1
|
Frias MA, Pagano S, Bararpour N, Sidibé J, Kamau F, Fétaud-Lapierre V, Hudson P, Thomas A, Lecour S, Strijdom H, Vuilleumier N. People living with HIV display increased anti-apolipoprotein A1 auto-antibodies, inflammation, and kynurenine metabolites: a case-control study. Front Cardiovasc Med 2024; 11:1343361. [PMID: 38414919 PMCID: PMC10896987 DOI: 10.3389/fcvm.2024.1343361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 01/25/2024] [Indexed: 02/29/2024] Open
Abstract
Objective This study aimed to study the relationship between auto-antibodies against apolipoprotein A1 (anti-apoA1 IgG), human immunodeficiency virus (HIV) infection, anti-retroviral therapy (ART), and the tryptophan pathways in HIV-related cardiovascular disease. Design This case-control study conducted in South Africa consisted of control volunteers (n = 50), people living with HIV (PLWH) on ART (n = 50), and untreated PLWH (n = 44). Cardiovascular risk scores were determined, vascular measures were performed, and an extensive biochemical characterisation (routine, metabolomic, and inflammatory systemic profiles) was performed. Methods Anti-apoA1 IgG levels were assessed by an in-house ELISA. Inflammatory biomarkers were measured with the Meso Scale Discovery® platform, and kynurenine pathway metabolites were assessed using targeted metabolomic profiling conducted by liquid chromatography-multiple reaction monitoring/mass spectrometry (LC-MRM/MS). Results Cardiovascular risk scores and vascular measures exhibited similarities across the three groups, while important differences were observed in systemic inflammatory and tryptophan pathways. Anti-apoA1 IgG seropositivity rates were 15%, 40%, and 70% in control volunteers, PLWH ART-treated, and PLWH ART-naïve, respectively. Circulating anti-apoA1 IgG levels were significantly negatively associated with CD4+ cell counts and positively associated with viremia and pro-inflammatory biomarkers (IFNγ, TNFα, MIPα, ICAM-1, VCAM-1). While circulating anti-apoA1 IgG levels were associated with increased levels of kynurenine in both control volunteers and PLWH, the kynurenine/tryptophan ratio was significantly increased in PLWH ART-treated. Conclusion HIV infection increases the humoral response against apoA1, which is associated with established HIV severity criteria and kynurenine pathway activation.
Collapse
Affiliation(s)
- Miguel A. Frias
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
- Department of Medical Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sabrina Pagano
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
- Department of Medical Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Nasim Bararpour
- Faculty Unit of Toxicology, CURML, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Department of Genetics, Stanford University, Stanford, CA, United States
- Stanford Center for Genomics and Personalized Medicine, Stanford, CA, United States
| | - Jonathan Sidibé
- Faculty Unit of Toxicology, CURML, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Festus Kamau
- Centre for Cardiometabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Vanessa Fétaud-Lapierre
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
- Department of Medical Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Peter Hudson
- Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Aurélien Thomas
- Faculty Unit of Toxicology, CURML, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Unit of Forensic Toxicology and Chemistry, CURML, Lausanne and Geneva University Hospitals, Lausanne, Geneva, Switzerland
| | - Sandrine Lecour
- Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Hans Strijdom
- Centre for Cardiometabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
- Department of Medical Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
2
|
Rodríguez-Carrio J, Alperi-López M, López P, Pérez-Álvarez ÁI, Robinson GA, Alonso-Castro S, Amigo-Grau N, Atzeni F, Suárez A. Humoral responses against HDL are linked to lipoprotein traits, atherosclerosis, inflammation and pathogenic pathways during early arthritis stages. Rheumatology (Oxford) 2023; 62:2898-2907. [PMID: 36617161 DOI: 10.1093/rheumatology/kead009] [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: 11/10/2022] [Revised: 12/20/2022] [Accepted: 01/02/2023] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE Chronic inflammation and immune dysregulation are crucial mechanisms for atherosclerosis in RA. Recent evidence suggests a link via humoral responses against high-density lipoproteins (HDL). This study aimed to characterize the specificity, clinical relevance and emergence of humoral responses against HDL along disease course, especially during the earliest phases of arthritis. METHODS IgG and IgM serum levels of antibodies against HDL (anti-HDL) and apolipoprotein A1 (anti-ApoA1) were measured in 82 early RA patients, 14 arthralgia individuals and 96 controls. Established RA patients (n = 42) were included for validation. Atherosclerosis and vascular stiffness were measured by Doppler ultrasound. Lipoprotein content, particle numbers and size were measured by H-NMR. Cytokines were measured by immunoassays. A cardiometabolic-related protein panel was evaluated using high-throughput targeted proteomics. RESULTS Anti-HDL and anti-ApoA1 responses were increased in early RA compared with controls (both P < 0.001) and were comparable to established disease. Only anti-ApoA1 antibodies were increased in arthralgia. IgG anti-HDL and anti-ApoA1 were associated with unfavourable lipoprotein traits in RA and arthralgia, respectively. A similar picture was observed for inflammatory mediators. No associations with clinical features or risk factors were found. IgG anti-HDL were independently associated with atherosclerosis occurrence in early RA, and outperformed patient stratification over conventional algorithms (mSCORE) and their anti-ApoA1 counterparts. Anti-HDL antibodies correlated with proteins involved in immune activation, remodelling and lipid metabolism pathways in early RA. CONCLUSION Humoral responses against HDL particles are an early event along the arthritis course, although quantitative and qualitative differences can be noticed among stages. These differences informed distinct capacities as biomarkers and underlying pathogenic circuits.
Collapse
Affiliation(s)
- Javier Rodríguez-Carrio
- Area of Immunology, Department of Functional Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain
- Area of Metabolism, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Mercedes Alperi-López
- Area of Metabolism, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Department of Rheumatology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Patricia López
- Area of Immunology, Department of Functional Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain
- Area of Metabolism, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | | | - George A Robinson
- Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, London, UK
| | - Sara Alonso-Castro
- Area of Metabolism, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Department of Rheumatology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Núria Amigo-Grau
- Biosfer Teslab, Reus, Spain
- Department of Basic Medical Sciences, Pere Virgili Health Research Institute (IISPV), Universitat Rovira i Virgili (URV), Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBER-DEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Fabiola Atzeni
- Rheumatology Unit, Department of Experimental and Internal Medicine, University of Messina, Messina, Italy
| | - Ana Suárez
- Area of Immunology, Department of Functional Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain
- Area of Metabolism, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| |
Collapse
|
3
|
Tollitt J, Allan SM, Chinnadurai R, Odudu A, Hoadley M, Smith C, Kalra PA. Does previous stroke modify the relationship between inflammatory biomarkers and clinical endpoints in CKD patients? BMC Nephrol 2022; 23:38. [PMID: 35042473 PMCID: PMC8767689 DOI: 10.1186/s12882-021-02625-2] [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: 04/30/2021] [Accepted: 11/17/2021] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Chronic kidney disease (CKD) is an independent risk factor for stroke. Stroke is also an independent risk factor for worse CKD outcomes and inflammation may contribute to this bidirectional relationship. This study aims to investigate inflammatory biomarkers in patients with non-dialysis CKD (ND-CKD) with and without stroke.
Methods
A propensity matched sample from > 3000 Salford Kidney Study (SKS) patients, differentiated by previous stroke at study recruitment, had stored plasma analyzed for interleukin- 6 (IL-6), Von Willebrand Factor (VWF) and C-reactive protein (CRP). Multivariable cox regression analysis investigated associations between inflammation and death, end-stage renal disease (ESRD) and future non-fatal cardiovascular events (NFCVE).
Results
A total of 157 previous stroke patients were compared against 162 non-stroke patients. There were no significant differences in inflammatory biomarkers between the two groups. Previous stroke was associated with greater mortality risk, hazard ratio (HR) (95% CI) was 1.45 (1.07–1.97). Higher inflammatory biomarker concentrations were independently associated with death but not ESRD or NFCVE in the total population. For each 1 standard deviation (SD) increase in log IL-6, VWF and CRP, the HR for all-cause mortality were 1.35 (1.10–1.70), 1.26 (1.05–1.51) and 1.34 (1.12–1.61), respectively. CRP retained its independent association (HR 1.47 (1.15–1.87)) with death in the stroke population.
Conclusion
Previous stroke is an important determinant of mortality. However, the adverse combination of stroke and ND-CKD does not seem to be driven by higher levels of inflammation detected after the stroke event. Biomarkers of inflammation were associated with worse outcome in both stroke and non-stroke ND-CKD patients.
Trial registration
15/NW/0818.
Collapse
|
4
|
Bariatric Surgery Leads to a Reduction in Antibodies to Apolipoprotein A-1: a Prospective Cohort Study. Obes Surg 2021; 32:355-364. [PMID: 34888742 PMCID: PMC8794910 DOI: 10.1007/s11695-021-05738-7] [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: 02/02/2021] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 10/31/2022]
Abstract
PURPOSE Autoantibodies against apolipoprotein A-1 have been associated with cardiovascular disease, poorer CV outcomes and all-cause mortality in obese individuals. The impact of bariatric surgery (BS) on the presence of circulating anti-apoA-1 IgG antibodies is unknown. This study aimed to determine the effect of bariatric surgery on auto-antibodies titres against Apolipoprotein A-1 (anti-apoA-1 IgG), looking for changes associated with lipid parameters, insulin resistance, inflammatory profile and percentage of excess body mass index loss (%EBMIL). MATERIALS AND METHODS We assessed 55 patients (40 women) before, 6 and 12 months post-operatively. Baseline and post-operative clinical history and measurements of body mass index (BMI), serum cholesterol, triglycerides, high- and low-density lipoprotein cholesterol (HDL-C and LDL-C), apoA-1, highly sensitive C-reactive protein (hsCRP), fasting glucose (FG), glycated haemoglobin (HbA1c) and HOMA-IR were taken at each point. Human anti-apoA-1 IgG were measured by ELISA. RESULTS The mean age of participants was 50 years. BS significantly improved BMI, %EBMIL triglycerides, HDL-C, apoA-1, hsCRP, HBA1c, FG and HOMA-IR. Baseline anti-apoA-1 IgG seropositivity was 25% and was associated with lower apoA-1 and higher hsCRP levels. One year after BS, anti-apoA-1 IgG seropositivity decreased to 15% (p = 0.007) and median anti-apoA-1 IgG values decreased from 0.70 (0.56-0.84) to 0.47 (0.37-0.61) AU (p < 0.001). Post-operative anti-apoA-1 IgG levels were significantly associated with a decreased post-surgical %EBMIL at 1 year. CONCLUSION Bariatric surgery results in significant reduction in anti-apoA-1 IgG levels, which may adversely influence weight loss. The exact mechanisms underpinning these results are elusive and require further study before defining any clinical recommendations.
Collapse
|
5
|
HDL in Atherosclerotic Cardiovascular Disease: In Search of a Role. Cells 2021; 10:cells10081869. [PMID: 34440638 PMCID: PMC8394469 DOI: 10.3390/cells10081869] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/14/2021] [Accepted: 07/20/2021] [Indexed: 12/17/2022] Open
Abstract
For a long time, high-density lipoprotein cholesterol (HDL-C) has been regarded as a cardiovascular disease (CVD) protective factor. Recently, several epidemiological studies, while confirming low plasma levels of HDL-C as an established predictive biomarker for atherosclerotic CVD, indicated that not only people at the lowest levels but also those with high HDL-C levels are at increased risk of cardiovascular (CV) mortality. This “U-shaped” association has further fueled the discussion on the pathophysiological role of HDL in CVD. In fact, genetic studies, Mendelian randomization approaches, and clinical trials have challenged the notion of HDL-C levels being causally linked to CVD protection, independent of the cholesterol content in low-density lipoproteins (LDL-C). These findings have prompted a reconsideration of the biological functions of HDL that can be summarized with the word “HDL functionality”, a term that embraces the many reported biological activities beyond the so-called reverse cholesterol transport, to explain this lack of correlation between HDL levels and CVD. All these aspects are summarized and critically discussed in this review, in an attempt to provide a background scenario for the “HDL story”, a lipoprotein still in search of a role.
Collapse
|
6
|
Investigation of brain damage mechanism in middle cerebral artery occlusion/reperfusion rats based on i-TRAQ quantitative proteomics. Exp Brain Res 2021; 239:1247-1260. [PMID: 33599834 DOI: 10.1007/s00221-021-06054-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 02/01/2021] [Indexed: 01/04/2023]
Abstract
The objective of this study is to analyze the differential protein expression profile in cerebral cortex of rats with middle cerebral ischemia/reperfusion (MCAO/R), explore the brain damage mechanism of MCAO/R at protein level, and provide experimental foundation for searching specific marker proteins of MCAO/R. Rat model of MCAO/R was established by modified suture-occluded method, and the model was evaluated by the results of brain 2,3,5-triphenyltetrazolium chloride (TTC) and hematoxylin-eosin (HE) staining. Cerebral cortex of rats from sham-operated group (Sham) and MCAO/R groups was used for FASP enzymatic hydrolysis, i-TRAQ quantitative labeling, and reverse-phase liquid chromatography purification and separation. Orbitrap Q Exactive mass spectrometry was used for qualitative and quantitative analyses of total differential protein expression profiles. MCAO/R rats had obvious cerebral infarction lesions, and the relative surface area of cerebral infarction was significantly different compared with sham rats, suggesting that MCAO/R rat model was successfully prepared. There were 199 significant difference proteins (MCAO/R vs Sham, p < 0.05, |fold change|> 1.2), including 104 up-regulated proteins and 95 down-regulated proteins. Gene ontology (GO) enrichment analysis showed that the up-regulated proteins were mainly concentrated in the biological processes of positive regulation of NF-κB transcription and I-κB kinase-NF-κB, etc. Down-regulated proteins were mainly concentrated in long-term synaptic potentiation, cellular response to DNA damage stimulus, etc. KEGG pathway analysis showed that the pathway involved in differential proteins includes oxidative phosphorylation, metabolic pathway, and Ras signaling pathway. Network analysis of differential proteins showed that Alb, ndufb5, ndufs7, ApoB, Cdc42, Ndufa3, Igf1r, P4hb, Mbp, Gc, Nme1, Akt2, and other proteins may play an important role in regulating oxidative stress, apoptosis, and inflammatory response in MCAO/R. Quantitative proteomics based on i-TRAQ labeling reveals the effect of inflammation and apoptosis in brain damage mechanism of MCAO/R. Besides, this research provide some experimental foundation for search and determination of potential therapeutic targets of MCAO/R.
Collapse
|
7
|
Timmerman N, Galyfos G, Sigala F, Thanopoulou K, de Borst GJ, Davidovic L, Eckstein HH, Filipovic N, Grugni R, Kallmayer M, de Kleijn DPV, Koncar I, Mantzaris MD, Marchal E, Matsagkas M, Mutavdzic P, Palombo D, Pasterkamp G, Potsika VT, Andreakos E, Fotiadis DI. The TAXINOMISIS Project: A multidisciplinary approach for the development of a new risk stratification model for patients with asymptomatic carotid artery stenosis. Eur J Clin Invest 2020; 50:e13411. [PMID: 32954520 PMCID: PMC7757200 DOI: 10.1111/eci.13411] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 08/21/2020] [Accepted: 08/23/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Asymptomatic carotid artery stenosis (ACAS) may cause future stroke and therefore patients with ACAS require best medical treatment. Patients at high risk for stroke may opt for additional revascularization (either surgery or stenting) but the future stroke risk should outweigh the risk for peri/post-operative stroke/death. Current risk stratification for patients with ACAS is largely based on outdated randomized-controlled trials that lack the integration of improved medical therapies and risk factor control. Furthermore, recent circulating and imaging biomarkers for stroke have never been included in a risk stratification model. The TAXINOMISIS Project aims to develop a new risk stratification model for cerebrovascular complications in patients with ACAS and this will be tested through a prospective observational multicentre clinical trial performed in six major European vascular surgery centres. METHODS AND ANALYSIS The risk stratification model will compromise clinical, circulating, plaque and imaging biomarkers. The prospective multicentre observational study will include 300 patients with 50%-99% ACAS. The primary endpoint is the three-year incidence of cerebrovascular complications. Biomarkers will be retrieved from plasma samples, brain MRI, carotid MRA and duplex ultrasound. The TAXINOMISIS Project will serve as a platform for the development of new computer tools that assess plaque progression based on radiology images and a lab-on-chip with genetic variants that could predict medication response in individual patients. CONCLUSION Results from the TAXINOMISIS study could potentially improve future risk stratification in patients with ACAS to assist personalized evidence-based treatment decision-making.
Collapse
Affiliation(s)
- Nathalie Timmerman
- Department of Vascular Surgery, Division of Surgical Specialties, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - George Galyfos
- First Propedeutic Department of Surgery, National and Kapodistrian University of Athens, Athens, Greece
| | - Fragiska Sigala
- First Propedeutic Department of Surgery, National and Kapodistrian University of Athens, Athens, Greece
| | - Kalliopi Thanopoulou
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Gert J de Borst
- Department of Vascular Surgery, Division of Surgical Specialties, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Lazar Davidovic
- Clinic for Vascular and Endovascular Surgery, Serbian Clinical Center, Belgrade, Serbia.,School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Hans-Henning Eckstein
- Clinic and Policlinik for vascular and endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Nenad Filipovic
- BioIRC, Research and Development Center for Bioengieering, Kragujevac, Serbia.,Faculty of Engineering, University of Kragujevac, Kragujevac, Serbia
| | | | - Michael Kallmayer
- Clinic and Policlinik for vascular and endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Dominique P V de Kleijn
- Department of Vascular Surgery, Division of Surgical Specialties, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Igor Koncar
- Clinic for Vascular and Endovascular Surgery, Serbian Clinical Center, Belgrade, Serbia.,School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Michalis D Mantzaris
- Department of Materials Science and Engineering, Unit of Medical Technology and Intelligent Information Systems, University of Ioannina, Ioannina, Greece
| | | | - Miltiadis Matsagkas
- Department of Vascular Surgery, Faculty of Medicine, University of Thessaly, Thessaly, Greece
| | - Perica Mutavdzic
- Clinic for Vascular and Endovascular Surgery, Serbian Clinical Center, Belgrade, Serbia
| | - Domenico Palombo
- Division of Vascular and Endovascular Surgery, IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Gerard Pasterkamp
- Division Laboratories and Pharmacy, Laboratory of Clinical Chemistry and Hematology, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Vassiliki T Potsika
- Department of Materials Science and Engineering, Unit of Medical Technology and Intelligent Information Systems, University of Ioannina, Ioannina, Greece
| | - Evangelos Andreakos
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Dimitrios I Fotiadis
- Department of Materials Science and Engineering, Unit of Medical Technology and Intelligent Information Systems, University of Ioannina, Ioannina, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (FORTH), Ioannina, Greece
| | | |
Collapse
|
8
|
Santiago-Raber ML, Montecucco F, Vuilleumier N, Miteva K, Baptista D, Carbone F, Pagano S, Roth A, Burger F, Mach F, Brandt KJ. Atherosclerotic plaque vulnerability is increased in mouse model of lupus. Sci Rep 2020; 10:18324. [PMID: 33110193 PMCID: PMC7591560 DOI: 10.1038/s41598-020-74579-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 10/05/2020] [Indexed: 01/11/2023] Open
Abstract
Anti-apolipoprotein A-1 (anti-apoA-1 IgG) and anti-double stranded DNA (anti-dsDNA IgG) autoantibodies have been described as mediators of atherogenesis in mice and humans. In the present study, we aim to investigate the association between atherosclerotic parameters, autoantibodies and plaque vulnerability in the context of systemic lupus erythematosus (SLE). We therefore bred a lupus prone-mouse model (Nba2.Yaa mice) with Apoe−/− mice resulting in Apoe−/−Nba2.Yaa mice spontaneously producing anti-apoA-1 IgG antibodies. Although Apoe−/−Nba2.Yaa and Apoe−/− mice subject to a high cholesterol diet displayed similar atherosclerosis lesions size in aortic roots and abdominal aorta, the levels of macrophage and neutrophil infiltration, collagen, MMP-8 and MMP-9 and pro-MMP-9 expression in Apoe−/−Nba2.Yaa mice indicated features of atherosclerotic plaque vulnerability. Even though Apoe−/−Nba2.Yaa mice and Apoe−/− mice had similar lipid levels, Apoe−/−Nba2.Yaa mice showed higher anti-apoA-1 and anti-dsDNA IgG levels. Apoe−/−Nba2.Yaa mice displayed a reduction of the size of the kidney, splenomegaly and lymph nodes (LN) hypertrophy. In addition, anti-apoA-1 and anti-dsDNA IgG increased also in relation with mRNA levels of GATA3, IL-4, Bcl-6 and CD20 in the spleen and aortic arch of Apoe−/−Nba2.Yaa mice. Our data show that although atherosclerosis-lupus-prone Apoe−/−Nba2.Yaa mice did not exhibit exacerbated atherosclerotic lesion size, they did show features of atherosclerotic plaque destabilization in correlation with the increase of pro-atherogenic autoantibodies.
Collapse
Affiliation(s)
- Marie-Laure Santiago-Raber
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,AMAL Therapeutics, Av. de la Roseraie 64, 1211, Geneva 4, Switzerland
| | - Fabrizio Montecucco
- Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, 10 Largo Benzi, 16132, Genoa, Italy.,First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy
| | - Nicolas Vuilleumier
- Department of Genetic Medicine, Laboratory and Pathology, Geneva University Hospitals, Geneva, Switzerland.,Division of Laboratory Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Kapka Miteva
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211, Geneva 4, Switzerland
| | - Daniela Baptista
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211, Geneva 4, Switzerland
| | - Federico Carbone
- Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, 10 Largo Benzi, 16132, Genoa, Italy.,First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy
| | - Sabrina Pagano
- Department of Genetic Medicine, Laboratory and Pathology, Geneva University Hospitals, Geneva, Switzerland.,Division of Laboratory Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Aline Roth
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211, Geneva 4, Switzerland
| | - Fabienne Burger
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211, Geneva 4, Switzerland
| | - Francois Mach
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211, Geneva 4, Switzerland
| | - Karim J Brandt
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211, Geneva 4, Switzerland.
| |
Collapse
|
9
|
da Silva RF, Baptista D, Roth A, Miteva K, Burger F, Vuilleumier N, Carbone F, Montecucco F, Mach F, J. Brandt K. Anti-Apolipoprotein A-1 IgG Influences Neutrophil Extracellular Trap Content at Distinct Regions of Human Carotid Plaques. Int J Mol Sci 2020; 21:ijms21207721. [PMID: 33086507 PMCID: PMC7588926 DOI: 10.3390/ijms21207721] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Neutrophils accumulate in atherosclerotic plaques. Neutrophil extracellular traps (NET) were recently identified in experimental atherosclerosis and in complex human lesions. However, not much is known about the NET marker citrullinated histone-3 (H3Cit) expression and functionality in human carotid plaques. Moreover, the association between the proatherosclerotic autoantibody anti-apolipoprotein A-1 (anti-ApoA-1 IgG) and NET has never been investigated. METHODS Atherosclerotic plaques have been obtained from 36 patients with severe carotid stenosis that underwent carotid endarterectomy for severe carotid stenosis. Samples were sectioned into upstream and downstream regions from the same artery segment. Plaque composition and expression of NET markers neutrophil elastase (NE) and H3Cit were quantified by immunohistochemistry. H3Cit expression and function was evaluated by immunofluorescence and confocal analysis in a subset of patients. RESULTS Pathological features of vulnerable phenotypes were exacerbated in plaques developed at downstream regions, including higher accumulation of neutrophils and enhanced expression of NE and H3Cit, as compared to plaques from upstream regions. The H3Cit signal was also more intense in downstream regions, with significant extracellular distribution in spaces outside of neutrophils. The percentage of H3Cit colocalization with CD66b (neutrophils) was markedly lower in downstream portions of carotid plaques, confirming the extrusion of NET in this region. In agreement, the maximum distance of the H3Cit signal from neutrophils, extrapolated from vortex distance calculation in all possible directions, was also higher in downstream plaques. The serum anti-ApoA-1index positively correlated with the expression of H3Cit in downstream segments of plaques. Expression of the H3Cit signal outside of neutrophils and H3Cit maximal distance from CD66b-positive cells increased in plaques from serum positive anti-ApoA-1 patients compared with serum negative patients. CONCLUSION NET elements are differentially expressed in upstream versus downstream regions of human carotid plaques and may be influenced by circulating levels of anti-ApoA-1 IgG. These findings could warrant the investigation of NET elements as potential markers of vulnerability.
Collapse
Affiliation(s)
- Rafaela F. da Silva
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland; (R.F.d.S.); (D.B.); (A.R.); (K.M.); (F.B.); (F.M.)
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, 31270-901 Belo Horizonte, Brazil
| | - Daniela Baptista
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland; (R.F.d.S.); (D.B.); (A.R.); (K.M.); (F.B.); (F.M.)
| | - Aline Roth
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland; (R.F.d.S.); (D.B.); (A.R.); (K.M.); (F.B.); (F.M.)
| | - Kapka Miteva
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland; (R.F.d.S.); (D.B.); (A.R.); (K.M.); (F.B.); (F.M.)
| | - Fabienne Burger
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland; (R.F.d.S.); (D.B.); (A.R.); (K.M.); (F.B.); (F.M.)
| | - Nicolas Vuilleumier
- Department of Diagnostics, Division of Laboratory Medicine, Geneva University Hospitals, 1211 Geneva, Switzerland;
- Department of Medical Specialities, Division of Laboratory Medicine, Faculty of Medicine, 1211 Geneva, Switzerland
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, viale Benedetto XV n6, 16132 Genoa, Italy; (F.C.); (F.M.)
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Largo Rosanna Benzi n10, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, viale Benedetto XV n6, 16132 Genoa, Italy; (F.C.); (F.M.)
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Largo Rosanna Benzi n10, 16132 Genoa, Italy
| | - François Mach
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland; (R.F.d.S.); (D.B.); (A.R.); (K.M.); (F.B.); (F.M.)
| | - Karim J. Brandt
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland; (R.F.d.S.); (D.B.); (A.R.); (K.M.); (F.B.); (F.M.)
- Correspondence: ; Tel.: +41-2237-94-647
| |
Collapse
|
10
|
Bonaventura A, Carbone F, Vecchié A, Meessen J, Ferraris S, Beck E, Keim R, Minetti S, Elia E, Ferrara D, Ansaldo AM, Novelli D, Caironi P, Latini R, Montecucco F. The role of resistin and myeloperoxidase in severe sepsis and septic shock: Results from the ALBIOS trial. Eur J Clin Invest 2020; 50:e13333. [PMID: 32585739 DOI: 10.1111/eci.13333] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 06/12/2020] [Accepted: 06/14/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Inflammatory biomarkers are useful in detecting patients with sepsis. The prognostic role of resistin and myeloperoxidase (MPO) has been investigated in sepsis. MATERIALS AND METHODS Plasma resistin and MPO were measured on days 1, 2 and 7 in 957 patients enrolled in the Albumin Italian Outcome Sepsis (ALBIOS) trial. The association between resistin and MPO levels on day 1, 2 and 7 and 90-day mortality was assessed. RESULTS Plasma resistin and MPO concentrations were higher at day 1 and decreased until day 7. Both biomarkers were positively correlated with each other and with physiological parameters. Higher levels of resistin and MPO on day 1 were associated with the development of new organ failures. Patients experiencing death at 90 days showed higher levels of resistin and MPO compared with survivors. At day 1, only MPO in the 4th quartile (Q4), but not resistin, was found to predict 90-day death (adjusted hazard ratio [aHR] 1.55 vs Q1). At day 2, resistin in the Q3 and Q4 predicted a > 40% increase in mortality as also did MPO in the Q4. On day 7, Q4 resistin was able to predict 90-day mortality, while all quartiles of MPO were not. CONCLUSIONS High levels of MPO, but not of resistin, on day 1 were able to predict 90-day mortality. These findings may either suggest that early hyper-activation of neutrophils is detrimental in patients with sepsis or reflect the burden of the inflammatory process caused by sepsis. Further studies are warranted to deepen these aspects (ALBIOS ClinicalTrials.gov Identifier: NCT00707122).
Collapse
Affiliation(s)
- Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.,Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1200 E Marshall St, Richmond, VA, 23298, USA
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino Genova-Italian Cardiovascular Network, Genoa, Italy
| | - Alessandra Vecchié
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.,Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1200 E Marshall St, Richmond, VA, 23298, USA
| | - Jennifer Meessen
- Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | | | - Roberto Keim
- UOC Anestesia Rianimazione e Terapia Intensiva - ASST Bergamo Est - Ospedale Bolognini di Seriate, Seriate, Italy
| | - Silvia Minetti
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Edoardo Elia
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Daniele Ferrara
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Anna Maria Ansaldo
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Deborah Novelli
- Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Pietro Caironi
- SCDU Anestesia e Rianimazione, Azienda Ospedaliero-Universitaria S. Luigi Gonzaga, Orbassano, Italy.,Dipartimento di Oncologia, Università degli Studi di Torino, Turin, Italy
| | - Roberto Latini
- Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genova-Italian Cardiovascular Network, Genoa, Italy.,First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| |
Collapse
|
11
|
Nigolian H, Ribi C, Courvoisier DS, Pagano S, Alvarez M, Trendelenburg M, Huynh-Do U, Vuilleumier N, Dayer JM, Chizzolini C, Roux-Lombard P. Anti-apolipoprotein A-1 autoantibodies correlate with disease activity in systemic lupus erythematosus. Rheumatology (Oxford) 2020; 59:534-544. [PMID: 31377780 DOI: 10.1093/rheumatology/kez306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/21/2019] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES Apolipoprotein A-1 (ApoA-1) is a protein fraction of the high-density lipoproteins with anti-inflammatory and antioxidant properties that play a major role in reverse cholesterol transport. The presence of anti-ApoA-1 IgG has been reported in SLE to be variably associated with disease activity or cardiovascular events (CVEs). We assessed the clinical performance of anti-ApoA-1 IgG and of antibodies directed against its immunodominant F3L1 peptide (F3L1 IgG) in a well-characterized Swiss SLE cohort study. METHODS A total of 354 biological samples and interviews from 176 individuals were studied. SLEDAI, clinical characteristics, anamnestic CVEs and therapy details were recorded. Sera were tested for the presence of anti-ApoA-1 IgG, anti-F3L1 IgG, anti-dsDNA IgG and aPL. RESULTS Anti-ApoA-1 and anti-F3L1 IgG positivity was associated with higher SLEDAI, mostly due to concomitant positivity of dsDNA IgG and low complement. Variations in time of anti-ApoA-1 IgG correlated positively with variations of anti-dsDNA IgG and inversely to variations of C3 levels. No cross-reactivity was found between anti-ApoA-1 and anti-dsDNA IgG. Positivity for anti-Apo-A1 IgG was more frequent in individuals receiving 10 mg/day or more of prednisone. We did not find any significant association between anti-ApoA-1 IgG positivity and CVEs. CONCLUSION Anti-ApoA-1 and anti-F3L1 IgG in SLE correlate strongly with laboratory markers of activity, particularly with the presence and titre of dsDNA IgG. These results confirm and extend previous findings and support the use of anti-ApoA1 IgG in the clinical setting. Their role in CVEs deserves further investigation.
Collapse
Affiliation(s)
- Haïg Nigolian
- Division of Immunology and Allergy, University Hospital and School of Medicine, Geneva, Switzerland
| | - Camillo Ribi
- Division of Immunology and Allergy, University Hospital of Lausanne and Lausanne University, Lausanne, Switzerland
| | | | - Sabrina Pagano
- Division of Laboratory Medicine, Diagnostic Department, Switzerland.,Department of Internal Medicine Specialties, University Hospital and School of Medicine, Geneva, Switzerland
| | - Montserrat Alvarez
- Division of Immunology and Allergy, University Hospital and School of Medicine, Geneva, Switzerland
| | - Marten Trendelenburg
- Laboratory for Clinical Immunology, Department of Biomedicine and Division of Internal Medicine, University Hospital of Basel, Basel, Switzerland
| | - Uyen Huynh-Do
- Division of Nephrology and Hypertension, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Diagnostic Department, Switzerland.,Department of Internal Medicine Specialties, University Hospital and School of Medicine, Geneva, Switzerland
| | | | - Carlo Chizzolini
- Division of Immunology and Allergy, University Hospital and School of Medicine, Geneva, Switzerland
| | - Pascale Roux-Lombard
- Division of Immunology and Allergy, University Hospital and School of Medicine, Geneva, Switzerland
| |
Collapse
|
12
|
Satta N, Frias MA, Vuilleumier N, Pagano S. Humoral Immunity Against HDL Particle: A New Perspective in Cardiovascular Diseases? Curr Pharm Des 2020; 25:3128-3146. [PMID: 31470782 DOI: 10.2174/1381612825666190830164917] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 08/24/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND Autoimmune diseases are closely associated with cardiovascular diseases (CVD). Over the last decades, the comprehension of atherosclerosis, the principal initiator of CVD, evolved from a lipidcentered disease to a predominant inflammatory and immune response-driven disease displaying features of autoimmunity against a broad range of auto-antigens, including lipoproteins. Among them, high density lipoproteins (HDL) are important actors of cholesterol transport and bear several anti-atherogenic properties, raising a growing interest as therapeutic targets to decrease atherosclerosis and CVD burden, with nevertheless rather disappointing results so far. Reflecting HDL composition complexity, autoimmune responses and autoantibodies against various HDL components have been reported. RESULTS In this review, we addressed the important complexity of humoral autoimmunity towards HDL and particularly how this autoimmune response could help improving our understanding of HDL biological implication in atherosclerosis and CVD. We also discussed several issues related to specific HDL autoantibody subclasses characteristics, including etiology, prognosis and pathological mechanisms according to Rose criteria. CONCLUSION Finally, we addressed the possible clinical value of using these antibodies not only as potential biomarkers of atherogenesis and CVD, but also as a factor potentially mitigating the benefit of HDL-raising therapies.
Collapse
Affiliation(s)
- Nathalie Satta
- Division of Laboratory Medicine, Department of Diagnostic, Geneva University Hospitals, 4 rue Gabrielle Perret-Gentil, 1205 Geneva, Switzerland.,Department of Medical Specialties, Faculty of Medicine, University of Geneva, 1 rue Michel Servet, 1211 Geneva, Switzerland
| | - Miguel A Frias
- Division of Laboratory Medicine, Department of Diagnostic, Geneva University Hospitals, 4 rue Gabrielle Perret-Gentil, 1205 Geneva, Switzerland.,Department of Medical Specialties, Faculty of Medicine, University of Geneva, 1 rue Michel Servet, 1211 Geneva, Switzerland
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Department of Diagnostic, Geneva University Hospitals, 4 rue Gabrielle Perret-Gentil, 1205 Geneva, Switzerland.,Department of Medical Specialties, Faculty of Medicine, University of Geneva, 1 rue Michel Servet, 1211 Geneva, Switzerland
| | - Sabrina Pagano
- Division of Laboratory Medicine, Department of Diagnostic, Geneva University Hospitals, 4 rue Gabrielle Perret-Gentil, 1205 Geneva, Switzerland.,Department of Medical Specialties, Faculty of Medicine, University of Geneva, 1 rue Michel Servet, 1211 Geneva, Switzerland
| |
Collapse
|
13
|
IgG Anti-High Density Lipoprotein Antibodies Are Elevated in Abdominal Aortic Aneurysm and Associated with Lipid Profile and Clinical Features. J Clin Med 2019; 9:jcm9010067. [PMID: 31888089 PMCID: PMC7019833 DOI: 10.3390/jcm9010067] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 12/21/2019] [Accepted: 12/24/2019] [Indexed: 02/06/2023] Open
Abstract
High-density lipoproteins cholesterol (HDLc) levels are decreased in abdominal aortic aneurysm (AAA), which is hallmarked by autoimmunity and lipid aortic deposits. To investigate whether IgG anti-HDL antibodies were present in AAA and their potential association with clinical features, IgG anti-HDL and total IgG along with HDLc plasma levels were measured in 488 AAA patients and 184 controls from the Viborg Vascular (VIVA) study, and in tissue-conditioned media from AAA intraluminal thrombus and media layer samples compared to control aortas. Higher IgG anti-HDL levels were found in AAA compared to controls, even after correcting for total IgG, and after adjusting for potential confounders. IgG anti-HDL levels were correlated with aortic diameter in univariate and adjusted multivariate analyses. IgG anti-HDL antibodies were negatively associated with HDLc levels before and after correcting for potential confounders. Increased anti-HDL antibodies were identified in tissue-conditioned media from AAA samples compared to healthy aortas, with higher levels being observed in the media layer. In conclusion, increased IgG anti-HDL levels (both in plasma and in tissue) are linked to AAA, associated with aortic diameter and HDLc levels. These data suggest a potential immune response against HDL in AAA and support an emerging role of anti-HDL antibodies in AAA.
Collapse
|
14
|
Anti-ApoA-1 IgGs in Familial Hypercholesterolemia Display Paradoxical Associations with Lipid Profile and Promote Foam Cell Formation. J Clin Med 2019; 8:jcm8122035. [PMID: 31766415 PMCID: PMC6947407 DOI: 10.3390/jcm8122035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 11/18/2019] [Indexed: 02/06/2023] Open
Abstract
AIMS Anti-Apolipoprotein A-1 autoantibodies (anti-ApoA-1 IgG) promote atherogenesis via innate immune receptors, and may impair cellular cholesterol homeostasis (CH). We explored the presence of anti-ApoA-1 IgG in children (5-15 years old) with or without familial hypercholesterolemia (FH), analyzing their association with lipid profiles, and studied their in vitro effects on foam cell formation, gene regulation, and their functional impact on cholesterol passive diffusion (PD). METHODS Anti-ApoA-1 IgG and lipid profiles were measured on 29 FH and 25 healthy children. The impact of anti-ApoA-1 IgG on key CH regulators (SREBP2, HMGCR, LDL-R, ABCA1, and miR-33a) and foam cell formation detected by Oil Red O staining were assessed using human monocyte-derived macrophages. PD experiments were performed using a validated THP-1 macrophage model. RESULTS Prevalence of high anti-ApoA-1 IgG levels (seropositivity) was about 38% in both study groups. FH children seropositive for anti-ApoA-1 IgG had significant lower total cholesterol LDL and miR-33a levels than those who were seronegative. On macrophages, anti-ApoA-1 IgG induced foam cell formation in a toll-like receptor (TLR) 2/4-dependent manner, accompanied by NF-kB- and AP1-dependent increases of SREBP-2, LDL-R, and HMGCR. Despite increased ABCA1 and decreased mature miR-33a expression, the increased ACAT activity decreased membrane free cholesterol, functionally culminating to PD inhibition. CONCLUSIONS Anti-ApoA-1 IgG seropositivity is frequent in children, unrelated to FH, and paradoxically associated with a favorable lipid profile. In vitro, anti-ApoA-1 IgG induced foam cell formation through a complex interplay between innate immune receptors and key cholesterol homeostasis regulators, functionally impairing the PD cholesterol efflux capacity of macrophages.
Collapse
|
15
|
Sciascia S, Cecchi I, Radin M, Rubini E, Suárez A, Roccatello D, Rodríguez-Carrio J. IgG Anti-high-Density Lipoproteins Antibodies Discriminate Between Arterial and Venous Events in Thrombotic Antiphospholipid Syndrome Patients. Front Med (Lausanne) 2019; 6:211. [PMID: 31612138 PMCID: PMC6775216 DOI: 10.3389/fmed.2019.00211] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/13/2019] [Indexed: 11/13/2022] Open
Abstract
Introduction: Recurrent thrombotic events are a hallmark of Antiphospholipid Syndrome (APS). However, biomarkers to identify if a patient with antiphospholipid antibodies (aPL) is at higher risk to develop an arterial or a venous event are lacking. Recently, the pathogenic role of anti-high-density lipoproteins antibodies (anti-HDL) in the occurrence of cardiovascular disease (CVD) in autoimmunity has emerged. The aim of the present study was to evaluate the presence of IgG anti-HDL antibodies in a cohort of thrombotic APS patients and to investigate their association with clinical outcomes. Methods: Serum levels of IgG anti-HDL antibodies, total IgG, and complete aPL profile were assessed in 60 APS patients and 80 healthy donors (HDs) by immunoassays. Results: Higher levels of IgG anti-HDL were found in APS patients compared to HDs (p < 0.001), even after correcting for total IgG levels (p < 0.001). No associations with treatments or traditional cardiovascular risk factors, except for smoking habit (p < 0.0001), were found. Patients who experienced at least one arterial event (n = 30) had significantly higher levels of anti-HDL antibodies when compared to patients with venous thrombosis (n = 30, p = 0.046), this difference being stronger when adjusting for total IgG (p = 0.007). Additionally, patients tested positive for antiphosphatidylserine/prothrombin (IgG/IgM) antibodies had significantly higher levels of anti-HDL antibodies (p = 0.045). Conclusions: Increased levels of IgG anti-HDL antibodies can be found in APS, mainly in patients with arterial thrombosis, independently of aPL antibodies and traditional risk factors. These findings point to a role of anti-HDL antibodies in APS and support their use as a potential biomarker for arterial thrombotic events.
Collapse
Affiliation(s)
- Savino Sciascia
- Center of Research of Immuno-pathology and Rare Diseases, Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, and SCDU Nephrology and Dialysis, Department of Clinical and Biological Sciences, S. Giovanni Bosco Hospital, University of Turin, Turin, Italy
| | - Irene Cecchi
- Center of Research of Immuno-pathology and Rare Diseases, Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, and SCDU Nephrology and Dialysis, Department of Clinical and Biological Sciences, S. Giovanni Bosco Hospital, University of Turin, Turin, Italy
| | - Massimo Radin
- Center of Research of Immuno-pathology and Rare Diseases, Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, and SCDU Nephrology and Dialysis, Department of Clinical and Biological Sciences, S. Giovanni Bosco Hospital, University of Turin, Turin, Italy
| | - Elena Rubini
- Center of Research of Immuno-pathology and Rare Diseases, Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, and SCDU Nephrology and Dialysis, Department of Clinical and Biological Sciences, S. Giovanni Bosco Hospital, University of Turin, Turin, Italy
| | - Ana Suárez
- Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Dario Roccatello
- Center of Research of Immuno-pathology and Rare Diseases, Coordinating Center of Piemonte and Valle d'Aosta Network for Rare Diseases, and SCDU Nephrology and Dialysis, Department of Clinical and Biological Sciences, S. Giovanni Bosco Hospital, University of Turin, Turin, Italy.,Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo, Oviedo, Spain
| | - Javier Rodríguez-Carrio
- Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain.,Bone and Mineral Research Unit, Instituto Reina Sofía de Investigación Nefrológica, REDinREN del ISCIII, Hospital Universitario Central de Asturias, Oviedo, Spain
| |
Collapse
|
16
|
Relationship between HDL Cholesterol Efflux Capacity, Calcium Coronary Artery Content, and Antibodies against ApolipoproteinA-1 in Obese and Healthy Subjects. J Clin Med 2019; 8:jcm8081225. [PMID: 31443207 PMCID: PMC6722652 DOI: 10.3390/jcm8081225] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/07/2019] [Accepted: 08/12/2019] [Indexed: 11/16/2022] Open
Abstract
AIMS To explore the associations between cholesterol efflux capacity (CEC), coronary artery calcium (CAC) score, Framingham risk score (FRS), and antibodies against apolipoproteinA-1 (anti-apoA-1 IgG) in healthy and obese subjects (OS). METHODS AND RESULTS ABCA1-, ABCG1-, passive diffusion (PD)-CEC and anti-apoA-1 IgG were measured in sera from 34 controls and 35 OS who underwent CAC score determination by chest computed tomography. Anti-apoA-1 IgG ability to modulate CEC and macrophage cholesterol content (MCC) was tested in vitro. Controls and OS displayed similar ABCG1-, ABCA1-, PD-CEC, CAC and FRS scores. Logistic regression analyses indicated that FRS was the only significant predictor of CAC lesion. Overall, anti-apoA-1 IgG were significantly correlated with ABCA1-CEC (r = 0.48, p < 0.0001), PD-CEC (r = -0.33, p = 0.004), and the CAC score (r = 0.37, p = 0.03). ABCA1-CEC was correlated with CAC score (r = 0.47, p = 0.004) and FRS (r = 0.18, p = 0.29), while PD-CEC was inversely associated with the same parameters (CAC: r = -0.46, p = 0.006; FRS: score r = -0.40, p = 0.01). None of these associations was replicated in healthy controls or after excluding anti-apoA-1 IgG seropositive subjects. In vitro, anti-apoA-1 IgG inhibited PD-CEC (p < 0.0001), increased ABCA1-CEC (p < 0.0001), and increased MCC (p < 0.0001). CONCLUSIONS We report a paradoxical positive association between ABCA1-CEC and the CAC score, with the latter being inversely associated with PD in OS. Corroborating our clinical observations, anti-apoA-1 IgG enhanced ABCA1 while repressing PD-CEC, leading to MCC increase in vitro. These results indicate that anti-apoA-1 IgG have the potential to interfere with CEC and macrophage lipid metabolism, and may underpin paradoxical associations between ABCA1-CEC and cardiovascular risk.
Collapse
|
17
|
Henson D, Tahhan AS, Nardo D, Quyyumi AA, Venditto VJ. Association Between ApoA-I (Apolipoprotein A-I) Immune Complexes and Adverse Cardiovascular Events-Brief Report. Arterioscler Thromb Vasc Biol 2019; 39:1884-1892. [PMID: 31315438 DOI: 10.1161/atvbaha.119.312964] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The immune response is linked to the progression of atherosclerotic cardiovascular disease (CVD). Free autoantibodies targeting ApoA-I (apolipoprotein A-I) have been identified as a component of the inflammatory milieu in patients and have a moderate association with CVD progression. Based on the presence of these antibodies and the high concentration of circulating ApoA-I, we hypothesized that antibodies bound to ApoA-I as an immune complex would be predictive of incident adverse CVD outcomes. Approach and Results: The presence of ApoA-I/IgG immune complexes (ICs) in plasma was confirmed by ELISA in 3 subject cohorts. Characterization of the protein components of ApoAI/IgG ICs indicate that ICs are not correlated with total ApoA-I concentration and are enriched in the anti-inflammatory subclass, IgG4, relative to total plasma IgG (>30% versus 6%). In 359 patients with coronary artery disease (CAD), there were 71 incident adverse CVD events (death, myocardial infarction, and stroke) during a median 4.1-year follow-up. In Cox proportional hazard regression analysis, low levels of ApoA-I/IgG ICs were independent predictors of adverse cardiovascular outcomes after adjustment for age, sex, diabetes mellitus, estimated glomerular filtration rate, presence of obstructive CAD, heart failure, total cholesterol, and HDL (high-density lipoprotein) cholesterol (adjusted hazard ratio of 1.90 [95% CI, 1.03-3.49; P=0.038] between the lowest and the highest tertiles). CONCLUSIONS Low levels of ApoA-I/IgG ICs are associated with an increased risk of adverse events in patients with CAD, raising their potential to be used as a biomarker to predict CVD progression.
Collapse
Affiliation(s)
- David Henson
- From the Department of Pharmaceutical Sciences, University of Kentucky, Lexington (D.H., D.N., V.J.V.)
| | - Ayman Samman Tahhan
- Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA (A.S.T., A.A.Q.)
| | - David Nardo
- From the Department of Pharmaceutical Sciences, University of Kentucky, Lexington (D.H., D.N., V.J.V.)
| | - Arshed Ali Quyyumi
- Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA (A.S.T., A.A.Q.)
| | - Vincent J Venditto
- From the Department of Pharmaceutical Sciences, University of Kentucky, Lexington (D.H., D.N., V.J.V.)
| |
Collapse
|
18
|
Non-Linear Relationship between Anti-Apolipoprotein A-1 IgGs and Cardiovascular Outcomes in Patients with Acute Coronary Syndromes. J Clin Med 2019; 8:jcm8071002. [PMID: 31324073 PMCID: PMC6679072 DOI: 10.3390/jcm8071002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 06/26/2019] [Accepted: 07/08/2019] [Indexed: 12/13/2022] Open
Abstract
Autoantibodies against apolipoprotein A-I (anti-apoA-I IgGs) are prevalent in atherosclerosis-related conditions. It remains elusive whether they improve the prognostic accuracy of the Global Registry of Acute Coronary Events (GRACE) score 2.0 (GS) in acute coronary syndromes (ACS). In this prospective multicenter registry, 1713 ACS patients were included and followed for 1 year. The primary endpoint (major adverse cardiovascular events (MACE)) was defined as the composite of myocardial infarction, stroke (including transient ischemic attack), or cardiovascular (CV) death with individual events independently adjudicated. Plasma levels of anti-apoA-I IgGs upon study inclusion were assessed using ELISA. The association between anti-apoA-I IgGs and incident MACE was assessed using Cox models with splines and C-statistics. One-year MACE incidence was 8.4% (144/1713). Anti-apoA-I IgG levels were associated with MACE with a non-linear relationship (p = 0.01), which remained unchanged after adjusting for the GS (p = 0.04). The hazard increased progressively across the two first anti-apoA-I IgG quartiles before decreasing thereafter. Anti-apoA-I IgGs marginally improved the prognostic accuracy of the GS (c-statistics increased from 0.68 to 0.70). In this multicenter study, anti-apoA-I IgGs were predictive of incident MACE in ACS independently of the GS but in a nonlinear manner. The practical implications of these findings remain to be defined.
Collapse
|
19
|
Anderson JLC, Pagano S, Virzi J, Dullaart RPF, Annema W, Kuipers F, Bakker SJL, Vuilleumier N, Tietge UJF. Autoantibodies to Apolipoprotein A-1 as Independent Predictors of Cardiovascular Mortality in Renal Transplant Recipients. J Clin Med 2019; 8:jcm8070948. [PMID: 31261925 PMCID: PMC6679113 DOI: 10.3390/jcm8070948] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/24/2019] [Accepted: 06/27/2019] [Indexed: 12/15/2022] Open
Abstract
Renal transplant recipients (RTRs) are known to have a high cardio-vascular disease (CVD) burden only partly explained by traditional CVD risk factors. The aim of this paper was therefore to determine: i) the prognostic value of autoantibodies against apoA-1 (anti-apoA-1 IgG) for incidence of CVD mortality, all-cause mortality and graft failure in RTR. Four hundred and sixty two (462) prospectively included RTRs were followed for 7.0 years. Baseline anti-apoA-1 IgG were determined and associations with incidence of CVD mortality (n = 48), all-cause mortality (n = 92) and graft failure (n = 39) were tested. Kaplan-Meier analyses demonstrated significant associations between tertiles of anti-apoA-1 IgG and CVD mortality (log rank test: p = 0.048). Adjusted Cox regression analysis showed a 54% increase in risk for CVD mortality for each anti-apoA-1 IgG levels standard deviation increase (hazard ratio [HR]: 1.54, 95% Confidence Interval [95%CI]: 1.14-2.05, p = 0.005), and a 33% increase for all-cause mortality (HR: 1.33; 95%CI: 1.06-1.67, p = 0.01), independent of CVD risk factors, renal function and HDL function. The association with all-cause mortality disappeared after excluding cases of CVD specific mortality. The sensitivity, specificity, positive predictive value, and negative predictive value of anti-apoA-1 positivity for CVD mortality were 18.0%, 89.3%, 17.0%, and 90.0%, respectively. HDL functionality was not associated with anti-apoA-1 IgG levels. This prospective study demonstrates that in RTR, anti-apoA-1 IgG are independent predictors of CVD mortality and are not associated with HDL functionality.
Collapse
Affiliation(s)
- Josephine L C Anderson
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, 1205 Groningen, The Netherlands
| | - Sabrina Pagano
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospital, 1205 Geneva, Switzerland
- Department of Medical Specialties, Faculty of Medicine, Geneva University, 1205 Geneva, Switzerland
| | - Julien Virzi
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospital, 1205 Geneva, Switzerland
- Department of Medical Specialties, Faculty of Medicine, Geneva University, 1205 Geneva, Switzerland
| | - Robin P F Dullaart
- Department of Endocrinology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Wijtske Annema
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, 1205 Groningen, The Netherlands
- Institute of Clinical Chemistry, University Hospital of Zurich and University of Zurich, 8006 Zurich, Switzerland
| | - Folkert Kuipers
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, 1205 Groningen, The Netherlands
- Laboratory Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Stephan J L Bakker
- Department of Nephrology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospital, 1205 Geneva, Switzerland
| | - Uwe J F Tietge
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, 1205 Groningen, The Netherlands.
- Division of Clinical Chemistry, Department of Laboratory Medicine H5, Karolinska Institutet, 14183 Stockholm, Sweden.
- Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, SE-141 86 Stockholm, Sweden.
| |
Collapse
|
20
|
Lagerstedt JO, Dalla-Riva J, Marinkovic G, Del Giudice R, Engelbertsen D, Burlin J, Petrlova J, Lindahl M, Bernfur K, Melander O, Nilsson J, Schiopu A. Anti-ApoA-I IgG antibodies are not associated with carotid artery disease progression and first-time cardiovascular events in middle-aged individuals. J Intern Med 2019; 285:49-58. [PMID: 30028049 DOI: 10.1111/joim.12817] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE IgG antibodies against apolipoprotein A-I (ApoA-I) have been found to be elevated in subjects from the general population with clinically manifest cardiovascular disease and in myocardial infarction patients with an adverse prognosis. Here, we investigated whether these antibodies are prospectively associated with carotid artery disease progression and with the risk for first-time cardiovascular events in individuals with no previous history of cardiovascular disease. APPROACH AND RESULTS We selected 383 subjects from the cardiovascular cohort of Malmö Diet and Cancer study who suffered a coronary event during a median follow-up period of 15.4 (10.3-16.4) years and 395 age- and sex-matched controls. None of the study participants had a previous history of coronary artery disease or stroke. Anti-ApoA-I IgG were measured by ELISA in serum samples collected at baseline. Intima-media thickness (IMT) was measured in the common carotid artery and in the carotid bifurcation at baseline and after 15.9 (±1.5) years. We found no associations between anti-ApoA-I IgG and carotid artery IMT at baseline or with IMT progression during follow-up. In Cox proportional hazards analyses adjusted for traditional cardiovascular risk factors, the hazard ratio (HR 95%CI) for the primary outcome, incident coronary events, was 0.97 (0.75-1.25), P = 0.782, in subjects with anti-ApoA-I IgG within the highest tertile compared with the lowest tertile. Similarly, we did not find any associations with the secondary outcome, incident first-time stroke. CONCLUSIONS Serum autoantibodies against ApoA-I do not correlate with disease progression and adverse events in cardiovascular disease-free individuals from the general population.
Collapse
Affiliation(s)
- J O Lagerstedt
- Medical Protein Science Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - J Dalla-Riva
- Medical Protein Science Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - G Marinkovic
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - R Del Giudice
- Medical Protein Science Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - D Engelbertsen
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - J Burlin
- Medical Protein Science Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - J Petrlova
- Medical Protein Science Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - M Lindahl
- Medical Protein Science Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - K Bernfur
- Department of Biochemistry and Structural Biology, Lund University, Lund, Sweden
| | - O Melander
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - J Nilsson
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - A Schiopu
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.,Department of Cardiology, Skåne University Hospital Malmö, Malmö, Sweden
| |
Collapse
|
21
|
Carbone F, Rigamonti F, Burger F, Roth A, Bertolotto M, Spinella G, Pane B, Palombo D, Pende A, Bonaventura A, Liberale L, Vecchié A, Dallegri F, Mach F, Montecucco F. Serum levels of osteopontin predict major adverse cardiovascular events in patients with severe carotid artery stenosis. Int J Cardiol 2018; 255:195-199. [PMID: 29317141 DOI: 10.1016/j.ijcard.2018.01.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 12/01/2017] [Accepted: 01/02/2018] [Indexed: 01/08/2023]
|
22
|
Serum matrix metalloproteinase-8, tissue inhibitor of metalloproteinase and myeloperoxidase in ischemic stroke. Atherosclerosis 2018; 271:9-14. [PMID: 29453088 DOI: 10.1016/j.atherosclerosis.2018.02.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/15/2018] [Accepted: 02/07/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Matrix metalloproteinase (MMP)-8 and myeloperoxidase (MPO) may contribute to cerebral damage in acute ischemic stroke. We tested the hypothesis that levels of MPO, MMP-8 and the ratio between MMP-8 and its regulator, tissue inhibitor of metalloproteinase (TIMP-1), are increased in acute ischemic stroke and its etiologic subgroups and they correlate with stroke severity. METHODS In a cross-sectional case-control study, serum concentrations of MMP-8, MPO and TIMP-1 were assessed within 24 h after admission in 470 first-ever ischemic stroke patients and 809 age- and sex-matched controls, randomly selected from the population. Odds ratios (OR) per decade of log transformed dependent variables were calculated and adjusted for age, sex and vascular risk factors. RESULTS Levels of MMP-8 (OR 4.9; 95% CI 3.4-7.2), MMP-8/TIMP-1 ratio (3.0; 2.2-4.1) and MPO (6.6; 4.0-11.0) were independently associated with ischemic stroke. MMP-8 levels differed between etiologic stroke subgroups (p = 0.019, ANOVA), with higher levels in cardioembolic stroke and stroke due to large vessel disease, and lower levels in microangiopathic stroke. MMP-8, MMP-8/TIMP-1 ratio and MPO (p < 0.001) concentrations showed positive associations with stroke severity independent of stroke etiology. CONCLUSIONS Concentrations of serum neutrophil markers are increased after ischemic stroke and associate with stroke severity and etiology. The value of these biomarkers in diagnostics and prognostics is worth being evaluated.
Collapse
|
23
|
Bridge SH, Pagano S, Jones M, Foster GR, Neely D, Vuilleumier N, Bassendine MF. Autoantibody to apolipoprotein A-1 in hepatitis C virus infection: a role in atherosclerosis? Hepatol Int 2018; 12:17-25. [PMID: 29423541 PMCID: PMC5814532 DOI: 10.1007/s12072-018-9842-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 01/08/2018] [Indexed: 12/22/2022]
Abstract
Background/purpose One to three per cent of the world’s population has hepatitis C virus (HCV) infection, which is not only a major cause of liver disease and cancer but also associated with an increased risk of atherosclerosis, despite an ostensibly favourable lipid profile. Autoantibodies are frequent in HCV infection and emerging evidence shows that autoantibodies could be valuable for cardiovascular disease (CVD) risk stratification. This study investigated a novel independent biomarker of CVD, autoantibodies to apolipoprotein A-1 (anti-apoA-1 IgG) and lipids in patients with chronic HCV before, during and after direct-acting anti-viral (DAA) therapy. Methods Eighty-nine blinded serum samples from 27 patients with advanced chronic HCV were assayed for lipids and anti-apoA-1 IgG by ELISA. Results Pre-treatment HCV viral load correlated with high-density lipoprotein cholesterol (HDL-C, r = 0.417; p = 0.042) and negatively with apolipoprotein (apo)B (r = − 0.497; p = 0.013) and markers of CVD risk, the apoB/apoA-1 ratio (r = − 0.490; p = 0.015) and triglyceride level (TG)/HDL-C ratio (r = − 0.450; p = 0.031). Fourteen (52%) of 27 patients had detectable anti-apoA-1 IgG autoantibodies pre-treatment; only two became undetectable with virological cure. Autoantibody-positive sera had lower apoA-1 (p = 0.012), HDL-C (p = 0.009) and total cholesterol (p = 0.006) levels. Conclusions This is the first report of the presence of an emerging biomarker for atherosclerosis, anti-apoA-1 IgG, in some patients with HCV infection. It may be induced by apoA-1 on the surface of HCV lipoviral particles. The autoantibodies inversely correlate with apoA-1 and HDL levels and may render HDL dysfunctional. Whether these hypothesis-generating findings have clinical implications in HCV patients requires further study.
Collapse
Affiliation(s)
- Simon H Bridge
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK.,Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Sabrina Pagano
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Meleri Jones
- The Liver Unit, Blizard Institute, Queen Mary University of London, London, UK
| | - Graham R Foster
- The Liver Unit, Blizard Institute, Queen Mary University of London, London, UK
| | - Dermot Neely
- Department of Clinical Biochemistry, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Margaret F Bassendine
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK. .,Department of Hepatology and Gastroenterology, Imperial College London, 10th Floor QEQM Wing, St. Mary's Hospital Campus, South Wharf Street, London, W2 1NY, UK.
| |
Collapse
|
24
|
Woudberg NJ, Pedretti S, Lecour S, Schulz R, Vuilleumier N, James RW, Frias MA. Pharmacological Intervention to Modulate HDL: What Do We Target? Front Pharmacol 2018; 8:989. [PMID: 29403378 PMCID: PMC5786575 DOI: 10.3389/fphar.2017.00989] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/22/2017] [Indexed: 12/24/2022] Open
Abstract
The cholesterol concentrations of low-density lipoprotein (LDL) and high-density lipoprotein (HDL) have traditionally served as risk factors for cardiovascular disease. As such, novel therapeutic interventions aiming to raise HDL cholesterol have been tested in the clinical setting. However, most trials led to a significant increase in HDL cholesterol with no improvement in cardiovascular events. The complexity of the HDL particle, which exerts multiple physiological functions and is comprised of a number of subclasses, has raised the question as to whether there should be more focus on HDL subclass and function rather than cholesterol quantity. We review current data regarding HDL subclasses and subclass-specific functionality and highlight how current lipid modifying drugs such as statins, cholesteryl ester transfer protein inhibitors, fibrates and niacin often increase cholesterol concentrations of specific HDL subclasses. In addition this review sets out arguments suggesting that the HDL3 subclass may provide better protective effects than HDL2.
Collapse
Affiliation(s)
- Nicholas J. Woudberg
- Hatter Institute for Cardiovascular Research in Africa and South African Medical Research Council Inter-University Cape Heart Group, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sarah Pedretti
- Hatter Institute for Cardiovascular Research in Africa and South African Medical Research Council Inter-University Cape Heart Group, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Division of Endocrinology, Diabetes, Hypertension and Nutrition, Department of Internal Medicine Specialities, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa and South African Medical Research Council Inter-University Cape Heart Group, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Richard W. James
- Division of Endocrinology, Diabetes, Hypertension and Nutrition, Department of Internal Medicine Specialities, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Miguel A. Frias
- Division of Endocrinology, Diabetes, Hypertension and Nutrition, Department of Internal Medicine Specialities, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
25
|
Satta N, Pagano S, Montecucco F, Gencer B, Mach F, Kaiser L, Calmy A, Vuilleumier N. Anti-apolipoprotein A-1 autoantibodies are associated with immunodeficiency and systemic inflammation in HIV patients. J Infect 2017; 76:186-195. [PMID: 29198606 DOI: 10.1016/j.jinf.2017.11.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 11/23/2017] [Accepted: 11/26/2017] [Indexed: 01/09/2023]
Abstract
OBJECTIVES To determine the existence of autoantibodies against apolipoprotein A-1 (anti-apoA-1 IgG) in HIV patients and explore their association with biological features of HIV infection and different inflammatory biomarkers. We also evaluated their impact on CD4+ lymphocytes survival. METHODS Anti-apoA-1 IgG plasma levels were assessed by ELISA in 237 HIV positive patients from a national prospective cohort with no current lipid-lowering therapy. RESULTS 58% of patients were found positive for anti-apoA-1 IgG and were associated with lower CD4+ counts, but higher viremia and systemic inflammation. Logistic regression analyses indicated that high anti-apoA-1 IgG levels were associated with a 16-fold increased risk of displaying low CD4+ levels, independent of HIV RNA levels and treatment (adjusted Odds ratio [OR]:16.1, 95% Confidence Interval [95%CI]:1.80-143.6; p = 0.01), and a 6-fold increased risk of having a detectable viremia, independent of antiretroviral treatment (OR:5.47; 95% CI:1.63-18.36; p = 0.006). In vitro, anti-apoA-1 IgG induced dose and time-dependent CD4+ apoptosis that was increased by exposure to HIV RNA. CONCLUSIONS In HIV patients, anti-apoA-1 IgG levels are associated with low CD4+ counts, high viremia and a pro-inflammatory systemic profile. Anti-apoA-1 IgG can promote CD4+ lymphocyte apoptosis via undefined pathways.
Collapse
Affiliation(s)
- Nathalie Satta
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, 4 rue Gabrielle Perret-Gentil, 1205 Geneva, Switzerland; Clinical Chemistry and Proteomic Group, Department of Human Protein Sciences, University of Geneva, Geneva, Switzerland.
| | - Sabrina Pagano
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, 4 rue Gabrielle Perret-Gentil, 1205 Geneva, Switzerland; Clinical Chemistry and Proteomic Group, Department of Human Protein Sciences, University of Geneva, Geneva, Switzerland
| | - Fabrizio Montecucco
- First Medical Clinic, Laboratory of Phagocyte Physiopathology and Inflammation, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV 16132 Genoa, Italy; IRCCS AOU San Martino - IST, Genova, largo Benzi 10 16143 Genoa, Italy; Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 9 viale Benedetto XV, 16132 Genoa, Italy
| | - Baris Gencer
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, 64 Avenue de la Roseraie, 1211 Geneva, Switzerland
| | | | - François Mach
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, 64 Avenue de la Roseraie, 1211 Geneva, Switzerland
| | - Laurent Kaiser
- Division of Infectious Diseases and of Laboratory Medicine, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Alexandra Calmy
- Division of Infectious Diseases and of Laboratory Medicine, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, 4 rue Gabrielle Perret-Gentil, 1205 Geneva, Switzerland; Clinical Chemistry and Proteomic Group, Department of Human Protein Sciences, University of Geneva, Geneva, Switzerland
| |
Collapse
|
26
|
Antiochos P, Marques-Vidal P, Virzi J, Pagano S, Satta N, Hartley O, Montecucco F, Mach F, Kutalik Z, Waeber G, Vollenweider P, Vuilleumier N. Impact of CD14 Polymorphisms on Anti-Apolipoprotein A-1 IgG-Related Coronary Artery Disease Prediction in the General Population. Arterioscler Thromb Vasc Biol 2017; 37:2342-2349. [DOI: 10.1161/atvbaha.117.309602] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 10/10/2017] [Indexed: 11/16/2022]
Abstract
Objective—
We aimed to determine whether autoantibodies against apoA-1 (apolipoprotein A-1; anti-apoA-1 IgG) predict incident coronary artery disease (CAD), defined as adjudicated incident myocardial infarction, angina, percutaneous coronary revascularization, or bypass grafting, in the general population. We further investigated whether this association is modulated by a functional CD14 receptor single nucleotide polymorphism.
Approach and Results—
In a prospectively studied, population-based cohort of 5220 subjects (mean age 52.6±10.7 years, 47.4% males), followed over a median period of 5.6 years, subjects positive versus negative for anti-apoA-1 IgG presented a total CAD rate of 3.9% versus 2.8% (
P
=0.077) and a nonfatal CAD rate of 3.6% versus 2.3% (
P
=0.018), respectively. After multivariate adjustment for established cardiovascular risk factors, the hazard ratios of anti-apoA-1 IgG for total and nonfatal CAD were: hazard ratio=1.36 (95% confidence interval, 0.94–1.97;
P
=0.105) and hazard ratio=1.53 (95% confidence interval, 1.03–2.26;
P
=0.034), respectively. In subjects with available genetic data for the C260T
rs2569190
single nucleotide polymorphism in the CD14 receptor gene (n=4247), we observed a significant interaction between anti-apoA-1 IgG and
rs2569190
allele status with regards to CAD risk, with anti-apoA-1 IgG conferring the highest risk for total and nonfatal CAD in non-TT carriers, whereas being associated with the lowest risk for total and nonfatal CAD in TT homozygotes (
P
for interaction =0.011 and
P
for interaction =0.033, respectively).
Conclusions—
Anti-apoA-1 IgG are independent predictors of nonfatal incident CAD in the general population. The strength of this association is dependent on a functional polymorphism of the CD14 receptor gene, a finding suggesting a gene–autoantibody interaction for the development of CAD.
Collapse
Affiliation(s)
- Panagiotis Antiochos
- From the Department of Internal Medicine, University Hospital of Lausanne, Switzerland (P.A., P.M.-V., G.W., P.V.); Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Switzerland (J.V., S.P., N.S., F. Montecucco, N.V.); Department of Human Protein Sciences, Faculty of Medicine, (J.V., S.P., N.S., N.V.), Department of Pathology and Immunology, Faculty of Medicine (O.H.), and Division of Cardiology, Foundation for Medical Researches,
| | - Pedro Marques-Vidal
- From the Department of Internal Medicine, University Hospital of Lausanne, Switzerland (P.A., P.M.-V., G.W., P.V.); Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Switzerland (J.V., S.P., N.S., F. Montecucco, N.V.); Department of Human Protein Sciences, Faculty of Medicine, (J.V., S.P., N.S., N.V.), Department of Pathology and Immunology, Faculty of Medicine (O.H.), and Division of Cardiology, Foundation for Medical Researches,
| | - Julien Virzi
- From the Department of Internal Medicine, University Hospital of Lausanne, Switzerland (P.A., P.M.-V., G.W., P.V.); Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Switzerland (J.V., S.P., N.S., F. Montecucco, N.V.); Department of Human Protein Sciences, Faculty of Medicine, (J.V., S.P., N.S., N.V.), Department of Pathology and Immunology, Faculty of Medicine (O.H.), and Division of Cardiology, Foundation for Medical Researches,
| | - Sabrina Pagano
- From the Department of Internal Medicine, University Hospital of Lausanne, Switzerland (P.A., P.M.-V., G.W., P.V.); Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Switzerland (J.V., S.P., N.S., F. Montecucco, N.V.); Department of Human Protein Sciences, Faculty of Medicine, (J.V., S.P., N.S., N.V.), Department of Pathology and Immunology, Faculty of Medicine (O.H.), and Division of Cardiology, Foundation for Medical Researches,
| | - Nathalie Satta
- From the Department of Internal Medicine, University Hospital of Lausanne, Switzerland (P.A., P.M.-V., G.W., P.V.); Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Switzerland (J.V., S.P., N.S., F. Montecucco, N.V.); Department of Human Protein Sciences, Faculty of Medicine, (J.V., S.P., N.S., N.V.), Department of Pathology and Immunology, Faculty of Medicine (O.H.), and Division of Cardiology, Foundation for Medical Researches,
| | - Oliver Hartley
- From the Department of Internal Medicine, University Hospital of Lausanne, Switzerland (P.A., P.M.-V., G.W., P.V.); Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Switzerland (J.V., S.P., N.S., F. Montecucco, N.V.); Department of Human Protein Sciences, Faculty of Medicine, (J.V., S.P., N.S., N.V.), Department of Pathology and Immunology, Faculty of Medicine (O.H.), and Division of Cardiology, Foundation for Medical Researches,
| | - Fabrizio Montecucco
- From the Department of Internal Medicine, University Hospital of Lausanne, Switzerland (P.A., P.M.-V., G.W., P.V.); Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Switzerland (J.V., S.P., N.S., F. Montecucco, N.V.); Department of Human Protein Sciences, Faculty of Medicine, (J.V., S.P., N.S., N.V.), Department of Pathology and Immunology, Faculty of Medicine (O.H.), and Division of Cardiology, Foundation for Medical Researches,
| | - François Mach
- From the Department of Internal Medicine, University Hospital of Lausanne, Switzerland (P.A., P.M.-V., G.W., P.V.); Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Switzerland (J.V., S.P., N.S., F. Montecucco, N.V.); Department of Human Protein Sciences, Faculty of Medicine, (J.V., S.P., N.S., N.V.), Department of Pathology and Immunology, Faculty of Medicine (O.H.), and Division of Cardiology, Foundation for Medical Researches,
| | - Zoltan Kutalik
- From the Department of Internal Medicine, University Hospital of Lausanne, Switzerland (P.A., P.M.-V., G.W., P.V.); Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Switzerland (J.V., S.P., N.S., F. Montecucco, N.V.); Department of Human Protein Sciences, Faculty of Medicine, (J.V., S.P., N.S., N.V.), Department of Pathology and Immunology, Faculty of Medicine (O.H.), and Division of Cardiology, Foundation for Medical Researches,
| | - Gerard Waeber
- From the Department of Internal Medicine, University Hospital of Lausanne, Switzerland (P.A., P.M.-V., G.W., P.V.); Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Switzerland (J.V., S.P., N.S., F. Montecucco, N.V.); Department of Human Protein Sciences, Faculty of Medicine, (J.V., S.P., N.S., N.V.), Department of Pathology and Immunology, Faculty of Medicine (O.H.), and Division of Cardiology, Foundation for Medical Researches,
| | - Peter Vollenweider
- From the Department of Internal Medicine, University Hospital of Lausanne, Switzerland (P.A., P.M.-V., G.W., P.V.); Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Switzerland (J.V., S.P., N.S., F. Montecucco, N.V.); Department of Human Protein Sciences, Faculty of Medicine, (J.V., S.P., N.S., N.V.), Department of Pathology and Immunology, Faculty of Medicine (O.H.), and Division of Cardiology, Foundation for Medical Researches,
| | - Nicolas Vuilleumier
- From the Department of Internal Medicine, University Hospital of Lausanne, Switzerland (P.A., P.M.-V., G.W., P.V.); Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Switzerland (J.V., S.P., N.S., F. Montecucco, N.V.); Department of Human Protein Sciences, Faculty of Medicine, (J.V., S.P., N.S., N.V.), Department of Pathology and Immunology, Faculty of Medicine (O.H.), and Division of Cardiology, Foundation for Medical Researches,
| |
Collapse
|
27
|
Braunersreuther V, Burger F, Lenglet S, Pelli G, Carbone F, Fraga-Silva R, Stergiopulos N, Monaco C, Mueller C, Pagano S, Dallegri F, Mach F, Vuilleumier N, Montecucco F. Anti-apoA-1 auto-antibodies increase mouse atherosclerotic plaque vulnerability, myocardial necrosis and mortality triggering TLR2 and TLR4. Thromb Haemost 2017; 114:410-22. [PMID: 25879306 DOI: 10.1160/th14-12-1039] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 02/19/2015] [Indexed: 12/12/2022]
Abstract
SummaryAuto-antibodies to apolipoprotein A-1 (anti-apoA-1 IgG) were shown to promote inflammation and atherogenesis, possibly through innate immune receptors signalling. Here, we aimed at investigating the role of Toll-like receptors (TLR) 2 and 4 on anti-apoA-1 IgG-induced athero-sclerotic plaque vulnerability, myocardial necrosis and mortality in mice. Adult male apolipoprotein E knockout (ApoE)-/- (n=72), TLR2-/-ApoE-/- (n=36) and TLR4-/-Apo-/- (n=28) mice were intravenously injected with 50 µg/mouse of endotoxin-free polyclonal anti-apoA-1 IgG or control isotype IgG (CTL IgG) every two weeks for 16 weeks. Atherosclerotic plaque size and vulnerability were assessed by histology. Myocardial ischaemia and necrosis, respectively, were determined by electrocardiographic (ECG) changes assessed by telemetry and serum troponin I (cTnI) measurements. Impact on survival was assessed by Kaplan-Meier analyses. In ApoE-/- mice, anti-apoA-1 IgG passive immunisation enhanced histological features of athero-sclerotic plaque vulnerability (increase in neutrophil and MMP-9 and reduction in collagen content), induced a substantial cTnI elevation (p=0.001), and increased mortality rate by 23 % (LogRank, p=0.04) when compared to CTL IgG. On a subgroup of ApoE-/- mice equipped with telemetry (n=4), a significant ST-segment depression was noted in anti-apoA-1 IgG-treated mice when compared to CTL IgG recipients (p< 0.001), and an acute ST-segment elevation myocardial infarction preceding mouse death was observed in one case. The deleterious effects of anti-apoA-1 IgG on atherosclerotic plaque vulnerability, myocardial necrosis and death were partially reversed in TLR2-/-ApoE-/- and TLR4-/-ApoE-/- backgrounds. In conclusion, anti-apoA-1 auto-antibodies seem to be active mediators of atherosclerotic plaque vulnerability, myocardial necrosis, and mortality in mice through TLR2- and TLR4-mediated pathways.
Collapse
|
28
|
Carbone F, Satta N, Burger F, Roth A, Lenglet S, Pagano S, Lescuyer P, Bertolotto M, Spinella G, Pane B, Palombo D, Pende A, Dallegri F, Mach F, Vuilleumier N, Montecucco F. Vitamin D receptor is expressed within human carotid plaques and correlates with pro-inflammatory M1 macrophages. Vascul Pharmacol 2016; 85:57-65. [PMID: 27555526 DOI: 10.1016/j.vph.2016.08.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/17/2016] [Accepted: 08/14/2016] [Indexed: 11/25/2022]
|
29
|
Bonaventura A, Mach F, Roth A, Lenglet S, Burger F, Brandt KJ, Pende A, Bertolotto M, Spinella G, Pane B, Palombo D, Dallegri F, Cea M, Vuilleumier N, Montecucco F, Carbone F. Intraplaque Expression of C-Reactive Protein Predicts Cardiovascular Events in Patients with Severe Atherosclerotic Carotid Artery Stenosis. Mediators Inflamm 2016; 2016:9153673. [PMID: 27738391 PMCID: PMC5050375 DOI: 10.1155/2016/9153673] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 08/22/2016] [Indexed: 01/15/2023] Open
Abstract
Serum c-reactive protein (CRP) was suggested for the assessment of intermediate cardiovascular (CV) risk. Here, systemic or intraplaque CRP levels were investigated as predictors of major adverse cardiovascular events (MACEs) in patients with severe carotid stenosis. CRP levels were assessed in the serum and within different portions (upstream and downstream) of carotid plaques of 217 patients undergoing endarterectomy. The association between CRP and intraplaque lipids, collagen, neutrophils, smooth muscle cells (SMC), and macrophage subsets was determined. No correlation between serum CRP and intraplaque biomarkers was observed. In upstream portions, CRP content was directly correlated with intraplaque neutrophils, total macrophages, and M1 macrophages and inversely correlated with SMC content. In downstream portions, intraplaque CRP correlated with M1 and M2 macrophages. According to the cut-off point (CRP > 2.9%) identified by ROC analysis in upstream portions, Kaplan-Meier analysis showed that patients with high CRP levels had a greater rate of MACEs. This risk of MACEs increased independently of age, male gender, serum CRP, and statin use. In conclusion, in patients with severe carotid artery stenosis, high CRP levels within upstream portions of carotid plaques directly and positively correlate with intraplaque inflammatory cells and predict MACEs at an 18-month follow-up period.
Collapse
Affiliation(s)
- Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
| | - François Mach
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, 64 Avenue de la Roseraie, 1211 Geneva, Switzerland
| | - Aline Roth
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, 64 Avenue de la Roseraie, 1211 Geneva, Switzerland
| | - Sébastien Lenglet
- Unit of Toxicology, University Centre of Legal Medicine, Geneva-Lausanne, Rue Michel-Servet 1, 1211 Geneva, Switzerland
| | - Fabienne Burger
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, 64 Avenue de la Roseraie, 1211 Geneva, Switzerland
| | - Karim J. Brandt
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, 64 Avenue de la Roseraie, 1211 Geneva, Switzerland
| | - Aldo Pende
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
- IRCCS AOU San Martino-IST, 10 Largo Benzi, 16132 Genoa, Italy
| | - Maria Bertolotto
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
| | - Giovanni Spinella
- Vascular and Endovascular Surgery Unit, Department of Surgery, IRCCS AOU San Martino-IST, 10 Largo Benzi, 16132 Genoa, Italy
| | - Bianca Pane
- Vascular and Endovascular Surgery Unit, Department of Surgery, IRCCS AOU San Martino-IST, 10 Largo Benzi, 16132 Genoa, Italy
| | - Domenico Palombo
- Vascular and Endovascular Surgery Unit, Department of Surgery, IRCCS AOU San Martino-IST, 10 Largo Benzi, 16132 Genoa, Italy
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
- IRCCS AOU San Martino-IST, 10 Largo Benzi, 16132 Genoa, Italy
| | - Michele Cea
- Clinic of Hematology, Department of Internal Medicine, University of Genoa, IRCCS AOU San Martino-IST, 10 Largo Benzi, 16132 Genoa, Italy
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, 4 Rue Gabrielle-Perret-Gentil, 1205 Geneva, Switzerland
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
- IRCCS AOU San Martino-IST, 10 Largo Benzi, 16132 Genoa, Italy
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 9 Viale Benedetto XV, 16132 Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
| |
Collapse
|
30
|
Carbone F, Satta N, Montecucco F, Virzi J, Burger F, Roth A, Roversi G, Tamborino C, Casetta I, Seraceni S, Trentini A, Padroni M, Dallegri F, Lalive PH, Mach F, Fainardi E, Vuilleumier N. Anti-ApoA-1 IgG serum levels predict worse poststroke outcomes. Eur J Clin Invest 2016; 46:805-17. [PMID: 27490973 DOI: 10.1111/eci.12664] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 08/01/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND Autoantibodies to apolipoprotein A-1 (anti-ApoA-1 IgG) were shown to predict major adverse cardiovascular events and promote atherogenesis. However, their potential relationship with clinical disability and ischaemic lesion volume after acute ischaemic stroke (AIS) remains unexplored. MATERIALS AND METHODS We included n = 76 patients admitted for AIS and we investigated whether baseline serum anti-ApoA-1 IgG levels could predict (i) AIS-induced clinical disability [assessed by the modified Rankin Scale (mRS)], and (ii) AIS-related ischaemic lesion volume [assessed by Computed Tomography (CT)]. We also evaluated the possible pro-apoptotic and pro-necrotic effects of anti-ApoA-1 IgG on human astrocytoma cell line (U251) using flow cytometry. RESULTS High levels of anti-ApoA-1 IgG were retrieved in 15·8% (12/76) of patients. Increased baseline levels of anti-ApoA-1 IgG were independently correlated with worse mRS [β = 0·364; P = 0·002; adjusted odds ratio (OR): 1·05 (95% CI 1·01-1·09); P = 0·017] and CT-assessed ischaemic lesion volume [β = 0·333; P < 0·001; adjusted OR: 1·06 (95% CI 1·01-1·12); P = 0·048] at 3 months. No difference in baseline clinical, biochemical and radiological characteristics was observed between patients with high vs. low levels of anti-ApoA-1 IgG. Incubating human astrocytoma cells with anti-ApoA-1 IgG dose dependently induced necrosis and apoptosis of U251 cells in vitro. CONCLUSION Anti-ApoA-1 IgG serum levels at AIS onset are associated with poorer clinical recovery and worse brain lesion volume 3 months after AIS. These observations could be partly explained by the deleterious effect of anti-ApoA-1 IgG on human brain cell survival in vitro and may have clinical implication in the prediction of poor outcome in AIS.
Collapse
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, School of Medicine, University of Genoa, Genoa, Italy
| | - Nathalie Satta
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, School of Medicine, University of Genoa, Genoa, Italy.,IRCCS AOU San Martino - IST, Genoa, Italy.,Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Julien Virzi
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Fabienne Burger
- Division of Cardiology, Department of Medical Specialties, Foundation for Medical Researches, University of Geneva, Geneva, Switzerland
| | - Aline Roth
- Division of Cardiology, Department of Medical Specialties, Foundation for Medical Researches, University of Geneva, Geneva, Switzerland
| | - Gloria Roversi
- Department of Biological, Psychiatric and Psychological Science, Azienda Ospedaliera-Universitaria, Arcispedale S. Anna, Ferrara, Italy
| | - Carmine Tamborino
- Department of Biological, Psychiatric and Psychological Science, Azienda Ospedaliera-Universitaria, Arcispedale S. Anna, Ferrara, Italy
| | - Ilaria Casetta
- Department of Biological, Psychiatric and Psychological Science, Azienda Ospedaliera-Universitaria, Arcispedale S. Anna, Ferrara, Italy
| | - Silva Seraceni
- Institute for Maternal and Child Health 'IRCCS Burlo Garofolo', Trieste, Italy
| | - Alessandro Trentini
- Section of Medical Biochemistry, Molecular Biology and Genetics, Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Marina Padroni
- Section of Medical Biochemistry, Molecular Biology and Genetics, Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, School of Medicine, University of Genoa, Genoa, Italy.,IRCCS AOU San Martino - IST, Genoa, Italy
| | - Patrice H Lalive
- Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospital, Geneva, Switzerland
| | - François Mach
- Division of Cardiology, Department of Medical Specialties, Foundation for Medical Researches, University of Geneva, Geneva, Switzerland
| | - Enrico Fainardi
- Neuroradiology Unit, Department of Neurosciences and Rehabilitation, Azienda Ospedaliera-Universitaria, Arcispedale S. Anna, Ferrara, Italy
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
31
|
Antiochos P, Marques-Vidal P, Virzi J, Pagano S, Satta N, Bastardot F, Hartley O, Montecucco F, Mach F, Waeber G, Vollenweider P, Vuilleumier N. Association between anti-apolipoprotein A-1 antibodies and cardiovascular disease in the general population. Results from the CoLaus study. Thromb Haemost 2016; 116:764-71. [PMID: 27384400 DOI: 10.1160/th16-03-0248] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 06/01/2016] [Indexed: 12/13/2022]
Abstract
We aimed to determine the association between autoantibodies against apolipoprotein A-1 (anti-apoA-1 IgG) and prevalent cardiovascular (CV) disease (CVD) as well as markers of CV risk in the general population. Cross-sectional data were obtained from 6649 subjects (age 52.6 ± 10.7 years, 47.4 % male) of the population-based CoLaus study. CVD was defined as myocardial infarction, angina pectoris, percutaneous revascularisation or bypass grafting for ischaemic heart disease stroke or transient ischaemic attack, and was assessed according to standardised medical records. Anti-apoA-1 IgG and biological markers were measured by ELISA and conventional automated techniques, respectively. Prevalence of high anti-apoA-1 IgG levels in the general population was 19.9 %. Presence of anti-apoA-1 IgG was significantly associated with CVD [odds ratio 1.34, 95 % confidence interval (1.05-1.70), p=0.018], independently of established CV risk factors (CVRFs) including age, sex, hypertension, smoking, diabetes, low and high-density lipoprotein cholesterol levels. The n=455 (6.8 %) study participants with a history of CVD (secondary prevention subgroup) presented higher median anti-ApoA-1 IgG values compared with subjects without CVD (p=0.029). Among patients in the secondary prevention subgroup, those with positive anti-apoA-1 IgG levels had lower HDL (p=0.002) and magnesium (p=0.001) levels, but increased uric acid and high-sensitivity C-reactive protein levels (p=0.022, and p<0.001, respectively) compared to patients with negative anti-apoA-1 IgG levels. In conclusion, anti-apoA-1 IgG levels are independently associated with CVD in the general population and also related to CV biomarkers in secondary prevention. These findings indicate that anti-apoA-1 IgG may represent a novel CVRF and need further study in prospective cohorts.
Collapse
Affiliation(s)
- Panagiotis Antiochos
- Dr. Panagiotis Antiochos, CoLaus Study, Bâtiment des Instituts, 19, Rue du Bugnon, CH-1005 Lausanne, Switzerland, Tel.: +41 79 556 03 11, Fax: +41 21 314 80 37, E-mail:
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Chistiakov DA, Orekhov AN, Bobryshev YV. ApoA1 and ApoA1-specific self-antibodies in cardiovascular disease. J Transl Med 2016; 96:708-18. [PMID: 27183204 DOI: 10.1038/labinvest.2016.56] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 03/21/2016] [Accepted: 04/03/2016] [Indexed: 12/15/2022] Open
Abstract
Apolipoprotein A1 (ApoA1) is a main protein moiety in high-density lipoprotein (HDL) particles. Generally, ApoA1 and HDL are considered as atheroprotective. In prooxidant and inflammatory microenvironment in the vicinity to the atherosclerotic lesion, ApoA1/HDL are subjected to modification. The chemical modifications such as oxidation, nitration, etc result in altering native architecture of ApoA1 toward dysfunctionality and abnormality. Neutrophil myeloperoxidase has a prominent role in this mechanism. Neo-epitopes could be formed and then exposed that makes them immunogenic. Indeed, these epitopes may be recognized by immune cells and induce production of proatherogenic ApoA1-specific IgG antibodies. These antibodies are biologically relevant because they are able to react with Toll-like receptor (TLR)-2 and TLR4 in target cells and induce a variety of pro-inflammatory responses. Epidemiological and functional studies underline a prognostic value of ApoA1 self-antibodies for several cardiovascular diseases, including myocardial infarction, acute coronary syndrome, and severe carotid stenosis.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Department of Molecular Genetic Diagnostics and Cell Biology, Division of Laboratory Medicine, Institute of Pediatrics, Research Center for Children's Health, Moscow, Russia
| | - Alexander N Orekhov
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow, Russia.,Faculty of Biology, Department of Biophysics, Lomonosov Moscow State University, Moscow, Russia
| | - Yuri V Bobryshev
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow, Russia.,Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.,School of Medicine, University of Western Sydney, Campbelltown, NSW, Australia
| |
Collapse
|
33
|
Annema W, von Eckardstein A. Dysfunctional high-density lipoproteins in coronary heart disease: implications for diagnostics and therapy. Transl Res 2016; 173:30-57. [PMID: 26972566 DOI: 10.1016/j.trsl.2016.02.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 02/15/2016] [Accepted: 02/17/2016] [Indexed: 12/18/2022]
Abstract
Low plasma levels of high-density lipoprotein (HDL) cholesterol are associated with increased risks of coronary heart disease. HDL mediates cholesterol efflux from macrophages for reverse transport to the liver and elicits many anti-inflammatory and anti-oxidative activities which are potentially anti-atherogenic. Nevertheless, HDL has not been successfully targeted by drugs for prevention or treatment of cardiovascular diseases. One potential reason is the targeting of HDL cholesterol which does not capture the structural and functional complexity of HDL particles. Hundreds of lipid species and dozens of proteins as well as several microRNAs have been identified in HDL. This physiological heterogeneity is further increased in pathologic conditions due to additional quantitative and qualitative molecular changes of HDL components which have been associated with both loss of physiological function and gain of pathologic dysfunction. This structural and functional complexity of HDL has prevented clear assignments of molecules to the functions of normal HDL and dysfunctions of pathologic HDL. Systematic analyses of structure-function relationships of HDL-associated molecules and their modifications are needed to test the different components and functions of HDL for their relative contribution in the pathogenesis of atherosclerosis. The derived biomarkers and targets may eventually help to exploit HDL for treatment and diagnostics of cardiovascular diseases.
Collapse
Affiliation(s)
- Wijtske Annema
- Institute of Clinical Chemistry, University Hospital Zurich, Zurich, Switzerland
| | | |
Collapse
|
34
|
Pagano S, Carbone F, Burger F, Roth A, Bertolotto M, Pane B, Spinella G, Palombo D, Pende A, Dallegri F, Satta N, Virzi J, Fontana P, Mach F, Montecucco F, Vuilleumier N. Anti-apolipoprotein A-1 auto-antibodies as active modulators of atherothrombosis. Thromb Haemost 2016; 116:554-64. [PMID: 27356567 DOI: 10.1160/th16-03-0229] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 05/25/2016] [Indexed: 12/18/2022]
Abstract
Humoral autoimmune-mediated inflammation plays a role in atherogenesis, and potentially in arterial thrombosis. Anti-apolipoprotein A-1 (apoA-1) IgG have been reported to represent emergent mediators of atherogenesis through Toll-like receptors (TLR) 2, 4 and CD14 signalling. We investigated the role of anti-apoA-1 IgG on tissue factor (TF) expression and activation, a key coagulation regulator underlying atherothrombosis. Atherothrombosis features were determined by immunohistochemical TF staining of human carotid biopsies derived from patients with severe carotid stenosis undergoing elective surgery (n=176), and on aortic roots of different genetic backgrounds mice (ApoE-/-; TLR2-/-ApoE-/- and TLR4-/-ApoE-/-) exposed to passive immunisation with anti-apoA-1 IgG. Human serum levels of anti-apoA-1 IgG were measured by ELISA. In vitro, on human-monocyte-derived-macrophages (HMDM) the anti-apoA-1 IgG increased TF expression and activity were analysed by FACS and chromogenic assays in presence of different pharmacological inhibitors. Human serum anti-apoA-1 IgG levels significantly correlated to intraplaque TF expression in carotid biopsies (r=0.31, p<0.001), which was predictive of clinically symptomatic lesions. On HMDM, anti-apoA-1 IgG induced a TLR2, 4 and CD14-dependent increase in TF expression and activity, involving NF-kappaB and a c-Jun N-terminal kinase-dependent AP-1 transcription factors. In ApoE-/- mice, anti-apoA-1 IgG passive immunisation significantly enhanced intraplaque TF expression when compared to control IgG. This effect was lost in both TLR2-/-ApoE-/- and TLR4-/-ApoE-/- mice. These results demonstrate that anti-apoA-1 IgG are associated with TF expression in human atherosclerotic plaques, induce TF expression in vitro and in vivo through TLR2 and 4 signalling, supporting a possible causal relationship between anti-apoA-1 IgG and atherothrombosis.
Collapse
Affiliation(s)
- Sabrina Pagano
- Sabrina Pagano, PhD, Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, 4 rue Gabrielle Perret-Gentil, 1205 Geneva, Switzerland, Tel.: +41 22 37 95 321, Fax: +41 22 3795502, E-mail:
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
El-Lebedy D, Rasheed E, Kafoury M, Abd-El Haleem D, Awadallah E, Ashmawy I. Anti-apolipoprotein A-1 autoantibodies as risk biomarker for cardiovascular diseases in type 2 diabetes mellitus. J Diabetes Complications 2016; 30:580-5. [PMID: 26965796 DOI: 10.1016/j.jdiacomp.2016.02.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 02/04/2016] [Accepted: 02/16/2016] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Anti-Apolipoprotein A-1 autoantibodies (anti-ApoA-1 IgG) represent an emerging prognostic cardiovascular marker in patients with myocardial infarction or autoimmune diseases associated with high thrombotic events. The aim of this work is to investigate the incidence of anti-apoA-1 autoantibodies in type 2 diabetes (T2DM) patients with and without CVD and to study potential association with disease risk and its effect on plasma lipid parameters. METHODS Qualitative determination of anti-apoA-1 IgG was assayed in sera from 302 subjects classified into T2DM patients (n=102), T2DM+CVD (n=112) and healthy controls (n=88). RESULTS The incidence of anti-apoA-1 IgG was significantly higher among CVD patients (35.7%) than T2DM patients (8.8%) or control subjects (6.1%), p<0.0001. A significant association with CVD was identified (p<0.0001) and subjects who were positive for anti-apoA-1 IgG were at 8.5 times increased risk to develop CVD when compared to controls. Diabetic patients who were positive for the antibodies showed 5.7 times increased CVD risk. ROC analysis indicated anti-apoA-1 IgG as a risk biomarker for CVD in T2DM patients with an AUC value of 0.76, sensitivity of 35.7% and specificity of 91.2%. Studying the effect on lipid parameters, anti-apoA-1 IgG associated with significantly higher serum concentrations of TC and non-HDL-C in all groups and with higher concentrations of LDL-C in diabetic patients and higher TC/HDL-C ratio in CVD patients. CONCLUSION Our results indicate that anti-apoA-1 IgG is a cardiovascular risk biomarker in T2DM patients.
Collapse
Affiliation(s)
- Dalia El-Lebedy
- Department of Clinical and Chemical Pathology, Medical Research Division, National Research Centre, Cairo, Egypt
| | - Enas Rasheed
- Department of Clinical and Chemical Pathology, Medical Research Division, National Research Centre, Cairo, Egypt
| | - Mona Kafoury
- Department of Clinical and Chemical Pathology, Medical Research Division, National Research Centre, Cairo, Egypt
| | - Dalia Abd-El Haleem
- Department of Clinical and Chemical Pathology, Medical Research Division, National Research Centre, Cairo, Egypt
| | - Eman Awadallah
- Department of Clinical and Chemical Pathology, Medical Research Division, National Research Centre, Cairo, Egypt
| | - Ingy Ashmawy
- Department of Clinical and Chemical Pathology, Medical Research Division, National Research Centre, Cairo, Egypt
| |
Collapse
|
36
|
Pende A, Artom N, Bertolotto M, Montecucco F, Dallegri F. Role of neutrophils in atherogenesis: an update. Eur J Clin Invest 2016; 46:252-63. [PMID: 26573245 DOI: 10.1111/eci.12566] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/07/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND The role of neutrophils in the beginning and the progression of the atherosclerotic process did not receive much attention until the last years. On the contrary, recent data, in both the experimental animals and humans, suggest important effects of these cells with possible clinical consequences. MATERIALS AND METHODS This narrative review was based on the papers found on PubMed and MEDLINE up to July 2015. The search terms used were 'neutrophil, atherosclerosis' in combination with 'recruitment, chemokine, plaque destabilization and pathophysiology'. RESULTS Different models demonstrate the presence and the actions of neutrophils in the early steps of the atherogenesis confirming the fundamental role of these cells in the response of the innate immune system to different pathogens (in this context the modified lipoproteins). However, also the late phases of the atherosclerotic process, in particular the destabilization of a mature plaque, seem to be modulated by the neutrophils, possibly through the interaction with recently discovered biological systems such as the endocannabinoids. CONCLUSIONS The understanding of the mechanisms involved in the modulation exerted by neutrophils in atherosclerosis is pivotal in terms of the complete definition of the overall picture. This approach will certainly give us new targets and new pharmacological opportunities for the anti-inflammatory strategy of the cardiovascular prevention.
Collapse
Affiliation(s)
- Aldo Pende
- Clinic of Internal Medicine 1, Department of Internal Medicine, University of Genoa School of Medicine, IRCCS Azienda Ospedaliera Universitaria San Martino - IST, Genoa, Italy
| | - Nathan Artom
- Clinic of Internal Medicine 1, Department of Internal Medicine, University of Genoa School of Medicine, IRCCS Azienda Ospedaliera Universitaria San Martino - IST, Genoa, Italy
| | - Maria Bertolotto
- Clinic of Internal Medicine 1, Department of Internal Medicine, University of Genoa School of Medicine, IRCCS Azienda Ospedaliera Universitaria San Martino - IST, Genoa, Italy
| | - Fabrizio Montecucco
- Clinic of Internal Medicine 1, Department of Internal Medicine, University of Genoa School of Medicine, IRCCS Azienda Ospedaliera Universitaria San Martino - IST, Genoa, Italy.,Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland.,Division of Cardiology, Faculty of Medicine, Foundation for Medical Researches, University of Geneva, Geneva, Switzerland
| | - Franco Dallegri
- Clinic of Internal Medicine 1, Department of Internal Medicine, University of Genoa School of Medicine, IRCCS Azienda Ospedaliera Universitaria San Martino - IST, Genoa, Italy
| |
Collapse
|
37
|
Mannic T, Satta N, Pagano S, Python M, Virzi J, Montecucco F, Frias MA, James RW, Maturana AD, Rossier MF, Vuilleumier N. CD14 as a Mediator of the Mineralocorticoid Receptor-Dependent Anti-apolipoprotein A-1 IgG Chronotropic Effect on Cardiomyocytes. Endocrinology 2015; 156:4707-19. [PMID: 26393305 DOI: 10.1210/en.2015-1605] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In vitro and animal studies point to autoantibodies against apolipoprotein A-1 (anti-apoA-1 IgG) as possible mediators of cardiovascular (CV) disease involving several mechanisms such as basal heart rate interference mediated by a mineralocorticoid receptor-dependent L-type calcium channel activation, and a direct pro-inflammatory effect through the engagement of the toll-like receptor (TLR) 2/CD14 complex. Nevertheless, the possible implication of these receptors in the pro-arrhythmogenic effect of anti-apoA-1 antibodies remains elusive. We aimed at determining whether CD14 and TLRs could mediate the anti-apoA-1 IgG chronotropic response in neonatal rat ventricular cardiomyocytes (NRVC). Blocking CD14 suppressed anti-apoA-1 IgG binding to NRVC and the related positive chronotropic response. Anti-apoA-1 IgG alone induced the formation of a TLR2/TLR4/CD14 complex, followed by the phosphorylation of Src, whereas aldosterone alone promoted the phosphorylation of Akt by phosphatidylinositol 3-kinase (PI3K), without affecting the chronotropic response. In the presence of both aldosterone and anti-apoA-1 IgG, the localization of TLR2/TLR4/CD14 was increased in membrane lipid rafts, followed by PI3K and Src activation, leading to an L-type calcium channel-dependent positive chronotropic response. Pharmacological inhibition of the Src pathway led to the decrease of L-type calcium channel activity and abrogated the NRVC chronotropic response. Activation of CD14 seems to be a key regulator of the mineralocorticoid receptor-dependent anti-apoA-1 IgG positive chronotropic effect on NRVCs, involving relocation of the CD14/TLR2/TLR4 complex into lipid rafts followed by PI3K and Src-dependent L-type calcium channel activation.
Collapse
Affiliation(s)
- Tiphaine Mannic
- Human Protein Sciences Department, Chemistry and Proteomic Group, Auto-immunity and Atherogenesis group; and Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine (T.M., N.S., J.V., F.M., N.V., M.F.R.), Geneva University Hospitals, 1201 Geneva, Switzerland; Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition (M.P., M.A.F., R.W.J.), Geneva University Hospitals, Switzerland; Department of Bioengineering Sciences (A.D.M.), Graduate School of Bioagricultural Sciences, Furo-cho, Chikusa-ku, Nagoya 464-8601, Nagoya University, Japan; and Central Institute of the Hospital of Valais (M.F.R.), 1951 Sion, Switzerland
| | - Nathalie Satta
- Human Protein Sciences Department, Chemistry and Proteomic Group, Auto-immunity and Atherogenesis group; and Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine (T.M., N.S., J.V., F.M., N.V., M.F.R.), Geneva University Hospitals, 1201 Geneva, Switzerland; Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition (M.P., M.A.F., R.W.J.), Geneva University Hospitals, Switzerland; Department of Bioengineering Sciences (A.D.M.), Graduate School of Bioagricultural Sciences, Furo-cho, Chikusa-ku, Nagoya 464-8601, Nagoya University, Japan; and Central Institute of the Hospital of Valais (M.F.R.), 1951 Sion, Switzerland
| | - Sabrina Pagano
- Human Protein Sciences Department, Chemistry and Proteomic Group, Auto-immunity and Atherogenesis group; and Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine (T.M., N.S., J.V., F.M., N.V., M.F.R.), Geneva University Hospitals, 1201 Geneva, Switzerland; Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition (M.P., M.A.F., R.W.J.), Geneva University Hospitals, Switzerland; Department of Bioengineering Sciences (A.D.M.), Graduate School of Bioagricultural Sciences, Furo-cho, Chikusa-ku, Nagoya 464-8601, Nagoya University, Japan; and Central Institute of the Hospital of Valais (M.F.R.), 1951 Sion, Switzerland
| | - Magaly Python
- Human Protein Sciences Department, Chemistry and Proteomic Group, Auto-immunity and Atherogenesis group; and Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine (T.M., N.S., J.V., F.M., N.V., M.F.R.), Geneva University Hospitals, 1201 Geneva, Switzerland; Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition (M.P., M.A.F., R.W.J.), Geneva University Hospitals, Switzerland; Department of Bioengineering Sciences (A.D.M.), Graduate School of Bioagricultural Sciences, Furo-cho, Chikusa-ku, Nagoya 464-8601, Nagoya University, Japan; and Central Institute of the Hospital of Valais (M.F.R.), 1951 Sion, Switzerland
| | - Julien Virzi
- Human Protein Sciences Department, Chemistry and Proteomic Group, Auto-immunity and Atherogenesis group; and Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine (T.M., N.S., J.V., F.M., N.V., M.F.R.), Geneva University Hospitals, 1201 Geneva, Switzerland; Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition (M.P., M.A.F., R.W.J.), Geneva University Hospitals, Switzerland; Department of Bioengineering Sciences (A.D.M.), Graduate School of Bioagricultural Sciences, Furo-cho, Chikusa-ku, Nagoya 464-8601, Nagoya University, Japan; and Central Institute of the Hospital of Valais (M.F.R.), 1951 Sion, Switzerland
| | - Fabrizio Montecucco
- Human Protein Sciences Department, Chemistry and Proteomic Group, Auto-immunity and Atherogenesis group; and Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine (T.M., N.S., J.V., F.M., N.V., M.F.R.), Geneva University Hospitals, 1201 Geneva, Switzerland; Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition (M.P., M.A.F., R.W.J.), Geneva University Hospitals, Switzerland; Department of Bioengineering Sciences (A.D.M.), Graduate School of Bioagricultural Sciences, Furo-cho, Chikusa-ku, Nagoya 464-8601, Nagoya University, Japan; and Central Institute of the Hospital of Valais (M.F.R.), 1951 Sion, Switzerland
| | - Miguel A Frias
- Human Protein Sciences Department, Chemistry and Proteomic Group, Auto-immunity and Atherogenesis group; and Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine (T.M., N.S., J.V., F.M., N.V., M.F.R.), Geneva University Hospitals, 1201 Geneva, Switzerland; Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition (M.P., M.A.F., R.W.J.), Geneva University Hospitals, Switzerland; Department of Bioengineering Sciences (A.D.M.), Graduate School of Bioagricultural Sciences, Furo-cho, Chikusa-ku, Nagoya 464-8601, Nagoya University, Japan; and Central Institute of the Hospital of Valais (M.F.R.), 1951 Sion, Switzerland
| | - Richard W James
- Human Protein Sciences Department, Chemistry and Proteomic Group, Auto-immunity and Atherogenesis group; and Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine (T.M., N.S., J.V., F.M., N.V., M.F.R.), Geneva University Hospitals, 1201 Geneva, Switzerland; Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition (M.P., M.A.F., R.W.J.), Geneva University Hospitals, Switzerland; Department of Bioengineering Sciences (A.D.M.), Graduate School of Bioagricultural Sciences, Furo-cho, Chikusa-ku, Nagoya 464-8601, Nagoya University, Japan; and Central Institute of the Hospital of Valais (M.F.R.), 1951 Sion, Switzerland
| | - Andres D Maturana
- Human Protein Sciences Department, Chemistry and Proteomic Group, Auto-immunity and Atherogenesis group; and Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine (T.M., N.S., J.V., F.M., N.V., M.F.R.), Geneva University Hospitals, 1201 Geneva, Switzerland; Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition (M.P., M.A.F., R.W.J.), Geneva University Hospitals, Switzerland; Department of Bioengineering Sciences (A.D.M.), Graduate School of Bioagricultural Sciences, Furo-cho, Chikusa-ku, Nagoya 464-8601, Nagoya University, Japan; and Central Institute of the Hospital of Valais (M.F.R.), 1951 Sion, Switzerland
| | - Michel F Rossier
- Human Protein Sciences Department, Chemistry and Proteomic Group, Auto-immunity and Atherogenesis group; and Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine (T.M., N.S., J.V., F.M., N.V., M.F.R.), Geneva University Hospitals, 1201 Geneva, Switzerland; Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition (M.P., M.A.F., R.W.J.), Geneva University Hospitals, Switzerland; Department of Bioengineering Sciences (A.D.M.), Graduate School of Bioagricultural Sciences, Furo-cho, Chikusa-ku, Nagoya 464-8601, Nagoya University, Japan; and Central Institute of the Hospital of Valais (M.F.R.), 1951 Sion, Switzerland
| | - Nicolas Vuilleumier
- Human Protein Sciences Department, Chemistry and Proteomic Group, Auto-immunity and Atherogenesis group; and Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine (T.M., N.S., J.V., F.M., N.V., M.F.R.), Geneva University Hospitals, 1201 Geneva, Switzerland; Department of Internal Medicine, Division of Endocrinology, Diabetology, Hypertension and Nutrition (M.P., M.A.F., R.W.J.), Geneva University Hospitals, Switzerland; Department of Bioengineering Sciences (A.D.M.), Graduate School of Bioagricultural Sciences, Furo-cho, Chikusa-ku, Nagoya 464-8601, Nagoya University, Japan; and Central Institute of the Hospital of Valais (M.F.R.), 1951 Sion, Switzerland
| |
Collapse
|
38
|
Pagano S, Gaertner H, Cerini F, Mannic T, Satta N, Teixeira PC, Cutler P, Mach F, Vuilleumier N, Hartley O. The Human Autoantibody Response to Apolipoprotein A-I Is Focused on the C-Terminal Helix: A New Rationale for Diagnosis and Treatment of Cardiovascular Disease? PLoS One 2015; 10:e0132780. [PMID: 26177543 PMCID: PMC4503694 DOI: 10.1371/journal.pone.0132780] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 06/19/2015] [Indexed: 12/16/2022] Open
Abstract
Background Cardiovascular disease (CVD) is the leading cause of death worldwide and new approaches for both diagnosis and treatment are required. Autoantibodies directed against apolipoprotein A-I (ApoA-I) represent promising biomarkers for use in risk stratification of CVD and may also play a direct role in pathogenesis. Methodology To characterize the anti-ApoA-I autoantibody response, we measured the immunoreactivity to engineered peptides corresponding to the different alpha-helical regions of ApoA-I, using plasma from acute chest pain cohort patients known to be positive for anti-ApoA-I autoantibodies. Principal Findings Our results indicate that the anti-ApoA-I autoantibody response is strongly biased towards the C-terminal alpha-helix of the protein, with an optimized mimetic peptide corresponding to this part of the protein recapitulating the diagnostic accuracy for an acute ischemic coronary etiology (non-ST segment elevation myocardial infarction and unstable angina) obtainable using intact endogenous ApoA-I in immunoassay. Furthermore, the optimized mimetic peptide strongly inhibits the pathology-associated capacity of anti-ApoA-I antibodies to elicit proinflammatory cytokine release from cultured human macrophages. Conclusions In addition to providing a rationale for the development of new approaches for the diagnosis and therapy of CVD, our observations may contribute to the elucidation of how anti-ApoA-I autoantibodies are elicited in individuals without autoimmune disease.
Collapse
Affiliation(s)
- Sabrina Pagano
- Department of Human Protein Sciences, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Hubert Gaertner
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Fabrice Cerini
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Tiphaine Mannic
- Department of Human Protein Sciences, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Nathalie Satta
- Department of Human Protein Sciences, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Priscila Camillo Teixeira
- Pharmaceutical Sciences, Pharma Research and Early Development, F.Hoffmann-La Roche, Basel, Switzerland
| | - Paul Cutler
- Pharmaceutical Sciences, Pharma Research and Early Development, F.Hoffmann-La Roche, Basel, Switzerland
| | - François Mach
- Division of Cardiology, Foundation for Medical Researches, University of Geneva, Geneva, Switzerland
| | - Nicolas Vuilleumier
- Department of Human Protein Sciences, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
- * E-mail: (OH); (NV)
| | - Oliver Hartley
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- * E-mail: (OH); (NV)
| |
Collapse
|
39
|
Rubini Gimenez M, Pagano S, Virzi J, Montecucco F, Twerenbold R, Reichlin T, Wildi K, Grueter D, Jaeger C, Haaf P, Vuilleumier N, Mueller C. Diagnostic and prognostic value of autoantibodies anti-apolipoprotein A-1 and anti-phosphorylcholine in acute non-ST elevation myocardial infarction. Eur J Clin Invest 2015; 45:369-79. [PMID: 25627775 DOI: 10.1111/eci.12411] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 01/22/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND Autoantibodies have been shown to play a critical role in predicting major adverse cardiovascular events in atherosclerotic patients. We aimed to assess the diagnostic accuracy of autoantibodies to apolipoprotein A-1 (anti-apoA-1 IgG) and to phosphorylcholine (anti-PC IgM) for non-ST segment elevation acute myocardial infarction (NSTEMI) and to explore their potential prognostic value. METHODS This prospective multicentre study included 1072 patients presenting to the emergency department for suspected NSTEMI. The final diagnosis was adjudicated by two independent cardiologists. For both antibodies alone or expressed as a ratio (anti-apoA-1 IgG/anti-PC IgM), we determined their (i) diagnostic accuracy for NSTEMI and (ii) prognostic accuracy for major adverse cardiovascular events (MACE) during 1-year follow-up. RESULTS A total of 154 patients (14%) had a final diagnosis of NSTEMI. Diagnostic accuracy for the diagnosis of NSTEMI as quantified by the area under the receiver operating characteristics curve (AUC) was very low for both autoantibodies separately as well as combined as a ratio: AUC anti-apoA-1 IgG 0.50 (95%CI, 0.47-0.53, P = 0.99), AUC anti-PC IgM 0.53 (95%CI, 0.50-0.56, P = 0.30) and AUC of the ratio 0.52 (95%CI, 0.49-0.55, P = 0.47). Adding the anti-apoA-1 IgG/Anti-PC IgM ratio to hs-cTnT did not provide incremental diagnostic value over hs-cTnT alone. MACE occurred in 221 patients (21%) during follow-up. The autoantibodies, separately or expressed as ratio, also had very low accuracy to predict MACE (p=ns). CONCLUSIONS Anti-apoA-1 IgG and anti-PC IgM autoantibodies did not have diagnostic or prognostic value in patients with NSTEMI.
Collapse
|
40
|
Carbone F, Montecucco F. Inflammation in arterial diseases. IUBMB Life 2015; 67:18-28. [PMID: 25631520 DOI: 10.1002/iub.1344] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 12/28/2014] [Indexed: 12/26/2022]
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine; Department of Internal Medicine; University of Genoa School of Medicine, IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro; Genoa Italy
- Division of Cardiology; Foundation for Medical Researches; Department of Medical Specialties; University of Geneva; Geneva Switzerland
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine; Department of Internal Medicine; University of Genoa School of Medicine, IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro; Genoa Italy
- Division of Cardiology; Foundation for Medical Researches; Department of Medical Specialties; University of Geneva; Geneva Switzerland
- Division of Laboratory Medicine; Department of Genetics and Laboratory Medicine; Geneva University Hospitals; Geneva Switzerland
| |
Collapse
|
41
|
Teixeira PC, Ducret A, Ferber P, Gaertner H, Hartley O, Pagano S, Butterfield M, Langen H, Vuilleumier N, Cutler P. Definition of human apolipoprotein A-I epitopes recognized by autoantibodies present in patients with cardiovascular diseases. J Biol Chem 2014; 289:28249-59. [PMID: 25170076 PMCID: PMC4192480 DOI: 10.1074/jbc.m114.589002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Autoantibodies to apolipoprotein A-I (anti-apoA-I IgG) have been shown to be both markers and mediators of cardiovascular disease, promoting atherogenesis and unstable atherosclerotic plaque. Previous studies have shown that high levels of anti-apoA-I IgGs are independently associated with major adverse cardiovascular events in patients with myocardial infarction. Autoantibody responses to apoA-I can be polyclonal and it is likely that more than one epitope may exist. To identify the specific immunoreactive peptides in apoA-I, we have developed a set of methodologies and procedures to isolate, purify, and identify novel apoA-I endogenous epitopes. First, we generated high purity apoA-I from human plasma, using thiophilic interaction chromatography followed by enzymatic digestion specifically at lysine or arginine residues. Immunoreactivity to the different peptides generated was tested by ELISA using serum obtained from patients with acute myocardial infarction and high titers of autoantibodies to native apoA-I. The immunoreactive peptides were further sequenced by mass spectrometry. Our approach successfully identified two novel immunoreactive peptides, recognized by autoantibodies from patients suffering from myocardial infarction, who contain a high titer of anti-apoA-I IgG. The discovery of these epitopes may open innovative prognostic and therapeutic opportunities potentially suitable to improve current cardiovascular risk stratification.
Collapse
Affiliation(s)
- Priscila Camillo Teixeira
- From the Pharma Research and Early Development, Roche Innovation Center, 4070 Basel, the Department of Genetics and Laboratory Medicine, Division of Laboratory Medicine, 1205 Geneva University Hospitals, 1205 Geneva, and
| | - Axel Ducret
- From the Pharma Research and Early Development, Roche Innovation Center, 4070 Basel
| | - Philippe Ferber
- From the Pharma Research and Early Development, Roche Innovation Center, 4070 Basel
| | - Hubert Gaertner
- the Department of Immunopathology, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
| | - Oliver Hartley
- the Department of Immunopathology, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
| | - Sabrina Pagano
- the Department of Genetics and Laboratory Medicine, Division of Laboratory Medicine, 1205 Geneva University Hospitals, 1205 Geneva, and
| | - Michelle Butterfield
- From the Pharma Research and Early Development, Roche Innovation Center, 4070 Basel
| | - Hanno Langen
- From the Pharma Research and Early Development, Roche Innovation Center, 4070 Basel
| | - Nicolas Vuilleumier
- the Department of Genetics and Laboratory Medicine, Division of Laboratory Medicine, 1205 Geneva University Hospitals, 1205 Geneva, and
| | - Paul Cutler
- From the Pharma Research and Early Development, Roche Innovation Center, 4070 Basel
| |
Collapse
|
42
|
Döring Y, Drechsler M, Soehnlein O, Weber C. Neutrophils in atherosclerosis: from mice to man. Arterioscler Thromb Vasc Biol 2014; 35:288-95. [PMID: 25147339 DOI: 10.1161/atvbaha.114.303564] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Infiltration of leukocyte subsets is a driving force of atherosclerotic lesion growth, and during the past decade, neutrophils have received growing attention in chronic inflammatory processes, such as atherosclerosis. Equipped with various ready to be released mediators, evolved to fight invading pathogens, neutrophils may also hold key functions in affecting sterile inflammation, such as in atherosclerosis. Many of their secretion products might instruct or activate other immune cells (particularly monocytes) to, for example, enter atherosclerotic lesions or release proinflammatory mediators. Despite the emerging evidence for the mechanistic contribution of neutrophils to early atherosclerosis in mice, their role in human atherogenesis, atheroprogression, and atherosclerotic plaque destabilization is still poorly understood. This brief review will summarize latest findings on the role of neutrophils in atherosclerosis and will pay special attention to studies describing a translation approach by combining measurements in mouse and human.
Collapse
Affiliation(s)
- Yvonne Döring
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (Y.D., M.D., O.S., C.W.); Department of Pathology, Academic Medical Center, Amsterdam University, Amsterdam, The Netherlands (M.D., O.S.); and German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (O.S., C.W.)
| | - Maik Drechsler
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (Y.D., M.D., O.S., C.W.); Department of Pathology, Academic Medical Center, Amsterdam University, Amsterdam, The Netherlands (M.D., O.S.); and German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (O.S., C.W.)
| | - Oliver Soehnlein
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (Y.D., M.D., O.S., C.W.); Department of Pathology, Academic Medical Center, Amsterdam University, Amsterdam, The Netherlands (M.D., O.S.); and German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (O.S., C.W.)
| | - Christian Weber
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (Y.D., M.D., O.S., C.W.); Department of Pathology, Academic Medical Center, Amsterdam University, Amsterdam, The Netherlands (M.D., O.S.); and German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (O.S., C.W.).
| |
Collapse
|
43
|
Vuilleumier N, Montecucco F, Hartley O. Autoantibodies to apolipoprotein A-1 as a biomarker of cardiovascular autoimmunity. World J Cardiol 2014; 6:314-326. [PMID: 24944761 PMCID: PMC4062126 DOI: 10.4330/wjc.v6.i5.314] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 03/18/2014] [Indexed: 02/06/2023] Open
Abstract
Immune-driven inflammation plays an important part in atherogenesis and is therefore believed to be key to the development of cardiovascular disease (CVD), which is currently the leading cause of death in the Western world. By fulfilling some of the Koch postulates, atherogenesis has even been proposed to be considered as an autoimmune disease, raising the hope that CVD could be prevented by immunomodulation. Nevertheless, the role of the immune system and autoimmune reactions in atherosclerosis appear to be a double edged-sword, with both pro-atherogenic and anti-atherogenic attributes. Hence, if immunomodulation is to become a therapeutic option for atherosclerosis and CVD, it will be crucial to correctly identify patients who might benefit from targeted suppression of deleterious autoimmune responses. This could be achieved, for example, by the detection of disease-associated autoantibodies. In this work, we will review the currently available clinical, in vitro, and animal studies dedicated to autoantibodies against apolipoprotein A-1 (anti-apoA-1 IgG), the major proteic fraction of high density lipoprotein. Current clinical studies indicate that high levels of anti-apoA-1 IgG are associated with a worse cardiovascular prognosis. In addition, in vitro and animal studies indicate a pro-inflammatory and pro-atherogenic role, supporting the hypothesis that these autoantibodies may play a direct causal role in CVD, and furthermore that they could potentially represent a therapeutic target for CVD in the future.
Collapse
|