birth. Extracted Abs were compared using a proteomics de novo sequencing approach. It was observed that children were born with very low levels of IgM anti-PC, whereas IgM anti-MDA was present at birth. Both IgM anti-PC and anti-MDA increased during the first 2 y of life, but IgM anti-PC in contrast to IgM anti-MDA was still significantly lower than in the mothers. IgG anti-PC decreased after 1 y but reached similar levels as mothers' after 2 y, whereas IgG anti-MDA reached similar levels as mothers' already after 1 y. Proteomics peptide sequencing analysis indicated large peptide sequence variation without specific clone expression during the early stage of life compared with the adult stage for which specific peptide sequences dominated. IgM anti-PC levels develop much slower than anti-MDA and are still relatively low at 2 y. We hypothesize that anti-PC is developed by a combination of preprogramming and exposure to the external world, in which infectious agents may play a role. For anti-MDA, preprogramming is likely to play a major role and at an earlier stage than for anti-PC.
Collapse
Affiliation(s)
- Divya Thiagarajan
- Institute of Environmental Medicine, Karolinska Institutet, SE 17177 Stockholm, Sweden
| | - Susanna L Lundström
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE 17177 Stockholm, Sweden
| | - Göran Pershagen
- Institute of Environmental Medicine, Karolinska Institutet, SE 17177 Stockholm, Sweden.,Centre for Occupational and Environmental Medicine, Region Stockholm, SE 17080 Stockholm, Sweden
| | - Catarina Almqvist
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, SE 17177 Stockholm, Sweden.,Pediatric Allergy and Pulmonology Unit, Astrid Lindgren Children's Hospital, Karolinska University Hospital, SE 17164 Stockholm, Sweden; and
| | - Ellika Andolf
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, SE 18288 Stockholm, Sweden
| | - Anna Hedman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, SE 17177 Stockholm, Sweden
| | - Oscar Berg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, SE 17177 Stockholm, Sweden
| | - Nina Oparina
- Institute of Environmental Medicine, Karolinska Institutet, SE 17177 Stockholm, Sweden
| | - Johan Frostegård
- Institute of Environmental Medicine, Karolinska Institutet, SE 17177 Stockholm, Sweden;
| |
Collapse
|
35
|
Thiagarajan D, Oparina N, Lundström S, Zubarev R, Sun J, Alarcon-Riquelme M, Frostegård J. IgM antibodies against malondialdehyde and phosphorylcholine in different systemic rheumatic diseases. Sci Rep 2020; 10:11010. [PMID: 32620913 PMCID: PMC7335044 DOI: 10.1038/s41598-020-66981-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 05/24/2020] [Indexed: 01/08/2023] Open
Abstract
IgM antibodies against phosphorylcholine (anti-PC) and malondialdehyde (anti-MDA) may have protective properties in cardiovascular and rheumatic diseases. We here compare these antibodies in systemic rheumatic conditions and study their properties. Anti-PC and anti-MDA was measured using ELISA in patients with SLE (374), RA (354), Mixed connective tissue disease (MCTD, 77), Systemic sclerosis (SSc, 331), Sjögren's syndrome (SjS, 324), primary antiphospholipid syndrome (PAPs, 65), undifferentiated connective tissue disease (UCTD, 118) and 515 matched healthy controls (HC). Cardiovascular score (CV) was broadly defined based on clinical disease symptoms. Anti-PC and anti-MDA peptide/protein characterization were compared using a proteomics de novo sequencing approach. anti-MDA and anti-PC were extracted from total IgM. The proportion of Treg cells was determined by flow cytometry. The maximal difference between cases and controls was shown for MCTD: significantly lower IgM Anti-PC but not anti-MDA among patients (median 49.3RU/ml vs 70.4 in healthy controls, p(t-test) = 0.0037). IgM low levels were more prevalent in MCTD, SLE, SjS, SSc and UCTD. IgM anti-PC variable region profiles were different from and more homologous than anti-MDA. Anti-PC but not anti-MDA were significantly negatively correlated with CV in the whole patient group. In contrast to IgM anti-PC, anti-MDA did not promote polarization of Tregs. Taken together, Anti-PC is decreased in MCTD and also in SLE, SjS and SSc but not in other studied diseases. Anti-PC may thus differentiate between these. In contrast, anti-MDA did not show these differences between diseases studied. Anti-PC level is negatively correlated with CV in the patient group cohort. In contrast to anti-PC, anti-MDA did not promote Treg polarization. These findings could have both diagnostic and therapeutic implications, one possibility being active or passive immunization with PC in some rheumatic conditions.
Collapse
Affiliation(s)
- Divya Thiagarajan
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Nina Oparina
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Susanna Lundström
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Roman Zubarev
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Jitong Sun
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Marta Alarcon-Riquelme
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,GENYO, Center for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Government, Parque tecnolуgico de la salud, 18016, Granada, Spain
| | - Johan Frostegård
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
36
|
Samal SK, Qureshi AR, Rahman M, Stenvinkel P, Frostegård J. Different subclasses and isotypes of antibodies against phosphorylcholine in haemodialysis patients: association with mortality. Clin Exp Immunol 2020; 201:94-104. [PMID: 32297318 PMCID: PMC7290086 DOI: 10.1111/cei.13441] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 12/13/2022] Open
Abstract
The risk of premature death is high among patients on haemodialysis (HD patients). We previously determined that immunoglobulin (Ig)M antibodies against phosphorylcholine (anti-PC) are negatively associated with increased risk of cardiovascular disease (CVD), atherosclerosis, some autoimmune diseases and mortality among HD patients in this cohort. Here, we also study other subclasses and isotypes of anti-PC in HD patients in relation to mortality, inflammation and gender. The study group is a cohort of 209 prevalent HD patients [median age = 66 years, interquartile range (IQR) = 51-74], vintage time = 29 months (IQR = 15-58; 56% men) with a mean follow-up period of 41 months (IQR = 20-60). Fifty-six per cent were men. We also divided patients into inflamed C-reactive protein (CRP) > 5·6 mg/ml and non-inflamed CRP. Antibody levels were determined by in-house enzyme-linked immunosorbent assay. IgG1 anti-PC below median was significantly associated with increased all-cause mortality (after adjustment for confounders: P = 0·02), while IgG, IgA and IgG2 anti-PC were not associated with this outcome. Among non-inflamed patients, IgM and IgG1 anti-PC were significantly associated with mortality (P = 0·047 and 0·02). IgG1 anti-PC was significantly associated with mortality among men (P = 0·03) and trending among women (P = 0·26). IgM (as previously reported) and IgG1 anti-PC are negatively associated with survival among HD patients and non-inflamed HD patients, but among inflamed patients there were no associations. IgG, IgA or IgG2 anti-PC were not associated with survival in these groups and subgroups. Further studies are needed to determine if raising anti-PC levels, especially IgM and IgG1 anti-PC, through immunization is beneficial.
Collapse
Affiliation(s)
- S. K. Samal
- Division of Immunology and Chronic DiseaseInstitute of Environmental MedicineKarolinska InstitutetStockholmSweden
| | - A. R. Qureshi
- Division of Renal MedicineDepartment of Clinical Science, Intervention and TechnologyKarolinska InstitutetStockholmSweden
| | - M. Rahman
- Division of Immunology and Chronic DiseaseInstitute of Environmental MedicineKarolinska InstitutetStockholmSweden
| | - P. Stenvinkel
- Division of Renal MedicineDepartment of Clinical Science, Intervention and TechnologyKarolinska InstitutetStockholmSweden
| | - J. Frostegård
- Division of Immunology and Chronic DiseaseInstitute of Environmental MedicineKarolinska InstitutetStockholmSweden
| |
Collapse
|
37
|
López P, Rodríguez-Carrio J, Martínez-Zapico A, Pérez-Álvarez ÁI, Benavente L, Caminal-Montero L, Suárez A. IgM anti-phosphorylcholine antibodies associate with senescent and IL-17+ T cells in SLE patients with a pro-inflammatory lipid profile. Rheumatology (Oxford) 2020; 59:407-417. [PMID: 31302689 DOI: 10.1093/rheumatology/kez264] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 06/04/2019] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE The aim was to evaluate whether T cell subsets and the lipid profile could be linked to the cardioprotective effect of IgM anti-phosphorylcholine (PC) antibodies in SLE. METHODS Anti-PC antibodies were quantified by ELISA in 197 patients and 99 controls and analysed in relationship to clinical features, treatments and serum lipids. Carotid atheromatosis was evaluated by ultrasonography; Th1, Th17, Treg and CD4+CD28null cells by flow cytometry; and cytokine serum levels by immunoassays, in a subgroup of 120 SLE patients and 33 controls. RESULTS IgM anti-PC serum levels were reduced in SLE patients compared with controls (P < 0.001) and were associated with age (β= -0.252; P = 0.002), high-density lipoprotein (HDL; β = 0.271; P = 0.001), low-density lipoprotein (LDL; β= -0.192; P = 0.017) and glucocorticoid treatment (β= -0.201; P = 0.012), whereas the IgG-to-IgM anti-PC ratio was increased (P = 0.007) and associated with age (β = 0.194; P = 0.028) and SLEDAI (β = 0.250; P = 0.005). Also, patients with clinical or subclinical cardiovascular disease exhibited reduced IgM anti-PC levels compared with their cardiovascular disease-free counterparts, regardless of glucocorticoid usage (P = 0.001). CD4+CD28null and Th17 cells were increased in SLE patients compared with controls (P < 0.01) and correlated inversely with IgM anti-PC levels. These associations were observed in patients displaying high triglyceride or low HDL levels, even after adjusting for clinical parameters and treatments (CD4+CD28null: β = -0.455, P = 0.001; Th17: β= -0.280, P = 0.035), but not in those with a normal lipid profile. High triglyceride and low HDL profiles were related to low IgM anti-PC and Treg levels, respectively, whereas both lipid profiles were associated with inflammatory markers and cytokines. CONCLUSION The present study provides evidence for an association of IgM anti-PC antibodies with pro-atherogenic T cell subsets in SLE, with a high triglyceride/low HDL lipid profile playing a facilitating major role.
Collapse
Affiliation(s)
- Patricia López
- Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo.,Group of Basic and Translational Research in Inflammatory Diseases, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)
| | - Javier Rodríguez-Carrio
- Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo.,Group of Basic and Translational Research in Inflammatory Diseases, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)
| | - Aleida Martínez-Zapico
- Department of Internal Medicine, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | - Lorena Benavente
- Department of Neurology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Luis Caminal-Montero
- Group of Basic and Translational Research in Inflammatory Diseases, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA).,Department of Internal Medicine, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Ana Suárez
- Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo.,Group of Basic and Translational Research in Inflammatory Diseases, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)
| |
Collapse
|
38
|
Thiagarajan D, Fiskesund R, Frostegård A, Steen J, Rahman M, Vikström M, Lundström S, Frostegård J. Immunoglobulin G1 Antibodies Against Phosphorylcholine Are Associated With Protection in Systemic Lupus Erythematosus and Atherosclerosis: Potential Underlying Mechanisms. ACR Open Rheumatol 2020; 2:344-356. [PMID: 32392632 PMCID: PMC7301871 DOI: 10.1002/acr2.11127] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 02/10/2020] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE Immunoglobulin M antibodies against phosphorylcholine (anti-PCs) may be protective in atherosclerosis, cardiovascular disease (CVD), and systemic lupus erythematosus (SLE). We study immunoglobulin G1 (IgG1) and immunoglobulin G2 (IgG2) anti-PCs, with a focus on atherosclerosis and SLE. METHODS We determined anti-PCs by using the enzyme-linked immunosorbent assay in 116 patients with SLE and 110 age- and sex-matched controls. For functional studies, we used three in-house-generated, fully human monoclonal IgG1 anti-PCs (A01, D05, and E01). Apoptosis was induced in Jurkat T cells and preincubated with A01, D05, E01, or IgG1 isotype control, and effects on efferocytosis by human macrophages were studied. Anti-PC peptide/protein characterization was determined using a proteomics de novo sequencing approach. RESULTS IgG1, but not IgG2, anti-PC levels were higher among patients with SLE (P = 0.02). IgG1 anti-PCs were negatively associated with Systemic Lupus International Collaborating Clinics (SLICC) damage index and Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) scores (odds ratio [OR]: 2.978 [confidence interval (CI): 0.876-10.098] and OR: 5.108 [CI 1.3-20.067], respectively) and negatively associated with CVD, atherosclerotic plaques, and echolucent plaques (potentially vulnerable plaques), but the association for the two former was not significant after controlling for confounders. D05 had a maximum effect on macrophage efferocytosis efficiency, followed by A01 and E01. The monoclonal antibodies showed differential binding specificity to PC and PC-associated neoepitopes. A peptide analysis showed a difference in the complementarity-determining region 3 of the three IgG1 anti-PC clones that are crucial for recognition of PC on apoptotic cell surfaces and other neoepitopes. CONCLUSION IgG1 anti-PCs are negatively associated with disease activity and disease damage in SLE, but the negative association with CVD is also dependent on confounding risk factors. One potential underlying mechanism could be increased clearance of dead cells.
Collapse
Affiliation(s)
| | - R Fiskesund
- Karolinska University Hospital, Huddinge, Sweden
| | | | - J Steen
- Karolinska Institutet, Stockholm, Sweden
| | - M Rahman
- Karolinska Institutet, Stockholm, Sweden
| | - M Vikström
- Karolinska Institutet, Stockholm, Sweden
| | | | | |
Collapse
|
39
|
Dai L, Debowska M, Lukaszuk T, Bobrowski L, Barany P, Söderberg M, Thiagarajan D, Frostegård J, Wennberg L, Lindholm B, Qureshi AR, Waniewski J, Stenvinkel P. Phenotypic features of vascular calcification in chronic kidney disease. J Intern Med 2020; 287:422-434. [PMID: 31823455 DOI: 10.1111/joim.13012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/30/2019] [Accepted: 11/05/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Patients with chronic kidney disease stage 5 (CKD5) are predisposed to vascular calcification (VC), but the combined effect of factors associated with VC was sparsely investigated. We applied the relaxed linear separability (RLS) feature selection model to identify features that concomitantly associate with VC in CKD5 patients. METHODS Epigastric arteries collected during surgery from living donor kidney transplant recipients were examined to score the histological extent of medial VC. Sixty-two phenotypic features in 152 patients were entered into RLS model to differentiate between no-minimal VC (n = 93; score 0-1) and moderate-extensive VC (n = 59; score 2-3). The subset of features associated with VC was selected on the basis of cross-validation procedure. The strength of association of the selected features with VC was expressed by the absolute value of 'RLS factor'. RESULTS Among 62 features, a subset of 17 features provided optimal prediction of VC with 89% of patients correctly classified into their groups. The 17 features included traditional risk factors (diabetes, age, cholesterol, BMI and male sex) and markers of bone metabolism, endothelial function, metabolites, serum antibodies and mitochondrial-derived peptide. Positive RLS factors range from 1.26 to 4.05 indicating features associated with increased risk of VC, and negative RLS factors range from -0.95 to -1.83 indicating features associated with reduced risk of VC. CONCLUSION The RLS model identified 17 features including novel biomarkers and traditional risk factors that together concomitantly associated with medial VC. These results may inform further investigations of factors promoting VC in CKD5 patients.
Collapse
Affiliation(s)
- L Dai
- From the, Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - M Debowska
- Department for Mathematical Modeling of Physiological Processes, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - T Lukaszuk
- Faculty of Computer Science, Bialystok University of Technology, Bialystok, Poland
| | - L Bobrowski
- Department for Mathematical Modeling of Physiological Processes, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland.,Faculty of Computer Science, Bialystok University of Technology, Bialystok, Poland
| | - P Barany
- From the, Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - M Söderberg
- Pathology, Clinical Pharmacology and Safety Sciences, AstraZeneca R&D, Gothenburg, Sweden
| | - D Thiagarajan
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - J Frostegård
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - L Wennberg
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - B Lindholm
- From the, Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - A R Qureshi
- From the, Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - J Waniewski
- Department for Mathematical Modeling of Physiological Processes, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - P Stenvinkel
- From the, Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
40
|
Samal SK, Qureshi AR, Rahman M, Stenvinkel P, Frostegård J. Antibodies against Malondialdehyde in Haemodialysis Patients and Its Association with Clinical Outcomes: Differences between Subclasses and Isotypes. J Clin Med 2020; 9:jcm9030753. [PMID: 32168733 PMCID: PMC7141181 DOI: 10.3390/jcm9030753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 01/22/2023] Open
Abstract
Patients on haemodialysis (HD-patients) have an increased risk of premature death. Low levels of IgM antibodies against malondialdehyde (anti-MDA) are associated with increased risk of cardiovascular disease (CVD) with underlying potential mechanisms described. Here, we studied subclasses and isotypes of anti-MDA in 210 HD-patients with mortality as outcome (56% men, median age 66, Interquartile range (IQR) 51-74 years, vintage time 29 (15-58) months, mean follow up period of 41 (20-60)months). Patients were also divided into inflamed c-reactive protein (CRP >5.6 mg/mL) and non-inflamed. Antibody levels were measured by ELISA. In multivariate risk analysis, patients in low tertile of IgM anti-MDA sub-distribution hazard ratio (sHR 0.54); 95% confidence interval (CI: 0.34-0.89) inversely and significantly associated with all-cause mortality after five years, after adjusting for confounders. Low tertile of IgG (sHR 0.48, 95%CI: 0.25-0.90, p = 0.02) and IgG1 (sHR 0.50, CI: 0.24-1.04, p = 0.06) was associated low mortality among non-inflamed patients. In contrast, anti-MDA IgG2 among inflamed patients was significantly associated with increased mortality, IgG2(sHR 2.33, CI: 1.16-4.68, p = 0.01). IgM anti-MDA was a novel biomarker among HD-patients with low levels being associated with mortality, while low levels of IgG and IgG1 but not IgA anti-MDA were associated with mortality only among non-inflamed patients. IgG2 anti-MDA was a significant risk marker among inflamed patients, which could be related to infection.
Collapse
Affiliation(s)
- Shailesh Kumar Samal
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, 17177 Stockholm, Sweden; (S.K.S.); (M.R.)
| | - Abdul Rashid Qureshi
- Divisions of Renal Medicine, Department of Clinical Science, Intervention and Technology, 14152 Huddinge, Sweden; (A.R.Q.); (P.S.)
| | - Mizanur Rahman
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, 17177 Stockholm, Sweden; (S.K.S.); (M.R.)
| | - Peter Stenvinkel
- Divisions of Renal Medicine, Department of Clinical Science, Intervention and Technology, 14152 Huddinge, Sweden; (A.R.Q.); (P.S.)
| | - Johan Frostegård
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, 17177 Stockholm, Sweden; (S.K.S.); (M.R.)
- Correspondence:
| |
Collapse
|
41
|
Rheumatoid arthritis patients display B-cell dysregulation already in the naïve repertoire consistent with defects in B-cell tolerance. Sci Rep 2019; 9:19995. [PMID: 31882654 PMCID: PMC6934703 DOI: 10.1038/s41598-019-56279-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/03/2019] [Indexed: 12/17/2022] Open
Abstract
B cells are postulated to be central in seropositive rheumatoid arthritis (RA). Here, we use exploratory mass cytometry (n = 23) and next-generation sequencing (n = 19) to study B-cell repertoire shifts in RA patients. Expression of several B-cell markers were significantly different in ACPA+ RA compared to healthy controls, including an increase in HLA-DR across subsets, CD22 in clusters of IgM+ B cells and CD11c in IgA+ memory. Moreover, both IgA+ and IgG+ double negative (IgD− CD27−) CD11c+ B cells were increased in ACPA+ RA, and there was a trend for elevation in a CXCR5/CCR6high transitional B-cell cluster. In the RA BCR repertoire, there were significant differences in subclass distribution and, notably, the frequency of VH with low somatic hypermutation (SHM) was strikingly higher, especially in IgG1 (p < 0.0001). Furthermore, both ACPA+ and ACPA− RA patients had significantly higher total serum IgA and IgM compared to controls, based on serology of larger cohorts (n = 3494 IgA; n = 397 IgM). The observed elevated Ig-levels, distortion in IgM+ B cells, increase in double negative B cells, change in B-cell markers, and elevation of unmutated IgG+ B cells suggests defects in B-cell tolerance in RA. This may represent an underlying cause of increased polyreactivity and autoimmunity in RA.
Collapse
|
42
|
Tmoyan NA, Afanasieva OI, Ezhov MV, Klesareva EA, Afanasieva MI, Razova OA, Balakhonova TV, Pokrovsky SN. [Lipoprotein(а) Level, Apolipoprotein(а) Polymorphism аnd Autoаntibodies Against Lipoprotein(а) in Patients with Stenotic Cаrotid Atherosclerosis]. ACTA ACUST UNITED AC 2019; 59:20-27. [PMID: 31849309 DOI: 10.18087/cardio.2019.12.n727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 07/20/2019] [Accepted: 09/17/2019] [Indexed: 11/18/2022]
Abstract
Аim. Comparative assessment of respiratory indicators according to multifunctional monitoring (PFM) with the recommended standard for a complete polysomnographic study and an assessment of the effect of blood pressure (BP) measurements in PFM on sleep quality. Triаls on the аssociаtion of Lp(а) and cаrotid аtherosclerosis аre limited. The аim of the study wаs to investigаte the аssociаtion of Lp(а), аpolipoprotein(а) [apo(а)] polymorphism аnd аutoаntibodies to Lp(а) with stenotic (≥50%) cаrotid аtherosclerosis in dependence on CHD presence. Materials and methods. The study included 785 pаtients аt the аge from 21 to 92 with dаtа of instrumentаl exаmination of coronаry, cаrotid аnd lower limbs аrteries. Stenotic cаrotid аtherosclerosis wаs diаgnosed in 447 pаtients who were divided into two groups depending on presence (n=344) or аbsence (n=103) of CHD. The control group comprised of 338 pаtients without stenotic аtherosclerosis of coronаry, cаrotid аnd lower limbs аrteries. In the blood serum of pаtients levels of Lp(а), аutoаntibodies to Lp(а) were determined аnd аlso аpo(а) phenotyping wаs conducted. Results. There were more mаles, higher аverаge аge аnd frequency of hypertension, type 2 diаbetes mellitus, smoking, Lp(а) concentrаtion (mediаn [interquаrtile rаnge]): 30 [11; 63] vs. 14 [5; 30] mg/dl, p<0.01) in the group with stenotic cаrotid аtherosclerosis in compаrison with control group. Besides, Lp(а) level wаs higher in CHD subgroup thаn in pаtients with stenotic cаrotid аtherosclerosis without CHD: 32 [12; 72] vs. 24 [8; 50] mg/dl, respectively, p=0.01. Elevаted (≥30 mg/dl) Lp(а) level, low moleculаr weight аpolipoprotein(а) [(LMW аpo(а)] phenotype were аssociаted with stenotic cаrotid аtherosclerosis (odds rаtio (OR) 2.9; 95% confidence intervаl (CI) 2.1-4.0, p<0.01 аnd OR 2.3; 95% CI 1.6-3.4, p<0.01, respectively). Logistic regression аnаlysis showed independent аssociаtion of elevаted Lp(а) level аnd LMW аpo(а) phenotype with stenotic cаrotid аtherosclerosis both in the presence аnd absence of CHD. The level of IgM аutoаntibodies to Lp(а) wаs higher in control group thаn in pаtients with stenotic cаrotid аtherosclerosis, p=0.02. Conclusion The level of Lp(a) ≥30 mg/dl and low molecular weight phenotype of aprotein(a) are predictors of stenotic atherosclerosis CA, regardless of the presence of coronary heart disease and other risk factors, while a reverse relationship was found between the level of autoantibodies of the IgM class against Lp(a) and the severity of atherosclerosis CA.
Collapse
Affiliation(s)
- N A Tmoyan
- National Medical Research Center for Cardiology
| | | | - M V Ezhov
- National Medical Research Center for Cardiology
| | | | | | - O A Razova
- National Medical Research Center for Cardiology
| | | | | |
Collapse
|
43
|
Emerging role of innate B1 cells in the pathophysiology of autoimmune and neuroimmune diseases: Association with inflammation, oxidative and nitrosative stress and autoimmune responses. Pharmacol Res 2019; 148:104408. [PMID: 31454534 DOI: 10.1016/j.phrs.2019.104408] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 12/16/2022]
|
44
|
Rahman M, Steuer J, Gillgren P, Végvári Á, Liu A, Frostegård J. Malondialdehyde Conjugated With Albumin Induces Pro-Inflammatory Activation of T Cells Isolated From Human Atherosclerotic Plaques Both Directly and Via Dendritic Cell-Mediated Mechanism. JACC Basic Transl Sci 2019; 4:480-494. [PMID: 31468003 PMCID: PMC6712057 DOI: 10.1016/j.jacbts.2019.03.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 03/11/2019] [Accepted: 03/13/2019] [Indexed: 12/14/2022]
Abstract
Human dendritic cells were differentiated from blood monocytes and treated with malondialdehyde (MDA) conjugated with human serum albumin (HSA). Autologous T cells from human plaques or blood were co-cultured with the pre-treated dendritic cells or treated directly. MDA modifications were studied by mass spectrometry. MDA-HSA induced a pro-inflammatory DC-mediated T-cell activation and also a strong direct effect on T cells, inhibited by an inhibitor of oxidative stress and antibodies against MDA. Atherogenic heat shock protein-60 was strongly induced in T cells activated by MDA-HSA. Two peptide modifications in atherosclerotic patients' HSA were similar to those present in in vitro MDA-modified HSA.
Collapse
Key Words
- ATP, adenosine triphosphate
- CVD, cardiovascular disease
- DC, dendritic cell
- GM-CSF, granulocyte-macrophage colony-stimulating factor
- HLA, human leukocyte antigen
- HSA, human serum albumin
- HSP, heat shock protein
- IFN, interferon
- IL, interleukin
- IgM, immunoglobulin M
- LDL, low-density lipoprotein
- MDA, malondialdehyde
- MS, mass spectrometry
- OxLDL, oxidized low-density lipoprotein
- PCR, polymerase chain reaction
- T cells
- TCR, T-cell receptor
- TGF, transforming growth factor
- TLR, Toll-like receptor
- TNF, tumor necrosis factor
- atherosclerosis
- dendritic cells
- malondialdehyde
- oxidized low-density lipoprotein
Collapse
Affiliation(s)
- Mizanur Rahman
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Johnny Steuer
- Section of Vascular Surgery, Department of Surgery, Södersjukhuset, Institution of Clinical Science and Education, Karolinska Institutet, Stockholm, Sweden
| | - Peter Gillgren
- Section of Vascular Surgery, Department of Surgery, Södersjukhuset, Institution of Clinical Science and Education, Karolinska Institutet, Stockholm, Sweden
| | - Ákos Végvári
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Biomedicum, Karolinska Institutet, Stockholm, Sweden
| | - Anquan Liu
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Johan Frostegård
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
45
|
Koulouri V, Koutsilieris M, Mavragani CP. B cells and atherosclerosis in systemic lupus erythematosus. Expert Rev Clin Immunol 2019; 15:417-429. [DOI: 10.1080/1744666x.2019.1571411] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Vasiliki Koulouri
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Michael Koutsilieris
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Clio P. Mavragani
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
46
|
|
47
|
Frostegård J, Hellström C, Nilsson P, Frostegård AG, Ajeganova S. Autoantibody profiling reveals four protein candidate autoantigens associated with systemic lupus erythematosus. Lupus 2018; 27:1670-1678. [PMID: 30041579 DOI: 10.1177/0961203318788153] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Objectives In systemic lupus erythematosus (SLE) there are typically many autoantibodies. The disease heterogeneity could be better understood with discovery of phenotype-specific antigens targeted by autoantibodies. We here aimed to identify novel autoantigens potentially related to SLE disease and a major complication, atherosclerosis. Methods Antigen microarrays were used to profile IgG autoantibody reactivity against 77 protein fragments (20-140 amino acids (aa) long, median 89 aa) produced within the Human Protein Atlas project, in serum samples from SLE patients ( n = 107) and age- and sex-matched population-based controls ( n = 107). Common carotid intima-media thickness, plaque occurrence and echogenicity were determined by B-mode ultrasound. Results We determined significant differences between patients and controls in IgG reactivity against four proteins. In patients compared to controls, there was an increase of IgG reactivity against zinc finger protein 688 (ZNF688), early B cell factor 2 (EBF2), crystallin, alpha B (CRYAB) and tumor necrosis factor receptor superfamily member 13C (TNFRSF13C). Of these four antigens, only anti-ZNF688 was associated with carotid atherosclerosis (plaque occurrence) and vulnerable plaques in SLE. There was a weak association between anti-EBF2 and SLE disease activity but no significant associations were determined for other measured IgG reactivity. Conclusions In this discovery screening we here demonstrate new candidate autoantigens with differential reactivity (reflecting autoantibody levels) in SLE patients and in controls and in relation to atherosclerosis in SLE.
Collapse
Affiliation(s)
- J Frostegård
- 1 Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - C Hellström
- 2 Affinity Proteomics, SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - P Nilsson
- 2 Affinity Proteomics, SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - A G Frostegård
- 1 Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - S Ajeganova
- 3 Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.,4 Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
48
|
Torrente-Segarra V, Monte TCS, Corominas H. Musculoskeletal involvement and ultrasonography update in systemic lupus erythematosus: New insights and review. Eur J Rheumatol 2018; 5:127-130. [PMID: 30183613 DOI: 10.5152/eurjrheum.2017.17198] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 01/13/2018] [Indexed: 12/25/2022] Open
Abstract
Ultrasonography has been rarely used to measure musculoskeletal and joint activity in systemic lupus erythematosus (SLE). The aim of this review is to discuss the utility and reliability of this non-invasive diagnostic tool for the assessment of joint disease in SLE patients. In the last decade, several reports have highlighted the role of ultrasonography for a better evaluation of SLE-related musculoskeletal symptoms. The symptoms have also been associated with worse outcomes in SLE; therefore, it is essential to seek useful and accessible techniques for better understanding of such patients who are insufficiently assessed by standard physical examination.
Collapse
Affiliation(s)
- Vicenç Torrente-Segarra
- Department of Rheumatology, Hospital Sant Joan Despí Moisès Broggi/Hospital General de l' Hospitalet, Barcelona, Spain
| | | | - Hèctor Corominas
- Rheumatology Unit, Hospital Universitari de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
49
|
Chen X, Gustafsson S, Whitington T, Borné Y, Lorentzen E, Sun J, Almgren P, Su J, Karlsson R, Song J, Lu Y, Zhan Y, Hägg S, Svensson P, Smedby KE, Slager SL, Ingelsson E, Lindgren CM, Morris AP, Melander O, Karlsson T, de Faire U, Caidahl K, Engström G, Lind L, Karlsson MCI, Pedersen NL, Frostegård J, Magnusson PKE. A genome-wide association study of IgM antibody against phosphorylcholine: shared genetics and phenotypic relationship to chronic lymphocytic leukemia. Hum Mol Genet 2018; 27:1809-1818. [PMID: 29547969 DOI: 10.1093/hmg/ddy094] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/12/2018] [Indexed: 11/13/2022] Open
Abstract
Phosphorylcholine (PC) is an epitope on oxidized low-density lipoprotein (oxLDL), apoptotic cells and several pathogens like Streptococcus pneumoniae. Immunoglobulin M against PC (IgM anti-PC) has the ability to inhibit uptake of oxLDL by macrophages and increase clearance of apoptotic cells. From our genome-wide association studies (GWASs) in four European-ancestry cohorts, six single nucleotide polymorphisms (SNPs) in 11q24.1 were discovered (in 3002 individuals) and replicated (in 646 individuals) to be associated with serum level of IgM anti-PC (the leading SNP rs35923643-G, combined β = 0.19, 95% confidence interval 0.13-0.24, P = 4.3 × 10-11). The haplotype tagged by rs35923643-G (or its proxy SNP rs735665-A) is also known as the top risk allele for chronic lymphocytic leukemia (CLL), and a main increasing allele for general IgM. By using summary GWAS results of IgM anti-PC and CLL in the polygenic risk score (PRS) analysis, PRS on the basis of IgM anti-PC risk alleles positively associated with CLL risk (explained 0.6% of CLL variance, P = 1.2 × 10-15). Functional prediction suggested that rs35923643-G might impede the binding of Runt-related transcription factor 3, a tumor suppressor playing a central role in the immune regulation of cancers. Contrary to the expectations from the shared genetics between IgM anti-PC and CLL, an inverse relationship at the phenotypic level was found in a nested case-control study (30 CLL cases with 90 age- and sex-matched controls), potentially reflecting reverse causation. The suggested function of the top variant as well as the phenotypic association between IgM anti-PC and CLL risk needs replication and motivates further studies.
Collapse
Affiliation(s)
- Xu Chen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Stefan Gustafsson
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Thomas Whitington
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Yan Borné
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Erik Lorentzen
- Department of Bioinformatics, Gothenburg University, Gothenburg, Sweden
| | - Jitong Sun
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Peter Almgren
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Jun Su
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Robert Karlsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jie Song
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Yi Lu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Statistical Genetics, Genetics and Computational Biology Department, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Yiqiang Zhan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Sara Hägg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Per Svensson
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden.,Department of Cardiology, Södersjukhuset, Stockholm, Sweden
| | - Karin E Smedby
- Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Susan L Slager
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Erik Ingelsson
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden.,Division of Cardiovascular Medicine, Department of Medicine, Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Cecilia M Lindgren
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK.,Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Andrew P Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK.,Department of Biostatistics, University of Liverpool, Liverpool, UK
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Thomas Karlsson
- Health Metrics, Institute of Medicine, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | - Ulf de Faire
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kenneth Caidahl
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Gunnar Engström
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Lars Lind
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Johan Frostegård
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Emergency Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Patrik K E Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
50
|
Hardt U, Larsson A, Gunnarsson I, Clancy RM, Petri M, Buyon JP, Silverman GJ, Svenungsson E, Grönwall C. Autoimmune reactivity to malondialdehyde adducts in systemic lupus erythematosus is associated with disease activity and nephritis. Arthritis Res Ther 2018; 20:36. [PMID: 29482604 PMCID: PMC5827973 DOI: 10.1186/s13075-018-1530-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 01/30/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Immunoglobulin M (IgM) autoreactivity to malondialdehyde (MDA) protein modifications is part of the natural antibody repertoire in health and may have beneficial functions. In contrast, IgG anti-MDA are increased in chronic inflammation and autoimmunity and may instead have pathogenic properties. METHODS Herein, we investigated serum IgG anti-MDA levels by enzyme-linked immunosorbent assay (ELISA) in 398 systemic lupus erythematosus (SLE) patients in the Swedish Karolinska SLE cohort and compared these to findings in 225 US SLE patients from New York University and Johns Hopkins University. RESULTS In two independent cohorts, IgG anti-MDA levels correlated positively with disease activity by the Systemic Lupus Erythematosus Disease Activity Index (SLEDAI; p < 0.0001, Spearman R = 0.3). Meta-analysis found an odds ratio of 2.7 (confidence interval (CI) 1.9-3.9; p < 0.0001) for high anti-MDA IgG levels with active disease (SLEDAI ≥ 6). Furthermore, IgG anti-MDA correlated directly with erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), soluble tumor necrosis factor receptors (sTNFR-1, sTNFR-2), and vascular cell adhesion molecule 1 (VCAM-1) measurements, and inversely with complement factors (C1q, C2, C3, C4). Importantly, IgG anti-MDA levels were significantly elevated in SLE patients with active nephritis (p = 0.0005) and correlated with cystatin C estimated glomerular filtration rate and albuminuria. CONCLUSIONS Elevated IgG anti-MDA in SLE patients was associated with high disease activity, with active lupus nephritis, and with biomarkers of systemic inflammation. This natural antibody reactivity may have potential prognostic utility, and may also actively contribute to pathogenesis.
Collapse
Affiliation(s)
- Uta Hardt
- Department of Medicine, Rheumatology Unit, Karolinska Institutet and Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Anders Larsson
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Iva Gunnarsson
- Department of Medicine, Rheumatology Unit, Karolinska Institutet and Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Robert M Clancy
- Department of Medicine, Division of Rheumatology, NYU School of Medicine, New York, NY, USA
| | - Michelle Petri
- Department of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jill P Buyon
- Department of Medicine, Division of Rheumatology, NYU School of Medicine, New York, NY, USA
| | - Gregg J Silverman
- Department of Medicine, Division of Rheumatology, NYU School of Medicine, New York, NY, USA
| | - Elisabet Svenungsson
- Department of Medicine, Rheumatology Unit, Karolinska Institutet and Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Caroline Grönwall
- Department of Medicine, Rheumatology Unit, Karolinska Institutet and Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden.
| |
Collapse
|