1
|
Gerdes N, Klingenberg R. Mini-Review: Immunogenic epitopes in apolipoprotein B-100 for atheroprotective immunization. Front Cardiovasc Med 2024; 11:1448664. [PMID: 39211769 PMCID: PMC11357920 DOI: 10.3389/fcvm.2024.1448664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Here, we provide a concise overview of recent developments in the identification of immunogenic epitopes in human apolipoprotein B-100 for immunization against atherosclerotic cardiovascular disease. Major steps forward toward a clinical application of vaccines include the design of humanized mouse models, tetramer-based identification of antigen-specific T cells, and novel analysis tools, such as single-cell RNA sequencing and cytometry by time of flight, to assess temporal and spatial changes in immune cells in atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Norbert Gerdes
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty and University Hospital, Heinrich Heine University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Roland Klingenberg
- Cardiology, Kerckhoff-Klinik, Bad Nauheim, Campus of the Justus-Liebig-University Giessen, Giessen, Germany
- Department of Cardiology, German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
| |
Collapse
|
2
|
Zhao Y, Xu Q, He N, Jiang M, Chen Y, Ren Z, Tang Z, Wu C, Liu L. Non-oxidative Modified Low-density Lipoproteins: The Underappreciated Risk Factors for Atherosclerosis. Curr Med Chem 2024; 31:5598-5611. [PMID: 37550912 DOI: 10.2174/0929867331666230807154019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/29/2023] [Accepted: 06/01/2023] [Indexed: 08/09/2023]
Abstract
Atherosclerosis, the pathological basis of most cardiovascular diseases, is a main risk factor causing about 20 million deaths each year worldwide. Oxidized low-density lipoprotein is recognized as the most important and independent risk factor in initiating and promoting atherosclerosis. Numerous antioxidants are extensively used in clinical practice, but they have no significant effect on reducing the morbidity and mortality of cardiovascular diseases. This finding suggests that researchers should pay more attention to the important role of non-oxidative modified low-density lipoprotein in atherosclerosis with a focus on oxidized low-density lipoprotein. This review briefly summarizes several important non-oxidative modified low-density lipoproteins associated with atherosclerosis, introduces the pathways through which these non-oxidative modified low-density lipoproteins induce the development of atherosclerosis in vivo, and discusses the mechanism of atherogenesis induced by these non-oxidative modified low-density lipoproteins. New therapeutic strategies and potential drug targets are provided for the prevention and treatment of atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Yimeng Zhao
- Key Laboratory for Arteriosclerology of Hunan Province, Department of Cardiovascular Medicine, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, The Third Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Qian Xu
- Key Laboratory for Arteriosclerology of Hunan Province, Department of Cardiovascular Medicine, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, The Third Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Naiqi He
- Key Laboratory for Arteriosclerology of Hunan Province, Department of Cardiovascular Medicine, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, The Third Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Mulin Jiang
- Key Laboratory for Arteriosclerology of Hunan Province, Department of Cardiovascular Medicine, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, The Third Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Yingzhuo Chen
- Key Laboratory for Arteriosclerology of Hunan Province, Department of Cardiovascular Medicine, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, The Third Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Zhong Ren
- Key Laboratory for Arteriosclerology of Hunan Province, Department of Cardiovascular Medicine, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, The Third Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Zhihan Tang
- Key Laboratory for Arteriosclerology of Hunan Province, Department of Cardiovascular Medicine, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, The Third Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Chunyan Wu
- Key Laboratory for Arteriosclerology of Hunan Province, Department of Cardiovascular Medicine, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, The Third Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Lushan Liu
- Key Laboratory for Arteriosclerology of Hunan Province, Department of Cardiovascular Medicine, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, The Third Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang 421001, China
| |
Collapse
|
3
|
Ketelhuth DFJ. ApoB100-reactive T cells: Does liver tolerance hold the key to modulating adaptive immunity in atherosclerosis? J Intern Med 2022; 291:530-532. [PMID: 35028996 DOI: 10.1111/joim.13436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Daniel F J Ketelhuth
- Division of Cardiovascular Medicine, Center for Molecular Medicine, Department of Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden.,Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
4
|
Atherosclerosis severity in patients with familial hypercholesterolemia: The role of T and B lymphocytes. ATHEROSCLEROSIS PLUS 2022; 48:27-36. [PMID: 36644561 PMCID: PMC9833267 DOI: 10.1016/j.athplu.2022.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 02/01/2023]
Abstract
Background and aims Familial hypercholesterolemia (FH) is characterized by lifelong exposure to high LDL-c concentrations and premature atherosclerotic cardiovascular disease; nevertheless, disease severity can be heterogeneous.We aimed at evaluating if the immune-inflammatory system could modulate atherosclerosis burden in FH. Methods From a cohort of subjects with confirmed FH (Dutch Lipid Clinic Network and genotype), 92 patients receiving high-intensity lipid-lowering therapy (statin ± ezetimibe) were included. The extension and severity of coronary atherosclerosis was assessed by standardized reporting systems (CAD-RADS) for coronary computed tomography angiography (CCTA) and coronary artery calcium (CAC) scores. Lipids, apolipoproteins, anti-oxLDL and anti-apolipoprotein B-D peptide (anti-ApoB-D) autoantibodies (IgM and IgG), lymphocytes subtypes, platelet, monocyte and endothelial microparticles (MP), IgM levels (circulating or produced by B1 cells) and cytokines in the supernatant of cultured cells were determined. Multiple linear regression models evaluated associations of these biomarkers with CAC and CAD-RADS scores. Results In univariate analysis CAC correlated with age, systolic blood pressure, TCD4+ cells, and titers of IgM anti-ApoB-D. In multiple linear regression [ANOVA F = 2.976; p = 0.024; R2 = 0.082), CD4+T lymphocytes (B = 35.289; beta = 0.277; p = 0.010; 95%CI for B 8.727 to 61.851), was independently associated with CAC. CAD-RADS correlated with age, systolic blood pressure, titers of IgM anti-ApoB-D, and endothelial MP in univariate analysis. In multiple linear regression, [ANOVA F = 2.790; p = 0.032; R2 = 0.119), only age (B = 0.027; beta = 0.234; p = 0.049; 95% CI for B 0.000 to 0.053) was independent predictor. Conclusions In subjects with FH, under high-intensity lipid-lowering therapy, age and CD4+T cells were associated to atherosclerosis burden.
Collapse
|
5
|
Afshar M, Yazdan-Ashoori S, Engert JC, Thanassoulis G. Drugs for Prevention and Treatment of Aortic Stenosis: How Close Are We? Can J Cardiol 2021; 37:1016-1026. [DOI: 10.1016/j.cjca.2021.02.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/18/2021] [Accepted: 02/25/2021] [Indexed: 12/25/2022] Open
|
6
|
Biphasic Production of Anti-ApoB100 Autoantibodies in Obese Humans and Mice. Pharmaceuticals (Basel) 2021; 14:ph14040330. [PMID: 33916621 PMCID: PMC8065440 DOI: 10.3390/ph14040330] [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: 02/17/2021] [Revised: 03/23/2021] [Accepted: 04/02/2021] [Indexed: 12/03/2022] Open
Abstract
Obesity is associated with autoimmunity, a phenomenon considered as harmful. Here we show that obese mice and humans produce IgG-type autoantibodies that specifically recognize apolipoprotein B-100 (ApoB100), its native epitope p210, and the synthetic p210 mimotope pB1. By contrast, antibodies against epitopes p45 and p240, which have been associated with atherosclerosis, were not detected in either the humans or mice. In a longitudinal analysis of high fat diet-fed mice, autoantibody production rose with increasing body weight, then decreased and plateaued at morbid obesity. Likewise, in a cross-sectional analysis of sera from 148 human volunteers spanning a wide BMI range and free of comorbidities, the immunoreactivity increased and then decreased with increasing BMI. Thus, the obesity-related ApoB100-specific natural autoantibodies characteristically showed the same epitope recognition, IgG-type, and biphasic serum levels in humans and mice. We previously reported that a pB1-based vaccine induces similar antibodies and can prevent obesity in mice. Therefore, our present results suggest that autoantibodies directed against native ApoB100 may mitigate obesity, and that the vaccination approach may be effective in humans.
Collapse
|
7
|
Sniderman AD, Thanassoulis G, Glavinovic T, Navar AM, Pencina M, Catapano A, Ference BA. Apolipoprotein B Particles and Cardiovascular Disease: A Narrative Review. JAMA Cardiol 2020; 4:1287-1295. [PMID: 31642874 DOI: 10.1001/jamacardio.2019.3780] [Citation(s) in RCA: 322] [Impact Index Per Article: 64.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Importance The conventional model of atherosclerosis presumes that the mass of cholesterol within very low-density lipoprotein particles, low-density lipoprotein particles, chylomicron, and lipoprotein (a) particles in plasma is the principal determinant of the mass of cholesterol that will be deposited within the arterial wall and will drive atherogenesis. However, each of these particles contains one molecule of apolipoprotein B (apoB) and there is now substantial evidence that apoB more accurately measures the atherogenic risk owing to the apoB lipoproteins than does low-density lipoprotein cholesterol or non-high-density lipoprotein cholesterol. Observations Cholesterol can only enter the arterial wall within apoB particles. However, the mass of cholesterol per apoB particle is variable. Therefore, the mass of cholesterol that will be deposited within the arterial wall is determined by the number of apoB particles that are trapped within the arterial wall. The number of apoB particles that enter the arterial wall is determined primarily by the number of apoB particles within the arterial lumen. However, once within the arterial wall, smaller cholesterol-depleted apoB particles have a greater tendency to be trapped than larger cholesterol-enriched apoB particles because they bind more avidly to the glycosaminoglycans within the subintimal space of the arterial wall. Thus, a cholesterol-enriched particle would deposit more cholesterol than a cholesterol-depleted apoB particle whereas more, smaller apoB particles that enter the arterial wall will be trapped than larger apoB particles. The net result is, with the exceptions of the abnormal chylomicron remnants in type III hyperlipoproteinemia and lipoprotein (a), all apoB particles are equally atherogenic. Conclusions and Relevance Apolipoprotein B unifies, amplifies, and simplifies the information from the conventional lipid markers as to the atherogenic risk attributable to the apoB lipoproteins.
Collapse
Affiliation(s)
- Allan D Sniderman
- Mike and Valeria Rosenbloom Centre for Cardiovascular Prevention, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| | - George Thanassoulis
- Mike and Valeria Rosenbloom Centre for Cardiovascular Prevention, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| | | | - Ann Marie Navar
- Duke Clinical Research Institute, Durham, North Carolina.,Associate Editor
| | - Michael Pencina
- Duke University School of Medicine, Durham, North Carolina.,Deputy Editor for Statistics
| | - Alberico Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Multimedica IRCCS, Milano, Italy
| | - Brian A Ference
- Centre for Naturally Randomized Trials, University of Cambridge, Cambridge, United Kingdom.,Institute for Advanced Studies, University of Bristol, Bristol, United Kingdom.,MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
8
|
Ketelhuth DFJ. The immunometabolic role of indoleamine 2,3-dioxygenase in atherosclerotic cardiovascular disease: immune homeostatic mechanisms in the artery wall. Cardiovasc Res 2020; 115:1408-1415. [PMID: 30847484 DOI: 10.1093/cvr/cvz067] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 01/30/2019] [Accepted: 03/05/2019] [Indexed: 01/05/2023] Open
Abstract
Coronary heart disease and stroke, the two most common cardiovascular diseases worldwide, are triggered by complications of atherosclerosis. Atherosclerotic plaques are initiated by a maladaptive immune response triggered by accumulation of lipids in the artery wall. Hence, disease is influenced by several non-modifiable and modifiable risk factors, including dyslipidaemia, hypertension, smoking, and diabetes. Indoleamine 2,3-dioxygenase (IDO), the rate-limiting enzyme in the kynurenine pathway of tryptophan (Trp) degradation, is modulated by inflammation and regarded as a key molecule driving immunotolerance and immunosuppressive mechanisms. A large body of evidence indicates that IDO-mediated Trp metabolism is involved directly or indirectly in atherogenesis. This review summarizes evidence from basic and clinical research showing that IDO is a major regulatory enzyme involved in the maintenance of immunohomeostasis in the vascular wall, as well as current knowledge about promising targets for the development of new anti-atherosclerotic drugs.
Collapse
Affiliation(s)
- Daniel F J Ketelhuth
- Cardiovascular Medicine Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden.,Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, Univ. of Southern Denmark, J. B. Winsløws Vej 21(3), Odense C, Denmark
| |
Collapse
|
9
|
Du R, Wu X, Peng K, Lin L, Li M, Xu Y, Xu M, Chen Y, Li D, Lu J, Bi Y, Wang W, Ning G. Serum apolipoprotein B is associated with increased risk of metabolic syndrome among middle-aged and elderly Chinese: A cross-sectional and prospective cohort study. J Diabetes 2019; 11:752-760. [PMID: 30652427 DOI: 10.1111/1753-0407.12904] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/02/2019] [Accepted: 01/12/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Serum apolipoprotein (apo) B has been associated with an increased risk of atherosclerotic cardiovascular diseases. However, findings on the association between apoB and the risk of metabolic syndrome (MetS) are inconsistent. The purpose of this study was to investigate the association between serum apoB and MetS risk in Chinese population. METHODS A baseline survey was conducted in a population-based cohort of 10 340 adults aged ≥40 years in Shanghai, China, in 2010. A follow-up visit was conducted to assess incident diabetes in 2015. RESULTS At baseline, 2794 of 10 340 participants (27.02%) had MetS. Serum apoB was significantly associated with an increased risk of prevalent MetS. Multivariable-adjusted odds ratios and 95% confidence intervals (CIs) for quintiles 2-5 compared with quintile 1 (reference) were 1.29 (1.02-1.63), 1.47 (1.18-1.84), 1.32 (1.06-1.65), and 2.02 (1.61-2.51), respectively (Ptrend < 0.05). During an average of 5.1 years follow-up, 4627 individuals without MetS at baseline showed a significant association between apoB and the risk of incident MetS. Multivariable-adjusted risk ratios (95% CIs) for subjects in apoB quintiles 2-5 compared with the reference were 1.43 (1.13-1.82), 1.57 (1.25-1.98), 1.74 (1.38-2.18), and 2.07 (1.66-2.58), respectively (Ptrend < 0.05). Stratified analysis suggested that the above association was much stronger among normal weight individuals than in those who were overweight or obese. CONCLUSION These cross-sectional and prospective studies provide evidence that serum apoB is associated with existing MetS and is a possible predictor of the risk of MetS, especially among normal weight individuals.
Collapse
Affiliation(s)
- Rui Du
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueyan Wu
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kui Peng
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Lin
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mian Li
- Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Yu Xu
- Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Min Xu
- Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Yuhong Chen
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Donghui Li
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jieli Lu
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Yufang Bi
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Weiqing Wang
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| |
Collapse
|
10
|
Faulin TDES, Kazuma SM, Tripodi GL, Cavalcante MF, Wakasuqui F, Oliveira CLP, Degenhardt MFDS, Michaloski J, Giordano RJ, Ketelhuth DFJ, Abdalla DSP. Proinflammatory Action of a New Electronegative Low-Density Lipoprotein Epitope. Biomolecules 2019; 9:biom9080386. [PMID: 31434316 PMCID: PMC6723646 DOI: 10.3390/biom9080386] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/03/2019] [Accepted: 08/13/2019] [Indexed: 01/02/2023] Open
Abstract
The electronegative low-density lipoprotein, LDL (-), is an endogenously modified LDL subfraction with cytotoxic and proinflammatory actions on endothelial cells, monocytes, and macrophages contributing to the progression of atherosclerosis. In this study, epitopes of LDL (-) were mapped using a phage display library of peptides and monoclonal antibodies reactive to this modified lipoprotein. Two different peptide libraries (X6 and CX8C for 6- and 8-amino acid-long peptides, respectively) were used in the mapping. Among all tested peptides, two circular peptides, P1A3 and P2C7, were selected based on their high affinities for the monoclonal antibodies. Small-angle X-ray scattering analysis confirmed their structures as circular rings. P1A3 or P2C7 were quickly internalized by bone marrow-derived murine macrophages as shown by confocal microscopy. P2C7 increased the expression of TNFα, IL-1 β and iNOS as well as the secretion of TNFα, CCL2, and nitric oxide by murine macrophages, similar to the responses induced by LDL (-), although less intense. In contrast, P1A3 did not show pro-inflammatory effects. We identified a mimetic epitope associated with LDL (-), the P2C7 circular peptide, that activates macrophages. Our data suggest that this conformational epitope represents an important danger-associated molecular pattern of LDL (-) that triggers proinflammatory responses.
Collapse
Affiliation(s)
- Tanize do Espirito Santo Faulin
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Soraya Megumi Kazuma
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Gustavo Luis Tripodi
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Marcela Frota Cavalcante
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Felipe Wakasuqui
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | | | | | - Jussara Michaloski
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, Sao Paulo 05508-000, SP, Brazil
| | - Ricardo José Giordano
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, Sao Paulo 05508-000, SP, Brazil
| | - Daniel Francisco Jacon Ketelhuth
- Centre for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, 17164 Stockholm, Sweden
- Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark (SDU), 5000 Odense, Denmark
| | - Dulcineia Saes Parra Abdalla
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil.
| |
Collapse
|
11
|
Jiang X, Wang F, Wang Y, Gisterå A, Roy J, Paulsson-Berne G, Hedin U, Lerman A, Hansson GK, Herrmann J, Yan ZQ. Inflammasome-Driven Interleukin-1α and Interleukin-1β Production in Atherosclerotic Plaques Relates to Hyperlipidemia and Plaque Complexity. JACC Basic Transl Sci 2019; 4:304-317. [PMID: 31312755 PMCID: PMC6610158 DOI: 10.1016/j.jacbts.2019.02.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 11/24/2018] [Accepted: 02/11/2019] [Indexed: 01/21/2023]
Abstract
Genetic and functional evidence suggests that there are additional inflammasome pathways, besides NLRP3, that contribute to IL-1 generation in human atherosclerotic plaques. Plaque generation of mature IL-1β is accompanied by secretion of similar levels of IL-1α, through a mechanism controlled by NLRP3 and caspase-1. Plaque IL-1β production is higher in patients with uncontrolled hyperlipidemia, on no or low-dose statin therapy, or with complex plaque imaging features. The present study lends support to high-intensity cholesterol lowering and anti-IL-1-directed therapies for patients at high cardiovascular risk.
CANTOS (Canakinumab Antiinflammatory Thrombosis Outcome Study) confirmed interleukin (IL)–1β as an appealing therapeutic target for human atherosclerosis and related complications. However, there are serious gaps in our understanding of IL-1 production in atherosclerosis. Herein the authors show that complex plaques, or plaques derived from patients with suboptimally controlled hyperlipidemia, or on no or low-intensity statin therapy, demonstrated higher recruitable IL-1β production. Generation of mature IL-1β was matched by IL-1α release, and both were attenuated by inhibition of NLR family pyrin domain containing 3 or caspase. These findings support the inflammasome as the main pathway for IL-1α/β generation in atherosclerosis and high-intensity lipid-lowering therapies as primary and additional anti-IL-1-directed therapies as secondary interventions in high-risk patients.
Collapse
Key Words
- ASC, apoptosis-associated speck-like protein containing a CARD
- ATP, adenosine 5′-triphosphate disodium salt hydrate
- BiKE, Biobank of Karolinska Carotid Endarterectomies
- CT, Computerized tomographic scanning
- IL, interleukin
- LDL, low-density lipoprotein
- LPS, lipopolysaccharide
- NLRC, nucleotide-binding oligomerization domain, leucine-rich repeat and CARD domain–containing protein
- NLRP, nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain–containing protein
- PBS, phosphate-buffered saline
- atherosclerosis
- hypercholesterolemia
- inflammasome
- inflammation
- interleukin-1
- mRNA, messenger ribonucleic acid
Collapse
Affiliation(s)
- Xintong Jiang
- Experimental Cardiovascular Research Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Feilong Wang
- Department of Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Yajuan Wang
- Experimental Cardiovascular Research Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Anton Gisterå
- Experimental Cardiovascular Research Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Joy Roy
- Department of Vascular Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Gabrielle Paulsson-Berne
- Experimental Cardiovascular Research Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ulf Hedin
- Department of Vascular Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Amir Lerman
- Department of Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Göran K Hansson
- Experimental Cardiovascular Research Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Joerg Herrmann
- Department of Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Zhong-Qun Yan
- Experimental Cardiovascular Research Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
12
|
Sniderman AD. Did the ACC/AHA/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA cholesterol guidelines get apoB right? J Clin Lipidol 2019; 13:360-366. [DOI: 10.1016/j.jacl.2019.05.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
13
|
Ma Z, Ketelhuth DFJ, Wirström T, Ohki T, Forteza MJ, Wang H, Grill V, Wollheim CB, Björklund A. Increased uptake of oxLDL does not exert lipotoxic effects in insulin-secreting cells. J Mol Endocrinol 2019; 62:159-168. [PMID: 30917339 DOI: 10.1530/jme-18-0146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/27/2019] [Indexed: 01/13/2023]
Abstract
Modified lipoproteins can negatively affect beta cell function and survival. However, the mechanisms behind interactions of modified lipoproteins with beta cells - and in particular, relationships to increased uptake - are only partly clarified. By over-expressing the scavenger receptor CD36 (Tet-on), we increased the uptake of fluorescent low-density modified lipoprotein (oxLDL) into insulin-secreting INS-1 cells. The magnitude of uptake followed the degree of CD36 over-expression. CD36 over-expression increased concomitant efflux of 3H-cholesterol in proportion to the cellular contents of 3H-cholesterol. Exposure to concentrations of oxLDL from 20 to 100 µg/mL dose-dependently increased toxicity (evaluated by MTT) as well as apoptosis. However, the increased uptake of oxLDL due to CD36 over-expression did not exert additive effects on oxLDL toxicity - neither on viability, nor on glucose-induced insulin release and cellular content. Reciprocally, blocking CD36 receptors by Sulfo-N-Succinimidyl Oleate decreased the uptake of oxLDL but did not diminish the toxicity. Pancreatic islets of CD36-/- mice displayed reduced uptake of 3H-cholesterol-labeled oxLDL vs wild type but similar toxicity to oxLDL. OxLDL was found to increase the expression of CD36 in islets and INS-1 cells. In summary, given the experimental conditions, our results indicate that (1) increased uptake of oxLDL is not responsible for toxicity of oxLDL, (2) increased efflux of the cholesterol moiety of oxLDL counterbalances, at least in part, increased uptake and (3) oxLDL participates in the regulation of CD36 in pancreatic islets and in INS-1 cells.
Collapse
Affiliation(s)
- Z Ma
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - D F J Ketelhuth
- Department of Medicine, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - T Wirström
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - T Ohki
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - M J Forteza
- Department of Medicine, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - H Wang
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - V Grill
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Institute of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - C B Wollheim
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - A Björklund
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
14
|
|
15
|
Abstract
Whether apoB adds significantly to the assessment of the risk and therapy of the atherogenic dyslipoproteinemias has been vigorously contested over many years. That trapping of apoB lipoprotein particles within the arterial wall is fundamental to the initiation and maturation of atherosclerotic lesions within the arterial wall is now widely accepted. At the same time, the concept that primary prevention should be based on the risk of a cardiovascular event, a measure that integrates the effects of age, sex, blood pressure, lipids and other factors, has also become widely accepted. Within the risk framework, the issue becomes whether apoB adds significantly to the assessment of risk. On the other hand, it can be argued that the risk model undervalues how important a role that LDL and blood pressure play as causes of atherosclerosis and that when considered as causes, the importance of apoB emerges. These are the two sides of the debate that will be presented in the article that follows: one will highlight the pros of measuring apoB, the second the cons. The reader can make up his or her mind which side of the issue they favour.
Collapse
Affiliation(s)
- Allan D Sniderman
- McGill University Health Centre, Royal Victoria Hospital, 1001 Decarie Boulevard, Montreal, Quebec, H4A 3J1, Canada.
| | - Jennifer G Robinson
- Departments of Epidemiology and Internal Medicine, Division of Cardiology, University of Iowa, 145 N Riverside Dr S455 CPHB, Iowa City, IA, 52242, United States
| |
Collapse
|
16
|
Ray M, Autieri MV. Regulation of pro- and anti-atherogenic cytokines. Cytokine 2017; 122:154175. [PMID: 29221669 DOI: 10.1016/j.cyto.2017.09.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/29/2017] [Accepted: 09/26/2017] [Indexed: 12/21/2022]
Abstract
Despite advances in prevention and treatment, vascular diseases continue to account for significant morbidity and mortality in the developed world. Incidence is expected to worsen as the number of patients with common co-morbidities linked with atherosclerotic vascular disease, such as obesity and diabetes, continues to increase, reaching epidemic proportions. Atherosclerosis is a lipid-driven vascular inflammatory disease involving multiple cell types in various stages of inflammation, activation, apoptosis, and necrosis. One commonality among these cell types is that they are activated and communicate with each other in a paracrine fashion via a complex network of cytokines. Cytokines mediate atherogenesis by stimulating expression of numerous proteins necessary for induction of a host of cellular responses, including inflammation, extravasation, proliferation, apoptosis, and matrix production. Cytokine expression is regulated by a number of transcriptional and post-transcriptional mechanisms. In this context, proteins that control and fine-tune cytokine expression can be considered key players in development of atherosclerosis and also represent targets for rational drug therapy to combat this disease. This review will describe the cellular and molecular mechanisms that drive atherosclerotic plaque progression and present key cytokines that participate in this process. We will also describe RNA binding proteins that mediate cytokine mRNA stability and regulate cytokine abundance. Identification and characterization of the cytokines and proteins that regulate their abundance are essential to our ability to identify therapeutic approaches to ameliorate atherosclerotic vascular disease.
Collapse
Affiliation(s)
- Mitali Ray
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Michael V Autieri
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
17
|
Abstract
Cardiovascular disease is the leading cause of death worldwide, both in the general population and among patients with chronic kidney disease (CKD). In most cases, the underlying cause of the cardiovascular event is atherosclerosis - a chronic inflammatory disease. CKD accelerates atherosclerosis via augmentation of inflammation, perturbation of lipid metabolism, and other mechanisms. In the artery wall, subendothelial retention of plasma lipoproteins triggers monocyte-derived macrophages and T helper type 1 (TH1) cells to form atherosclerotic plaques. Inflammation is initiated by innate immune reactions to modified lipoproteins and is perpetuated by TH1 cells that react to autoantigens from the apolipoprotein B100 protein of LDL. Other T cells are also active in atherosclerotic lesions; regulatory T cells inhibit pathological inflammation, whereas TH17 cells can promote plaque fibrosis. The slow build-up of atherosclerotic plaques is asymptomatic, but plaque rupture or endothelial erosion can induce thrombus formation, leading to myocardial infarction or ischaemic stroke. Targeting risk factors for atherosclerosis has reduced mortality, but a need exists for novel therapies to stabilize plaques and to treat arterial inflammation. Patients with CKD would likely benefit from such preventive measures.
Collapse
Affiliation(s)
- Anton Gisterå
- Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, SE-17176 Stockholm, Sweden
| | - Göran K Hansson
- Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, SE-17176 Stockholm, Sweden
| |
Collapse
|
18
|
Gisterå A, Hermansson A, Strodthoff D, Klement ML, Hedin U, Fredrikson GN, Nilsson J, Hansson GK, Ketelhuth DFJ. Vaccination against T-cell epitopes of native ApoB100 reduces vascular inflammation and disease in a humanized mouse model of atherosclerosis. J Intern Med 2017; 281:383-397. [PMID: 28194913 DOI: 10.1111/joim.12589] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND AND OBJECTIVES The T-cell response to low-density lipoprotein (LDL) in the vessel wall plays a critical role in atherosclerotic plaque formation and stability. In this study, we used a new translational approach to investigate epitopes from human apolipoprotein B100 (ApoB100), the protein component of LDL, which triggers T-cell activation. We also evaluated the potential of two selected native ApoB100 epitopes to modulate atherosclerosis in human ApoB100-transgenic Ldlr-/- (HuBL) mice. METHODS AND RESULTS HuBL mice were immunized with human atherosclerotic plaque homogenate to boost cellular autoimmune response to tissue-derived ApoB100 epitopes. In vitro challenge of splenocytes from immunized mice with a library of overlapping native peptides covering human ApoB100 revealed several sequences eliciting T-cell proliferation. Of these sequences, peptide (P) 265 and P295 were predicted to bind several human leucocyte antigen (HLA) haplotypes and induced high levels of interferon (IFN)-γ. Vaccination of HuBL mice with these peptides mounted a strong adaptive immune response to native ApoB100, including high levels of epitope-specific plasma IgGs. Interestingly, P265 and P295 vaccines significantly decreased plaque size, reduced macrophage infiltration and increased IgG1 deposition in the plaques. Purified IgGs from vaccinated mice displayed anti-inflammatory properties against macrophages in vitro, reducing their response to LPS in a dose-dependent manner. CONCLUSION We identified two specific epitopes from human native ApoB100 that trigger T-cell activation and protect HuBL mice against atherosclerosis when used in a vaccine. Our data suggest that vaccination-induced protective mechanisms may be mediated at least in part through specific antibody responses to LDL that inhibit macrophage activation.
Collapse
Affiliation(s)
- A Gisterå
- Department of Medicine, Cardiovascular Medicine Unit, Center for Molecular Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - A Hermansson
- Department of Medicine, Cardiovascular Medicine Unit, Center for Molecular Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - D Strodthoff
- Department of Medicine, Cardiovascular Medicine Unit, Center for Molecular Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - M L Klement
- Department of Medicine, Cardiovascular Medicine Unit, Center for Molecular Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - U Hedin
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden.,Department of Vascular Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - G N Fredrikson
- Department of Clinical Sciences, Skåne University Hospital, Lund University, Malmö, Sweden
| | - J Nilsson
- Department of Clinical Sciences, Skåne University Hospital, Lund University, Malmö, Sweden
| | - G K Hansson
- Department of Medicine, Cardiovascular Medicine Unit, Center for Molecular Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - D F J Ketelhuth
- Department of Medicine, Cardiovascular Medicine Unit, Center for Molecular Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW Lipoprotein(a) [Lp(a)] is the strongest independent genetic risk factor for both myocardial infarction and aortic stenosis. It has also been associated with other forms of atherosclerotic cardiovascular disease (CVD) including ischemic stroke. Its levels are genetically determined and remain fairly stable throughout life. Elevated Lp(a), above 50 mg/dl, affects one in five individuals worldwide. RECENT FINDINGS Herein, we review the recent epidemiologic and genetic evidence supporting the causal role of Lp(a) in CVD, highlight recommendations made by European and Canadian guidelines regarding Lp(a) and summarize the rapidly evolving field of Lp(a)-lowering therapies including antisense therapies and Proprotein Convertase Subtilisin/Kexin Type 9 inhibitors. SUMMARY With novel therapies on the horizon, Lp(a) is poised to gain significant clinical relevance and its lowering could have a significant impact on the burden of CVD. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Mehdi Afshar
- aDepartment of Medicine, McGill University bPreventive and Genomic Cardiology, McGill University Health Center and Research Institute cDepartment of Clinical Epidemiology, McGill University Health Center, Montreal, Quebec, Canada
| | | |
Collapse
|
20
|
Wilkins JT, Sniderman A, Lloyd-Jones DM. Reply. J Am Coll Cardiol 2016; 67:3024-5. [DOI: 10.1016/j.jacc.2016.03.579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 03/22/2016] [Indexed: 10/21/2022]
|
21
|
Affiliation(s)
- Kevin Jon Williams
- From the Section of Endocrinology, Diabetes, & Metabolism, Temple University School of Medicine, Philadelphia, PA (K.J.W.); Department of Molecular and Clinical Medicine, Sahlgrenska Academy of the University of Gothenburg, Göteborg, Sweden (K.J.W.); Departments of Medicine, Pathology & Cell Biology, and Physiology, Columbia University Medical Center, New York, NY (I.T.); and Department of Medicine (Cardiology), the Marc and Ruti Bell Program in Vascular Biology and The Center for the Prevention of Cardiovascular Disease, NYU School of Medicine, New York, NY (E.A.F.).
| | - Ira Tabas
- From the Section of Endocrinology, Diabetes, & Metabolism, Temple University School of Medicine, Philadelphia, PA (K.J.W.); Department of Molecular and Clinical Medicine, Sahlgrenska Academy of the University of Gothenburg, Göteborg, Sweden (K.J.W.); Departments of Medicine, Pathology & Cell Biology, and Physiology, Columbia University Medical Center, New York, NY (I.T.); and Department of Medicine (Cardiology), the Marc and Ruti Bell Program in Vascular Biology and The Center for the Prevention of Cardiovascular Disease, NYU School of Medicine, New York, NY (E.A.F.)
| | - Edward A Fisher
- From the Section of Endocrinology, Diabetes, & Metabolism, Temple University School of Medicine, Philadelphia, PA (K.J.W.); Department of Molecular and Clinical Medicine, Sahlgrenska Academy of the University of Gothenburg, Göteborg, Sweden (K.J.W.); Departments of Medicine, Pathology & Cell Biology, and Physiology, Columbia University Medical Center, New York, NY (I.T.); and Department of Medicine (Cardiology), the Marc and Ruti Bell Program in Vascular Biology and The Center for the Prevention of Cardiovascular Disease, NYU School of Medicine, New York, NY (E.A.F.)
| |
Collapse
|
22
|
Thanassoulis G. Lipoprotein (a) in calcific aortic valve disease: from genomics to novel drug target for aortic stenosis. J Lipid Res 2015; 57:917-24. [PMID: 26685327 DOI: 10.1194/jlr.r051870] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Indexed: 12/21/2022] Open
Abstract
Calcific aortic stenosis (AS) is the most common form of valve disease in the Western world and affects over 2.5 million individuals in North America. Despite the large burden of disease, there are no medical treatments to slow the development of AS, due at least in part to our incomplete understanding of its causes. The Cohorts for Heart and Aging Research in Genetic Epidemiology extra-coronary calcium consortium reported a genome-wide association study demonstrating that genetic variants in LPA are strongly associated with aortic valve (AV) calcium and clinical AS. Using a Mendelian randomization study design, it was demonstrated that the effect of this genetic variant is mediated by plasma lipoprotein (a) [Lp(a)], directly implicating elevations in Lp(a) as a cause of AV calcium and progression to AS. This discovery has sparked intense interest in Lp(a) as a modifiable cause for AV disease. Herein, we will review the mounting epidemiological and genetic findings in support of Lp(a)-mediated valve disease, discuss potential mechanisms underlying this observation, and outline the steps to translate this discovery to a much needed novel preventive and/or therapeutic strategy for AV disease.
Collapse
Affiliation(s)
- George Thanassoulis
- Preventive and Genomic Cardiology, McGill University Health Center and Research Institute and Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
23
|
Fonseca HAR, Fonseca FA, Lins LC, Monteiro AM, Bianco HT, Brandão SA, Povoa RM, Juliano L, Figueiredo-Neto AM, Boschcov P, Gidlund M, Izar MC. Antihypertensive therapy increases natural immunity response in hypertensive patients. Life Sci 2015; 143:124-30. [PMID: 26514303 DOI: 10.1016/j.lfs.2015.10.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 10/04/2015] [Accepted: 10/23/2015] [Indexed: 12/21/2022]
Abstract
AIMS The aim of this work was to evaluate the effects of treatment of hypertension on the autoantibodies to apolipoprotein B-derived peptides (anti-ApoB-D peptide Abs) response, inflammation markers and vascular function. MAIN METHODS Eighty-eight patients with hypertension (stage 1 or 2) were recruited and advised to receive perindopril (4mg), hydrochlorothiazide (25mg), or indapamide (1.5mg) for 12weeks in a blinded fashion. Office and 24-h ambulatory blood pressure monitoring (24h ABPM), flow-mediated dilatation (FMD), nitrate-induced dilatation (NID), titers of IgG and IgM anti-ApoB-D peptide Abs, hsCRP, and interleukins (IL-8 and IL-10) were evaluated at baseline and 12weeks after therapies. KEY FINDINGS All treatments reduced office BP, and improved FMD (P<0.05 vs. baseline). The NID was improved only in the perindopril arm (P<0.05 vs. baseline). The 24h-ABPM was reduced with perindopril and hydrochlorothiazide therapies (P<0.05 vs. baseline), but not with indapamide, and this effect was followed by increase in titers of IgM Anti-ApoB-D peptide Abs (P<0.05 vs. baseline), without modifications in titers IgG Anti-ApoB-D peptide Abs and interleukins. Multivariable regression analysis has shown that change in the titers of IgM anti-ApoB-D peptide was associated with the changes in FMD (β -0.347; P<0.05). SIGNIFICANCE These findings shed light to a possible modulator effect of the antihypertensive therapy on the natural immunity responses and vascular function.
Collapse
Affiliation(s)
| | - Francisco A Fonseca
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Lívia C Lins
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Andrea M Monteiro
- Department of Immunology, Institute of Biomedical Sciences IV, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Henrique T Bianco
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Sergio A Brandão
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Rui M Povoa
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Luiz Juliano
- Department of Biophysics, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | | | - Paulo Boschcov
- Department of Biophysics, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Magnus Gidlund
- Department of Immunology, Institute of Biomedical Sciences IV, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Maria C Izar
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
24
|
Liu H, Wei X, Kong L, Liu X, Cheng L, Yan S, Zhang X, Chen L. NOD2 is involved in the inflammatory response after cerebral ischemia-reperfusion injury and triggers NADPH oxidase 2-derived reactive oxygen species. Int J Biol Sci 2015; 11:525-35. [PMID: 25892960 PMCID: PMC4400384 DOI: 10.7150/ijbs.10927] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 02/23/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Increasing evidences suggest that innate immunity is involved in cerebral ischemia-reperfusion (I/R) injury, but the liable innate immune receptors have not been completely elucidated. Here, we explored the role of the nucleotide-binding oligomerization domain (NOD)2, a member of the cytosolic NOD-like receptor family, in acute focal cerebral I/R injury. METHODS An in vivo middle cerebral artery occlusion (MCAO) model that in wild type (WT) and NOD2 deficient (NOD2(-/-)) mice and in vitro model of oxygen glucose deprivation and reoxygenation (OGD/R) in cultured primary microglia and astrocytes were used to investigate the expression of NOD2 and explore the roles of NOD2 in ischemic stroke. RESULTS Our results showed that NOD2 expression was significantly increased in microglia and astrocytes in response to the I/R insult. Pretreatment with muramyl dipeptide, an extrinsic ligand of NOD2, significantly increased the infarct volume and neurological dysfunction in mice subjected to MCAO. Genetic ablation of the NOD2 gene significantly improved stroke outcomes and reduced inflammation, as evidenced by a lower expression of the pro-inflammatory cytokines IL-1β, IL-6 and TNFα in conjunction with attenuated activation of nuclear factor κB (NF-κB), p38 mitogen activated protein kinases (MAPK) and JNK. Moreover, NOD2 deficiency prevented the upregulation of the NADPH oxidase (NOX) 2 and ROS generation induced by I/R. Mechanistically, NOD2-induced production of IL-6 in primary cultured microglia was mediated through activation of NOX2. CONCLUSIONS This study showed the contribution of NOD2 to inflammatory response and provided direct evidence that NOX2-mediated oxidative stress as an important target molecule linked NOD2 to inflammatory damage in ischemic stroke. Pharmacological targeting of NOD2-mediated inflammatory response at multiple levels may help design a new approach to develop therapeutic strategies for prevention of deterioration of cerebral function and for the treatment of stroke.
Collapse
Affiliation(s)
- Huiqing Liu
- 1. Department of Pharmacology, School of Medicine, Shandong University, Wenhua West Road 44, Jinan, Shandong 250012, P.R. China
| | - Xinbing Wei
- 1. Department of Pharmacology, School of Medicine, Shandong University, Wenhua West Road 44, Jinan, Shandong 250012, P.R. China
| | - Lingjun Kong
- 1. Department of Pharmacology, School of Medicine, Shandong University, Wenhua West Road 44, Jinan, Shandong 250012, P.R. China
| | - Xiaoqian Liu
- 1. Department of Pharmacology, School of Medicine, Shandong University, Wenhua West Road 44, Jinan, Shandong 250012, P.R. China. ; 2. Key Laboratory of Molecular Pharmacology and Drug Evaluation (Ministry of Education of China), School of Pharmacy, Yantai University, Qingquan Road 30, Yantai, Shandong 264005, P.R. China
| | - Li Cheng
- 1. Department of Pharmacology, School of Medicine, Shandong University, Wenhua West Road 44, Jinan, Shandong 250012, P.R. China
| | - Shi Yan
- 3. Department of Obstetrics and Gynaecology, Qilu Hospital of Shandong University, Wenhua West Road 107, Jinan, Shandong 250012, P.R. China
| | - Xiumei Zhang
- 1. Department of Pharmacology, School of Medicine, Shandong University, Wenhua West Road 44, Jinan, Shandong 250012, P.R. China
| | - Lin Chen
- 1. Department of Pharmacology, School of Medicine, Shandong University, Wenhua West Road 44, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
25
|
Johansson ME, Zhang XY, Edfeldt K, Lundberg AM, Levin MC, Borén J, Li W, Yuan XM, Folkersen L, Eriksson P, Hedin U, Low H, Sviridov D, Rios FJ, Hansson GK, Yan ZQ. Innate immune receptor NOD2 promotes vascular inflammation and formation of lipid-rich necrotic cores in hypercholesterolemic mice. Eur J Immunol 2014; 44:3081-92. [PMID: 25042478 DOI: 10.1002/eji.201444755] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 06/16/2014] [Accepted: 07/14/2014] [Indexed: 01/07/2023]
Abstract
Atherosclerosis is an inflammatory disease associated with the activation of innate immune TLRs and nucleotide-binding oligomerization domain-containing protein (NOD)-like receptor pathways. However, the function of most innate immune receptors in atherosclerosis remains unclear. Here, we show that NOD2 is a crucial innate immune receptor influencing vascular inflammation and atherosclerosis severity. 10-week stimulation with muramyl dipeptide (MDP), the NOD2 cognate ligand, aggravated atherosclerosis, as indicated by the augmented lesion burden, increased vascular inflammation and enlarged lipid-rich necrotic cores in Ldlr(-/-) mice. Myeloid-specific ablation of NOD2, but not its downstream kinase, receptor-interacting serine/threonine-protein kinase 2, restrained the expansion of the lipid-rich necrotic core in Ldlr(-/-) chimeric mice. In vitro stimulation of macrophages with MDP enhanced the uptake of oxidized low-density lipoprotein and impaired cholesterol efflux in concordance with upregulation of scavenger receptor A1/2 and downregulation of ATP-binding cassette transporter A1. Ex vivo stimulation of human carotid plaques with MDP led to increased activation of inflammatory signaling pathways p38 MAPK and NF-κB-mediated release of proinflammatory cytokines. Altogether, this study suggests that NOD2 contributes to the expansion of the lipid-rich necrotic core and promotes vascular inflammation in atherosclerosis.
Collapse
Affiliation(s)
- Maria E Johansson
- Center for Molecular Medicine, Department of Medicine, Karolinska Institute, Stockholm, Sweden; Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Ait-Oufella H, Sage AP, Mallat Z, Tedgui A. Adaptive (T and B cells) immunity and control by dendritic cells in atherosclerosis. Circ Res 2014; 114:1640-60. [PMID: 24812352 DOI: 10.1161/circresaha.114.302761] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Chronic inflammation in response to lipoprotein accumulation in the arterial wall is central in the development of atherosclerosis. Both innate and adaptive immunity are involved in this process. Adaptive immune responses develop against an array of potential antigens presented to effector T lymphocytes by antigen-presenting cells, especially dendritic cells. Functional analysis of the role of different T-cell subsets identified the Th1 responses as proatherogenic, whereas regulatory T-cell responses exert antiatherogenic activities. The effect of Th2 and Th17 responses is still debated. Atherosclerosis is also associated with B-cell activation. Recent evidence established that conventional B-2 cells promote atherosclerosis. In contrast, innate B-1 B cells offer protection through secretion of natural IgM antibodies. This review discusses the recent development in our understanding of the role of T- and B-cell subsets in atherosclerosis and addresses the role of dendritic cell subpopulations in the control of adaptive immunity.
Collapse
Affiliation(s)
- Hafid Ait-Oufella
- From INSERM UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (H.A.-O., Z.M., A.T.); Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Paris, France (H.A.-O.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S., Z.M.)
| | | | | | | |
Collapse
|
27
|
Libby P, Tabas I, Fredman G, Fisher EA. Inflammation and its resolution as determinants of acute coronary syndromes. Circ Res 2014; 114:1867-79. [PMID: 24902971 PMCID: PMC4078767 DOI: 10.1161/circresaha.114.302699] [Citation(s) in RCA: 389] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 05/12/2014] [Indexed: 12/28/2022]
Abstract
Inflammation contributes to many of the characteristics of plaques implicated in the pathogenesis of acute coronary syndromes. Moreover, inflammatory pathways not only regulate the properties of plaques that precipitate acute coronary syndromes but also modulate the clinical consequences of the thrombotic complications of atherosclerosis. This synthesis will provide an update on the fundamental mechanisms of inflammatory responses that govern acute coronary syndromes and also highlight the ongoing balance between proinflammatory mechanisms and endogenous pathways that can promote the resolution of inflammation. An appreciation of the countervailing mechanisms that modulate inflammation in relation to acute coronary syndromes enriches our fundamental understanding of the pathophysiology of this important manifestation of atherosclerosis. In addition, these insights provide glimpses into potential novel therapeutic interventions to forestall this ultimate complication of the disease.
Collapse
Affiliation(s)
- Peter Libby
- From the Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (P.L.); Department of Medicine, Columbia University Medical Center, New York, NY (I.T.); and Division of Cardiology, Department of Medicine, New York University School of Medicine (E.A.F.).
| | - Ira Tabas
- From the Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (P.L.); Department of Medicine, Columbia University Medical Center, New York, NY (I.T.); and Division of Cardiology, Department of Medicine, New York University School of Medicine (E.A.F.)
| | - Gabrielle Fredman
- From the Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (P.L.); Department of Medicine, Columbia University Medical Center, New York, NY (I.T.); and Division of Cardiology, Department of Medicine, New York University School of Medicine (E.A.F.)
| | - Edward A Fisher
- From the Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (P.L.); Department of Medicine, Columbia University Medical Center, New York, NY (I.T.); and Division of Cardiology, Department of Medicine, New York University School of Medicine (E.A.F.)
| |
Collapse
|
28
|
Assinger A, Wang Y, Butler LM, Hansson GK, Yan ZQ, Söderberg-Nauclér C, Ketelhuth DFJ. Apolipoprotein B100 danger-associated signal 1 (ApoBDS-1) triggers platelet activation and boosts platelet-leukocyte proinflammatory responses. Thromb Haemost 2014; 112:332-41. [PMID: 24816772 DOI: 10.1160/th13-12-1026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 03/20/2014] [Indexed: 12/21/2022]
Abstract
Low-density lipoproteins (LDL), occurring in vivo in both their native and oxidative form, modulate platelet function and thereby contribute to atherothrombosis. We recently identified and demonstrated that 'ApoB100 danger-associated signal 1' (ApoBDS-1), a native peptide derived from Apolipoprotein B-100 (ApoB100) of LDL, induces inflammatory responses in innate immune cells. Platelets are critically involved in the development as well as in the lethal consequences of atherothrombotic diseases, but whether ApoBDS-1 has also an impact on platelet function is unknown. In this study we examined the effect of ApoBDS-1 on human platelet function and platelet-leukocyte interactions in vitro. Stimulation with ApoBDS-1 induced platelet activation, degranulation, adhesion and release of proinflammatory cytokines. ApoBDS-1-stimulated platelets triggered innate immune responses by augmenting leukocyte activation, adhesion and transmigration to/through activated HUVEC monolayers, under flow conditions. These platelet-activating effects were sequence-specific, and stimulation of platelets with ApoBDS-1 activated intracellular signalling pathways, including Ca2+, PI3K/Akt, PLC, and p38- and ERK-MAPK. Moreover, our data indicates that ApoBDS-1-induced platelet activation is partially dependent of positive feedback from ADP on P2Y1 and P2Y12, and TxA2. In conclusion, we demonstrate that ApoBDS-1 is an effective platelet agonist, boosting platelet-leukocyte's proinflammatory responses, and potentially contributing to the multifaceted inflammatory-promoting effects of LDL in the pathogenesis of atherothrombosis.
Collapse
Affiliation(s)
- A Assinger
- Dr. Alice Assinger, Institute of Physiology, Center for Physiology and Pathophysiology, Medical University of Vienna, Schwarzspanierstrasse 17, A-1090 Vienna, Austria, E-mail:
| | | | | | | | | | | | - D F J Ketelhuth
- Dr. Daniel FJ Ketelhuth, Cardiovascular Research Unit, Center for Molecular Medicine, L8:03, Karolinska University Hospital, S-17176 Stockholm, Sweden, Fax: +46 8 313147, E-mail:
| |
Collapse
|
29
|
de Nijs T, Sniderman A, de Graaf J. ApoB versus non-HDL-cholesterol: Diagnosis and cardiovascular risk management. Crit Rev Clin Lab Sci 2013; 50:163-71. [DOI: 10.3109/10408363.2013.847897] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
30
|
Abstract
At least 468 individual genes have been manipulated by molecular methods to study their effects on the initiation, promotion, and progression of atherosclerosis. Most clinicians and many investigators, even in related disciplines, find many of these genes and the related pathways entirely foreign. Medical schools generally do not attempt to incorporate the relevant molecular biology into their curriculum. A number of key signaling pathways are highly relevant to atherogenesis and are presented to provide a context for the gene manipulations summarized herein. The pathways include the following: the insulin receptor (and other receptor tyrosine kinases); Ras and MAPK activation; TNF-α and related family members leading to activation of NF-κB; effects of reactive oxygen species (ROS) on signaling; endothelial adaptations to flow including G protein-coupled receptor (GPCR) and integrin-related signaling; activation of endothelial and other cells by modified lipoproteins; purinergic signaling; control of leukocyte adhesion to endothelium, migration, and further activation; foam cell formation; and macrophage and vascular smooth muscle cell signaling related to proliferation, efferocytosis, and apoptosis. This review is intended primarily as an introduction to these key signaling pathways. They have become the focus of modern atherosclerosis research and will undoubtedly provide a rich resource for future innovation toward intervention and prevention of the number one cause of death in the modern world.
Collapse
Affiliation(s)
- Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
31
|
Rios FJO, Ferracini M, Pecenin M, Koga MM, Wang Y, Ketelhuth DFJ, Jancar S. Uptake of oxLDL and IL-10 production by macrophages requires PAFR and CD36 recruitment into the same lipid rafts. PLoS One 2013; 8:e76893. [PMID: 24130805 PMCID: PMC3793910 DOI: 10.1371/journal.pone.0076893] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 08/28/2013] [Indexed: 01/24/2023] Open
Abstract
Macrophage interaction with oxidized low-density lipoprotein (oxLDL) leads to its differentiation into foam cells and cytokine production, contributing to atherosclerosis development. In a previous study, we showed that CD36 and the receptor for platelet-activating factor (PAFR) are required for oxLDL to activate gene transcription for cytokines and CD36. Here, we investigated the localization and physical interaction of CD36 and PAFR in macrophages stimulated with oxLDL. We found that blocking CD36 or PAFR decreases oxLDL uptake and IL-10 production. OxLDL induces IL-10 mRNA expression only in HEK293T expressing both receptors (PAFR and CD36). OxLDL does not induce IL-12 production. The lipid rafts disruption by treatment with βCD reduces the oxLDL uptake and IL-10 production. OxLDL induces co-immunoprecipitation of PAFR and CD36 with the constitutive raft protein flotillin-1, and colocalization with the lipid raft-marker GM1-ganglioside. Finally, we found colocalization of PAFR and CD36 in macrophages from human atherosclerotic plaques. Our results show that oxLDL induces the recruitment of PAFR and CD36 into the same lipid rafts, which is important for oxLDL uptake and IL-10 production. This study provided new insights into how oxLDL interact with macrophages and contributing to atherosclerosis development.
Collapse
Affiliation(s)
- Francisco J O Rios
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil ; BHF-Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
32
|
Sniderman AD, Islam S, Yusuf S, McQueen MJ. Is the superiority of apoB over non-HDL-C as a marker of cardiovascular risk in the INTERHEART study due to confounding by related variables? J Clin Lipidol 2013; 7:626-31. [PMID: 24314360 DOI: 10.1016/j.jacl.2013.08.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 08/20/2013] [Accepted: 08/27/2013] [Indexed: 12/14/2022]
Abstract
BACKGROUND Patients with increased numbers of cholesterol-depleted apolipoprotein B (apoB) particles frequently have multiple other abnormalities, which might confound the comparison of apoB and non-high-density-lipoprotein-cholesterol (non-HDL-C) as markers of cardiovascular risk. OBJECTIVE We wanted to determine whether the superiority of apoB over non-HDL-C as a marker of cardiovascular risk in the INTERHEART study is due to such variables that act as confounders of the primary comparison. METHODS To test for confounding, cases and controls were first separated into 3 groups on the basis of the percentile levels within the study of non-HDL-C and apoB with discordance defined as a difference of 5 percentile points. Logistic regression was used to compute odds ratio of myocardial infarction (as an outcome) for different categories, assuming concordance as reference adjusted for other confounders. RESULTS Plasma triglyceride and non-HDL-C levels were highest in the discordant group with lowest risk and lowest in the discordant group with highest risk, whereas apoB was highest in the discordant group with the highest risk and lowest in the discordant group with the lowest group. Moreover, no significant change was found in the odds ratio for either discordant group when adjusted for the effect of any of the variables examined, evidence that none confounded the primary comparison. CONCLUSION Factors such as hypertriglyceridemia do not confound the comparison of apoB and non-HDL-C, further evidence that apoB is superior to non-HDL-C as a marker of the importance of the apoB atherogenic lipoproteins in cardiovascular risk.
Collapse
Affiliation(s)
- Allan D Sniderman
- Mike Rosenbloom Laboratory for Cardiovascular Research, McGill University Health Centre, Royal Victoria Hospital, Room H7.22, 687 Pine Avenue West, Montreal, Québec, Canada H3A 1A1.
| | | | | | | |
Collapse
|
33
|
Witztum JL, Lichtman AH. The influence of innate and adaptive immune responses on atherosclerosis. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2013; 9:73-102. [PMID: 23937439 DOI: 10.1146/annurev-pathol-020712-163936] [Citation(s) in RCA: 220] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Both the chronic development of atherosclerotic lesions and the acute changes in lesion phenotype that lead to clinical cardiovascular events are significantly influenced by the innate and adaptive immune responses to lipoprotein deposition and oxidation in the arterial wall. The rapid pace of discovery of mechanisms of immunologic recognition, effector functions, and regulation has significantly influenced the study of atherosclerosis, and our new knowledge is beginning to affect how we treat this ubiquitous disease. In this review, we discuss recent advances in our understanding of how innate and adaptive immunity contribute to atherosclerosis, as well as therapeutic opportunities that arise from this knowledge.
Collapse
Affiliation(s)
- Joseph L Witztum
- Department of Medicine, University of California, San Diego, La Jolla, California 92093;
| | | |
Collapse
|
34
|
Obesity Modulates the Immune Response to Oxidized LDL in Hypertensive Patients. Cell Biochem Biophys 2013; 67:1451-60. [DOI: 10.1007/s12013-013-9585-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
35
|
Sniderman A, Kwiterovich PO. Update on the detection and treatment of atherogenic low-density lipoproteins. Curr Opin Endocrinol Diabetes Obes 2013; 20:140-7. [PMID: 23422241 DOI: 10.1097/med.0b013e32835ed9cb] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE OF REVIEW To explain why epidemiological studies have reached such diverse views as to whether apolipoprotein B (apoB) and/or low-density lipoprotein particle number (LDL-P) are more accurate markers of the risk of cardiovascular disease than LDL-C or non-high-density lipoprotein cholesterol (HDL-C) and to review the treatment options to lower LDL. RECENT FINDINGS The Emerging Risk Factor Collaboration, a large prospective participant level analysis, a meta-analysis of statin clinical trials, and the Heart Protection Study have each reported that apoB does not add significantly to the cholesterol markers as indices of cardiovascular risk. By contrast, a meta-analysis of published prospective studies demonstrated that non-HDL-C was superior to LDL-C, and apoB was superior to non-HDL-C. As well, three studies using discordance analysis each demonstrated that apoB and LDL-P were superior to the cholesterol markers. Two approaches to resolve these differences are brought to bear in this article: first, which results are credible and second, how does taking the known differences in LDL composition into account, help resolve them. The best identification of individuals at risk of coronary artery disease or with coronary artery disease allows the most efficacious treatment of elevated LDL-P and will permit a more extensive use of some of the more novel LDL-lowering agents. SUMMARY Much of the controversy vanishes once the physiologically driven differences in the composition of the apoB lipoprotein particles are taken into account, illustrating that epidemiology, not directed by physiology, is like shooting without aiming.
Collapse
Affiliation(s)
- Allan Sniderman
- Division of Cardiology, McGill University Health Centre, Montreal, Quebec, Canada
| | | |
Collapse
|
36
|
Rios FJ, Koga MM, Pecenin M, Ferracini M, Gidlund M, Jancar S. Oxidized LDL induces alternative macrophage phenotype through activation of CD36 and PAFR. Mediators Inflamm 2013; 2013:198193. [PMID: 24062612 PMCID: PMC3767076 DOI: 10.1155/2013/198193] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Accepted: 07/22/2013] [Indexed: 12/14/2022] Open
Abstract
OxLDL is recognized by macrophage scavenger receptors, including CD36; we have recently found that Platelet-Activating Factor Receptor (PAFR) is also involved. Since PAFR in macrophages is associated with suppressor function, we examined the effect of oxLDL on macrophage phenotype. It was found that the presence of oxLDL during macrophage differentiation induced high mRNA levels to IL-10, mannose receptor, PPAR γ and arginase-1 and low levels of IL-12 and iNOS. When human THP-1 macrophages were pre-treated with oxLDL then stimulated with LPS, the production of IL-10 and TGF- β significantly increased, whereas that of IL-6 and IL-8 decreased. In murine TG-elicited macrophages, this protocol significantly reduced NO, iNOS and COX2 expression. Thus, oxLDL induced macrophage differentiation and activation towards the alternatively activated M2-phenotype. In murine macrophages, oxLDL induced TGF- β , arginase-1 and IL-10 mRNA expression, which were significantly reduced by pre-treatment with PAFR antagonists (WEB and CV) or with antibodies to CD36. The mRNA expression of IL-12, RANTES and CXCL2 were not affected. We showed that this profile of macrophage activation is dependent on the engagement of both CD36 and PAFR. We conclude that oxLDL induces alternative macrophage activation by mechanisms involving CD36 and PAFR.
Collapse
Affiliation(s)
- Francisco J Rios
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Avenida Professor Lineu Prestes 1730, ICB IV-Sala 140/146, 05508-900 Sao Paulo, SP, Brazil.
| | | | | | | | | | | |
Collapse
|
37
|
Ortea I, Roschitzki B, Ovalles JG, Longo JL, de la Torre I, González I, Gómez-Reino JJ, González A. Discovery of serum proteomic biomarkers for prediction of response to infliximab (a monoclonal anti-TNF antibody) treatment in rheumatoid arthritis: An exploratory analysis. J Proteomics 2012; 77:372-82. [DOI: 10.1016/j.jprot.2012.09.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 09/07/2012] [Accepted: 09/11/2012] [Indexed: 12/22/2022]
|
38
|
Zhang L, Ovchinnikova O, Jönsson A, Lundberg AM, Berg M, Hansson GK, Ketelhuth DF. The tryptophan metabolite 3-hydroxyanthranilic acid lowers plasma lipids and decreases atherosclerosis in hypercholesterolaemic mice. Eur Heart J 2012; 33:2025-34. [DOI: 10.1093/eurheartj/ehs175] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
39
|
Rios FJO, Koga MM, Ferracini M, Jancar S. Co-stimulation of PAFR and CD36 is required for oxLDL-induced human macrophages activation. PLoS One 2012; 7:e36632. [PMID: 22570732 PMCID: PMC3343035 DOI: 10.1371/journal.pone.0036632] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 04/04/2012] [Indexed: 11/18/2022] Open
Abstract
The oxidative process of LDL particles generates molecules which are structurally similar to platelet-activating factor (PAF), and some effects of oxidized LDL (oxLDL) have been shown to be dependent on PAF receptor (PAFR) activation. In a previous study, we showed that PAFR is required for upregulation of CD36 and oxLDL uptake. In the present study we analyzed the molecular mechanisms activated by oxLDL in human macrophages and the contribution of PAFR to this response. Human adherent monocytes/macrophages were stimulated with oxLDL. Uptake of oxLDL and CD36 expression were determined by flow cytometry; MAP kinases and Akt phosphorylation by Western blot; IL-8 and MCP-1 concentration by ELISA and mRNA expression by real-time PCR. To investigate the participation of the PI3K/Akt pathway, Gαi-coupled protein or PAFR, macrophages were treated with LY294002, pertussis toxin or with the PAFR antagonists WEB2170 and CV3988, respectively before addition of oxLDL. It was found that the addition of oxLDL to human monocytes/macrophages activates the PI3K/Akt pathway which in turn activates the MAPK (p38 and JNK). Phosphorylation of Akt requires the engagement of PAFR and a Gαi-coupled protein. The upregulation of CD36 protein and the uptake of oxLDL as well as the IL-8 production are dependent on PI3K/Akt pathway activation. The increased CD36 protein expression is dependent on PAFR and Gαi-coupled protein. Transfection studies using HEK 293t cells showed that oxLDL uptake occurs with either PAFR or CD36, but IL-8 production requires the co-transfection of both PAFR and CD36. These findings show that PAFR has a pivotal role in macrophages response to oxLDL and suggest that pharmacological intervention at the level of PAFR activation might be beneficial in atherosclerosis.
Collapse
Affiliation(s)
- Francisco J O Rios
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, Sao Paulo, Brazil.
| | | | | | | |
Collapse
|