1
|
Deroissart J, Binder CJ, Porsch F. Role of Antibodies and Their Specificities in Atherosclerotic Cardiovascular Disease. Arterioscler Thromb Vasc Biol 2024; 44:2154-2168. [PMID: 39114917 DOI: 10.1161/atvbaha.124.319843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Atherosclerosis is a lipid-driven chronic inflammatory disease that is modulated by innate and adaptive immunity including humoral immunity. Importantly, antibody alterations achieved by genetic means or active and passive immunization strategies in preclinical studies can improve or aggravate atherosclerosis. Additionally, a wide range of epidemiological data demonstrate not only an association between the total levels of different antibody isotypes but also levels of antibodies targeting specific antigens with atherosclerotic cardiovascular disease. Here, we discuss the potential role of atherogenic dyslipidemia on the antibody repertoire and review potential antibody-mediated effector mechanisms involved in atherosclerosis development highlighting the major atherosclerosis-associated antigens that trigger antibody responses.
Collapse
Affiliation(s)
- Justine Deroissart
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - Florentina Porsch
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| |
Collapse
|
2
|
Jackson ML, Bond AR, Ascione R, Johnson JL, George SJ. FGL2/FcγRIIB Signalling Mediates Arterial Shear Stress-Mediated Endothelial Cell Apoptosis: Implications for Coronary Artery Bypass Vein Graft Pathogenesis. Int J Mol Sci 2024; 25:7638. [PMID: 39062880 PMCID: PMC11277082 DOI: 10.3390/ijms25147638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
The sudden exposure of venous endothelial cells (vECs) to arterial fluid shear stress (FSS) is thought to be a major contributor to coronary artery bypass vein graft failure (VGF). However, the effects of arterial FSS on the vEC secretome are poorly characterised. We propose that analysis of the vEC secretome may reveal potential therapeutic approaches to suppress VGF. Human umbilical vein endothelial cells (HUVECs) pre-conditioned to venous FSS (18 h; 1.5 dynes/cm2) were exposed to venous or arterial FSS (15 dynes/cm2) for 24 h. Tandem Mass Tagging proteomic analysis of the vEC secretome identified significantly increased fibroleukin (FGL2) in conditioned media from HUVECs exposed to arterial FSS. This increase was validated by Western blotting. Application of the NFκB inhibitor BAY 11-7085 (1 µM) following pre-conditioning reduced FGL2 release from vECs exposed to arterial FSS. Exposure of vECs to arterial FSS increased apoptosis, measured by active cleaved caspase-3 (CC3) immunocytochemistry, which was likewise elevated in HUVECs treated with recombinant FGL2 (20 ng/mL) for 24 h under static conditions. To determine the mechanism of FGL2-induced apoptosis, HUVECs were pre-treated with a blocking antibody to FcγRIIB, a receptor FGL2 is proposed to interact with, which reduced CC3 levels. In conclusion, our findings indicate that the exposure of vECs to arterial FSS results in increased release of FGL2 via NFκB signalling, which promotes endothelial apoptosis via FcγRIIB signalling. Therefore, the inhibition of FGL2/FcγRIIB signalling may provide a novel approach to reduce arterial FSS-induced vEC apoptosis in vein grafts and suppress VGF.
Collapse
Affiliation(s)
| | | | | | | | - Sarah J. George
- Translational Health Sciences, Bristol Medical School, Faculty of Health and Life Sciences, University of Bristol, Bristol BS2 8HW, UK; (M.L.J.); (A.R.B.); (R.A.); (J.L.J.)
| |
Collapse
|
3
|
Raposo-Gutiérrez I, Rodríguez-Ronchel A, Ramiro AR. Atherosclerosis antigens as targets for immunotherapy. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1129-1147. [PMID: 39196152 DOI: 10.1038/s44161-023-00376-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 10/18/2023] [Indexed: 08/29/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arteries that can lead to thrombosis, infarction and stroke, underlying the first cause of mortality worldwide. Adaptive immunity plays critical roles in atherosclerosis, and numerous studies have ascribed both atheroprotective and atherogenic functions to specific subsets of T and B cells. However, less is known on how antigen specificity determines the protective or adverse outcome of such adaptive responses. Understanding antigen triggers in atherosclerosis is crucial to delve deeper into mechanisms of disease initiation and progression and to implement specific immunotherapeutic approaches, including vaccination strategies. Here we review the role of adaptive immunity in atherosclerosis and the insights that single-cell technology has provided into the function of distinct immune cell subsets. We outline the most relevant atherosclerosis antigens and antibodies reported to date and examine their immunotherapeutic potential. Finally, we review the most promising vaccination-based clinical trials targeting the adaptive immune system.
Collapse
Affiliation(s)
- Irene Raposo-Gutiérrez
- B Lymphocyte Lab, Novel Mechanisms of Atherosclerosis Program, Spanish National Center for Cardiovascular Research, Madrid, Spain
| | - Ana Rodríguez-Ronchel
- B Lymphocyte Lab, Novel Mechanisms of Atherosclerosis Program, Spanish National Center for Cardiovascular Research, Madrid, Spain
| | - Almudena R Ramiro
- B Lymphocyte Lab, Novel Mechanisms of Atherosclerosis Program, Spanish National Center for Cardiovascular Research, Madrid, Spain.
| |
Collapse
|
4
|
López-Sanz L, Bernal S, Jiménez-Castilla L, Pardines M, Hernández-García A, Blanco-Colio L, Martín-Ventura JL, Gómez Guerrero C. The presence of activating IgG Fc receptors in macrophages aggravates the development of experimental abdominal aortic aneurysm. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2023; 35:185-194. [PMID: 36737385 DOI: 10.1016/j.arteri.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/16/2022] [Accepted: 12/22/2022] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Abdominal aortic aneurysm (AAA) is a multifactorial, degenerative disease characterized by progressive aortic dilation and chronic activation of inflammation, proteolytic activity, and oxidative stress in the aortic wall. The immune response triggered by antibodies against antigens present in the vascular wall participates in the formation and progression of AAA through mechanisms not completely understood. This work analyses the function of specific IgG receptors (FcγR), especially those expressed by monocytes/macrophages, in the development of experimental AAA. METHODS In the elastase-induced AAA model, the abdominal aortas from wildtype and FcγR deficient mice with/without macrophage adoptive transfer were analysed by histology and quantitative PCR. In vitro, mouse macrophages were transfected with RNA interference of FcγRIV/CD16.2 or treated with Syk kinase inhibitor before stimulation with IgG immune complexes. RESULTS Macrophage adoptive transfer in FcγR deficient mice increased the susceptibility to AAA development. Mice receiving macrophages with functional FcγR exhibited higher aortic diameter increase, higher content of macrophages and B lymphocytes, and upregulated expression of chemokine CCL2, cytokines (TNF-α and IL-17), metalloproteinase MMP2, prooxidant enzyme NADPH oxidase-2, and the isoforms FcγRIII/CD16 and FcγRIV/CD16.2. In vitro, both FcγRIV/CD16.2 gene silencing and Syk inhibition reduced cytokines and reactive oxygen species production induced by immune complexes in macrophages. CONCLUSIONS Activation of macrophage FcγR contributes to AAA development by inducing mediators of inflammation, proteolysis, and oxidative stress. Modulation of FcγR or effector molecules may represent a potential target for AAA treatment.
Collapse
Affiliation(s)
- Laura López-Sanz
- Laboratorio de Patología Vascular y Renal, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD/UAM), Madrid, España; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Madrid, España
| | - Susana Bernal
- Laboratorio de Patología Vascular y Renal, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD/UAM), Madrid, España; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Madrid, España
| | - Luna Jiménez-Castilla
- Laboratorio de Patología Vascular y Renal, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD/UAM), Madrid, España; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Madrid, España
| | - Marisa Pardines
- Laboratorio de Patología Vascular y Renal, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD/UAM), Madrid, España
| | - Ana Hernández-García
- Laboratorio de Patología Vascular y Renal, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD/UAM), Madrid, España
| | - Luis Blanco-Colio
- Laboratorio de Patología Vascular y Renal, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD/UAM), Madrid, España; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, España
| | - José Luis Martín-Ventura
- Laboratorio de Patología Vascular y Renal, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD/UAM), Madrid, España; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, España
| | - Carmen Gómez Guerrero
- Laboratorio de Patología Vascular y Renal, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD/UAM), Madrid, España; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Madrid, España.
| |
Collapse
|
5
|
Porsch F, Mallat Z, Binder CJ. Humoral immunity in atherosclerosis and myocardial infarction: from B cells to antibodies. Cardiovasc Res 2021; 117:2544-2562. [PMID: 34450620 DOI: 10.1093/cvr/cvab285] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/30/2021] [Accepted: 08/24/2021] [Indexed: 02/06/2023] Open
Abstract
Immune mechanisms are critically involved in the pathogenesis of atherosclerosis and its clinical manifestations. Associations of specific antibody levels and defined B cell subsets with cardiovascular disease activity in humans as well as mounting evidence from preclinical models demonstrate a role of B cells and humoral immunity in atherosclerotic cardiovascular disease. These include all aspects of B cell immunity, the generation of antigen-specific antibodies, antigen presentation and co-stimulation of T cells, as well as production of cytokines. Through their impact on adaptive and innate immune responses and the regulation of many other immune cells, B cells mediate both protective and detrimental effects in cardiovascular disease. Several antigens derived from (oxidised) lipoproteins, the vascular wall and classical autoantigens have been identified. The unique antibody responses they trigger and their relationship with atherosclerotic cardiovascular disease are reviewed. In particular, we focus on the different effector functions of specific IgM, IgG, and IgE antibodies and the cellular responses they trigger and highlight potential strategies to target B cell functions for therapy.
Collapse
Affiliation(s)
- Florentina Porsch
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom.,INSERM U970, Paris Cardiovascular Research Centre, Paris, France.,Unversité Paris Descartes, Sorbonne Paris Cité, Paris France
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
6
|
Hutchinson MA, Park HS, Zanotti KJ, Alvarez-Gonzalez J, Zhang J, Zhang L, Telljohann R, Wang M, Lakatta EG, Gearhart PJ, Maul RW. Auto-Antibody Production During Experimental Atherosclerosis in ApoE-/- Mice. Front Immunol 2021; 12:695220. [PMID: 34305930 PMCID: PMC8299997 DOI: 10.3389/fimmu.2021.695220] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/21/2021] [Indexed: 12/22/2022] Open
Abstract
Current models stipulate that B cells and antibodies function during atherosclerosis in two distinct ways based on antibody isotype, where IgM is protective and IgG is inflammatory. To examine this model, we generated ApoE-/- Aid-/- mice, which are unable to produce IgG antibodies due to the absence of activation-induced deaminase (AID) but maintain high plasma cholesterol due to the absence of apolipoprotein E (APOE). We saw a dramatic decrease in plaque formation in ApoE-/- Aid-/- mice compared to ApoE-/- mice. Rigorous analysis of serum antibodies revealed both ApoE-/- and ApoE-/- Aid-/- mice had substantially elevated titers of IgM antibodies compared to C57BL/6J controls, suggesting a more complex dynamic than previously described. Analysis of antigen specificity demonstrated that ApoE-/- Aid-/- mice had elevated titers of antibodies specific to malondialdehyde-oxidized low density lipoprotein (MDA-oxLDL), which has been shown to block macrophage recruitment into plaques. Conversely, ApoE-/- mice showed low levels of MDA-oxLDL specificity, but had antibodies specific to numerous self-proteins. We provide evidence for a hierarchical order of antibody specificity, where elevated levels of MDA-oxLDL specific IgM antibodies inhibit plaque formation. If the level of MDA-oxLDL specific IgM is insufficient, self-reactive IgM and IgG antibodies are generated against debris within the arterial plaque, resulting in increased inflammation and further plaque expansion.
Collapse
Affiliation(s)
- Mark A. Hutchinson
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, MD, United States
| | - Han-Sol Park
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, MD, United States
| | - Kimberly J. Zanotti
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, MD, United States
| | - Juan Alvarez-Gonzalez
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, MD, United States
| | - Jing Zhang
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, United States
| | - Li Zhang
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, United States
| | - Richard Telljohann
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, United States
| | - Mingyi Wang
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, United States
| | - Edward G. Lakatta
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, United States
| | - Patricia J. Gearhart
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, MD, United States
| | - Robert W. Maul
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, MD, United States
| |
Collapse
|
7
|
Lopez‐Sanz L, Bernal S, Jimenez‐Castilla L, Prieto I, La Manna S, Gomez‐Lopez S, Blanco‐Colio LM, Egido J, Martin‐Ventura JL, Gomez‐Guerrero C. Fcγ receptor activation mediates vascular inflammation and abdominal aortic aneurysm development. Clin Transl Med 2021; 11:e463. [PMID: 34323424 PMCID: PMC8255062 DOI: 10.1002/ctm2.463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/24/2021] [Accepted: 05/30/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA), a degenerative vascular pathology characterized by permanent dilation of the aorta, is considered a chronic inflammatory disease involving innate/adaptive immunity. However, the functional role of antibody-dependent immune response against antigens present in the damaged vessel remains unresolved. We hypothesized that engagement of immunoglobulin G (IgG) Fc receptors (FcγR) by immune complexes (IC) in the aortic wall contributes to AAA development. We therefore evaluated FcγR expression in AAA lesions and analysed whether inhibition of FcγR signaling molecules (γ-chain and Syk kinase) influences AAA formation in mice. METHODS FcγR gene/protein expression was assessed in human and mouse AAA tissues. Experimental AAA was induced by aortic elastase perfusion in wild-type (WT) mice and γ-chain knockout (γKO) mice (devoid of activating FcγR) in combination with macrophage adoptive transfer or Syk inhibitor treatment. To verify the mechanisms of FcγR in vitro, vascular smooth muscle cells (VSMC) and macrophages were stimulated with IgG IC. RESULTS FcγR overexpression was detected in adventitia and media layers of human and mouse AAA. Elastase-perfused γKO mice exhibited a decrease in AAA incidence, aortic dilation, elastin degradation, and VSMC loss. This was associated with (1) reduced infiltrating leukocytes and immune deposits in AAA lesions, (2) inflammatory genes and metalloproteinases downregulation, (3) redox balance restoration, and (4) converse phenotype of anti-inflammatory macrophage M2 and contractile VSMC. Adoptive transfer of FcγR-expressing macrophages aggravated aneurysm in γKO mice. In vitro, FcγR deficiency attenuated inflammatory gene expression, oxidative stress, and phenotypic switch triggered by IC. Additionally, Syk inhibition prevented IC-mediated cell responses, reduced inflammation, and mitigated AAA formation. CONCLUSION Our findings provide insight into the role and mechanisms mediating IgG-FcγR-associated inflammation and aortic wall injury in AAA, which might represent therapeutic targets against AAA disease.
Collapse
MESH Headings
- Animals
- Antigen-Antibody Complex/adverse effects
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/prevention & control
- Disease Models, Animal
- Humans
- Immunoglobulin gamma-Chains/genetics
- Immunoglobulin gamma-Chains/metabolism
- Inflammation/metabolism
- Inflammation/pathology
- Macrophages/cytology
- Macrophages/immunology
- Macrophages/metabolism
- Male
- Matrix Metalloproteinases/genetics
- Matrix Metalloproteinases/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Niacinamide/analogs & derivatives
- Niacinamide/therapeutic use
- Oxidative Stress
- Pancreatic Elastase/adverse effects
- Pyrimidines/therapeutic use
- Receptors, IgG/genetics
- Receptors, IgG/metabolism
- Syk Kinase/antagonists & inhibitors
- Syk Kinase/metabolism
Collapse
Affiliation(s)
- Laura Lopez‐Sanz
- Renal, Vascular and Diabetes Research LabIIS‐Fundacion Jimenez Diaz (IIS‐FJD)MadridSpain
- Universidad Autonoma de Madrid (UAM)MadridSpain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)MadridSpain
| | - Susana Bernal
- Renal, Vascular and Diabetes Research LabIIS‐Fundacion Jimenez Diaz (IIS‐FJD)MadridSpain
- Universidad Autonoma de Madrid (UAM)MadridSpain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)MadridSpain
| | - Luna Jimenez‐Castilla
- Renal, Vascular and Diabetes Research LabIIS‐Fundacion Jimenez Diaz (IIS‐FJD)MadridSpain
- Universidad Autonoma de Madrid (UAM)MadridSpain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)MadridSpain
| | - Ignacio Prieto
- Renal, Vascular and Diabetes Research LabIIS‐Fundacion Jimenez Diaz (IIS‐FJD)MadridSpain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)MadridSpain
| | - Sara La Manna
- Renal, Vascular and Diabetes Research LabIIS‐Fundacion Jimenez Diaz (IIS‐FJD)MadridSpain
- Universidad Autonoma de Madrid (UAM)MadridSpain
| | | | - Luis Miguel Blanco‐Colio
- Renal, Vascular and Diabetes Research LabIIS‐Fundacion Jimenez Diaz (IIS‐FJD)MadridSpain
- Spanish Biomedical Research Centre in Cardiovascular Diseases (CIBERCV)MadridSpain
| | - Jesus Egido
- Renal, Vascular and Diabetes Research LabIIS‐Fundacion Jimenez Diaz (IIS‐FJD)MadridSpain
- Universidad Autonoma de Madrid (UAM)MadridSpain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)MadridSpain
| | - Jose Luis Martin‐Ventura
- Renal, Vascular and Diabetes Research LabIIS‐Fundacion Jimenez Diaz (IIS‐FJD)MadridSpain
- Universidad Autonoma de Madrid (UAM)MadridSpain
- Spanish Biomedical Research Centre in Cardiovascular Diseases (CIBERCV)MadridSpain
| | - Carmen Gomez‐Guerrero
- Renal, Vascular and Diabetes Research LabIIS‐Fundacion Jimenez Diaz (IIS‐FJD)MadridSpain
- Universidad Autonoma de Madrid (UAM)MadridSpain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)MadridSpain
| |
Collapse
|
8
|
Sun J, Shen Z, Niu X, Zhang Y, Zhang B, Zhang T, He K, Xu H, Liu S, Ho SSH, Li X, Cao J. Cytotoxicity and Potential Pathway to Vascular Smooth Muscle Cells Induced by PM 2.5 Emitted from Raw Coal Chunks and Clean Coal Combustion. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:14482-14493. [PMID: 33138382 DOI: 10.1021/acs.est.0c02236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Coal combustion emits a large amount of PM2.5 (particulate matters with aerodynamic diameters less than 2.5 μm) and causes adverse damages to the cardiovascular system. In this study, emissions from anthracite and bitumite were examined. Red mud (RM) acts as an additive and is mixed in coal briquettes with a content of 0-10% as a single variable to demonstrate the reduction in the PM2.5 emissions. Burnt in a regulated combustion chamber, the 10% RM-containing bitumite and anthracite briquettes showed 52.3 and 18.6% reduction in PM2.5, respectively, compared with their chunk coals. Lower cytotoxicity (in terms of oxidative stresses and inflammation factors) was observed for PM2.5 emitted from the RM-containing briquettes than those from non-RM briquettes, especially for the bitumite groups. Besides, the results of western blotting illustrated that the inhibition of NF-κB and MAPK was the potential pathway for the reduction of cytokine levels by the RM addition. The regression analyses further demonstrated that the reduction was attributed to the lower emissions of transition metals (i.e., Mn) and PAHs (i.e., acenaphthene). This pilot study provides solid evidence for the cytotoxicity to vascular smooth muscle cells induced by PM2.5 from coal combustion and potential solutions for reducing the emission of toxic pollutants from human health perspectives.
Collapse
Affiliation(s)
- Jian Sun
- Department of Environmental Science and Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Zhenxing Shen
- Department of Environmental Science and Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Xinyi Niu
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Shatin 999077, Hong Kong, China
| | - Yue Zhang
- Department of Environmental Science and Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Bin Zhang
- Department of Environmental Science and Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Tian Zhang
- Department of Environmental Science and Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Kun He
- Department of Environmental Science and Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Hongmei Xu
- Department of Environmental Science and Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Suixin Liu
- Key Lab of Aerosol Chemistry & Physics, Institute of Earth Environment, Chinese Academy of Sciences, Xi'an 710049, China
| | - Steven Sai Hang Ho
- Divison of Atmospheric Sciences, Desert Research Institute, Reno, Nevada 89512, United States
| | - Xuxiang Li
- School of Human Settlements and Civil Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Junji Cao
- Key Lab of Aerosol Chemistry & Physics, Institute of Earth Environment, Chinese Academy of Sciences, Xi'an 710049, China
| |
Collapse
|
9
|
Poria cocos polysaccharides attenuated ox-LDL-induced inflammation and oxidative stress via ERK activated Nrf2/HO-1 signaling pathway and inhibited foam cell formation in VSMCs. Int Immunopharmacol 2020; 80:106173. [DOI: 10.1016/j.intimp.2019.106173] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 12/24/2019] [Accepted: 12/28/2019] [Indexed: 12/31/2022]
|
10
|
Yu D, Wang T, Huang J, Fang X, Fan H, Yi G, Liu Q, Zhang Y, Zeng X, Liu Q. MicroRNA‐9 overexpression suppresses vulnerable atherosclerotic plaque and enhances vascular remodeling through negative regulation of the p38MAPK pathway via OLR1 in acute coronary syndrome. J Cell Biochem 2019; 121:49-62. [DOI: 10.1002/jcb.27830] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/14/2018] [Indexed: 12/16/2022]
Affiliation(s)
- Dao‐Rui Yu
- Department of Pharmacology School of Basic Medicine and Life Science, Hainan Medical University Haikou China
| | - Tao Wang
- Department of nursing humanities, International Nursing School, Hainan Medical University
| | - Jing Huang
- Department of Pharmacology School of Basic Medicine and Life Science, Hainan Medical University Haikou China
| | - Xing‐Yue Fang
- Department of Pharmacology School of Basic Medicine and Life Science, Hainan Medical University Haikou China
| | - Hao‐Fei Fan
- Department of Pharmacology School of Basic Medicine and Life Science, Hainan Medical University Haikou China
| | - Guo‐Hui Yi
- Instrument testing center, Public Research Laboratory, Hainan Medical University
| | - Qiang Liu
- Department of Pharmacology School of Basic Medicine and Life Science, Hainan Medical University Haikou China
| | - Yu Zhang
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University
| | - Xiang‐Zhou Zeng
- Department of Pharmacology School of Basic Medicine and Life Science, Hainan Medical University Haikou China
| | - Qi‐Bing Liu
- Department of Pharmacology School of Basic Medicine and Life Science, Hainan Medical University Haikou China
| |
Collapse
|
11
|
Bagchi-Chakraborty J, Francis A, Bray T, Masters L, Tsiantoulas D, Nus M, Harrison J, Broekhuizen M, Leggat J, Clatworthy MR, Espéli M, Smith KG, Binder CJ, Mallat Z, Sage AP. B Cell Fcγ Receptor IIb Modulates Atherosclerosis in Male and Female Mice by Controlling Adaptive Germinal Center and Innate B-1-Cell Responses. Arterioscler Thromb Vasc Biol 2019; 39:1379-1389. [PMID: 31092015 PMCID: PMC6636804 DOI: 10.1161/atvbaha.118.312272] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/22/2019] [Indexed: 02/02/2023]
Abstract
Objective- Investigate the impact of modulating B cell FcγRIIb (Fcγ receptor IIb) expression on atherosclerosis. Approach and Results- Western diet-induced atherosclerosis was assessed in Ldlr-/- or Apoe-/- mice with B cell-specific overexpression of FcγRIIb or with an FcγRIIb promoter mutation that alters FcγRIIb expression in germinal center (GC) B cells. In males, overexpression of FcγRIIb on B cells severely reduced activated, class switched B cell responses, as indicated by reductions in GC B cells, plasma cells, and serum IgG but not IgM antibodies. Male mice overexpressing FcγRIIb developed less atherosclerosis, suggesting a pathogenic role for GC B cell IgG responses. In support of this hypothesis, male mice with a promoter polymorphism-driven reduction in FcγRIIb on GC B cells but not plasma cells have a converse phenotype of enhanced GC responses and IgG2c antibodies and enhanced atherosclerosis. IgG2c significantly enhanced TNF (tumor necrosis factor) secretion by CD11b+ CD11c+ cells expressing the high-affinity receptor FcγRIV. In females, overexpression of FcγRIIb on B cells not only reduced GC B cell responses but also substantially reduced B-1 cells and IgM antibodies, which translated into acceleration of atherosclerosis. Promoter-driven reduction in FcγRIIb did not alter GC B cell responses in females and, therefore, had no impact on atherosclerosis. Conclusions- B cell FcγRIIb differentially alters proatherogenic adaptive GC B cell and atheroprotective innate B-1 responses in male and female mice fed a western diet. Our results highlight the importance of a better understanding and ability to selectively target B cell responses in future immunotherapeutic approaches against human cardiovascular disease. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Jayashree Bagchi-Chakraborty
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Anna Francis
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Toni Bray
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Leanne Masters
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Dimitrios Tsiantoulas
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Meritxell Nus
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - James Harrison
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Michelle Broekhuizen
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Jennifer Leggat
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| | - Menna R. Clatworthy
- Division of Immunology (M.R.C., K.G.C.S., Z.M.), Department of Medicine, University of Cambridge, United Kingdom
| | - Marion Espéli
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Paris, France (M.E.)
| | - Kenneth G.C. Smith
- Division of Immunology (M.R.C., K.G.C.S., Z.M.), Department of Medicine, University of Cambridge, United Kingdom
| | - Christoph J. Binder
- Department of Laboratory Medicine, Medical University of Vienna, Austria (C.J.B.)
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (C.J.B.)
| | - Ziad Mallat
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
- Division of Immunology (M.R.C., K.G.C.S., Z.M.), Department of Medicine, University of Cambridge, United Kingdom
- Institut National de la Santé et de la Recherche Médicale, Universite Paris-Descartes, Paris Cardiovascular Research Center, and Université Paris-Descartes, France (Z.M.)
| | - Andrew P. Sage
- From the Division of Cardiovascular Medicine (J.B.-C., A.F., T.B., L.M., D.T., M.N., J.H., M.B., J.L., Z.M., A.P.S.), Department of Medicine, University of Cambridge, United Kingdom
| |
Collapse
|
12
|
Marvin J, Rhoads JP, Major AS. FcγRIIb on CD11c + cells modulates serum cholesterol and triglyceride levels and differentially affects atherosclerosis in male and female Ldlr -/- mice. Atherosclerosis 2019; 285:108-119. [PMID: 31051414 DOI: 10.1016/j.atherosclerosis.2019.04.221] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/08/2019] [Accepted: 04/12/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Circulating levels of oxidized lipoprotein (oxLDL) correlate with myocardial infarction risk and atherosclerosis severity. Our previous study demonstrates that oxLDL immune complexes (oxLDL-ICs) can signal through FcγRs on bone marrow-derived dendritic cells (BMDCs) and enhance their activation and inflammatory cytokine secretion. While global FcγR-/- studies have shown that activating FcγRs are proatherogenic, the role of the inhibitory FcγRIIb is unclear. We sought to determine the role of DC-specific FcγRIIb in atherosclerosis. METHODS Bone marrow chimeras were generated by rescuing lethally irradiated Ldlr-/- mice with hematopoietic cells from littermate CD11c-Cre+ or CD11c-Cre-Fcgr2bfl/fl donors. Four weeks following transplant, recipients were placed on a Western diet for eight weeks. Various tissues and organs were analyzed for differences in inflammation. RESULTS Quantitation of atherosclerosis in the proximal aorta demonstrated a 58% increase in female CD11c-Cre+Fcgr2bfl/fl recipients, but a surprising 44% decrease in male recipients. Hepatic cholesterol and triglycerides were increased in female CD11c-Cre+Fcgr2bfl/fl recipients. This was associated with an increase in CD36 and MHC Class II expression on hepatic CD11c+CD11b+ DCs in female livers. In contrast, male CD11c-Cre+Fcgr2bfl/fl recipients had decreased hepatic lipids with a corresponding decrease in CD36 and MHC Class II expression on CD11c+ cells. Interestingly, both sexes of CD11c-Cre+Fcgr2bfl/fl recipients had significant decreases in serum cholesterol and TGs with corresponding decreases in liver Fasn transcripts. CONCLUSIONS The absence of FcγRIIb expression on CD11c+ cells results in sex-dependent alteration in liver inflammation influencing atherogenesis and sex-independent modulation of serum cholesterol and TGs.
Collapse
Affiliation(s)
- Jennifer Marvin
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt Medical Center, Nashville, TN, 37232, USA
| | - Jillian P Rhoads
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt Medical Center, Nashville, TN, 37232, USA
| | - Amy S Major
- Tennessee Valley Healthcare System, U.S. Department of Veterans Affairs, Nashville, TN, 37212, USA; Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt Medical Center, Nashville, TN, 37232, USA.
| |
Collapse
|
13
|
Fc Gamma Receptor IIb Expressed in Hepatocytes Promotes Lipid Accumulation and Gluconeogenesis. Int J Mol Sci 2018; 19:ijms19102932. [PMID: 30261661 PMCID: PMC6213401 DOI: 10.3390/ijms19102932] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/03/2018] [Accepted: 09/08/2018] [Indexed: 01/01/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is characterized by ectopic lipid accumulation in the liver, usually combined with hepatic insulin resistance. Fc-gamma receptor-IIb (FcγRIIb) and its ligand are reported to be associated with obesity and type 2 diabetes mellitus (T2DM). As knowledge about FcγRIIb in the literature is mostly generated from studies on skeletal muscle tissue, the expression and function of FcγRIIb in the liver and hepatocytes are largely unknown. In this study, we identified the expression of FcγRIIb in primary cultured mouse hepatocytes: FcγRIIb was upregulated in response to oleic acid (OA) in a dose dependent manner. FcγRIIb knockdown using shRNA suppressed the lipid and triglyceride accumulation, and mRNA expression of ACC1, FASn, CD36, MTTP, and ApoB in OA-treated HepG2 cells. FcγRIIb deficiency mice fed with high fat diet (HFD) had significantly lower liver weight and liver to body weight ratio, as well as less triglyceride accumulation in the livers. In glycometabolism, FcγRIIb hindered insulin-induced phosphorylation of AKT and FOXO1, and in turn upregulated G6Pase and PEPCK mRNA expression, suggesting that FcγRIIb promotes gluconeogenesis by suppressing the AKT/FOXO1/G6Pase/PEPCK pathway in hepatocytes. This study reveals a novel role for FcγRIIb in regulating lipid metabolism and glycometabolism, and provides a new therapeutic target to improve NAFLD.
Collapse
|
14
|
Sage AP, Nus M, Bagchi Chakraborty J, Tsiantoulas D, Newland SA, Finigan AJ, Masters L, Binder CJ, Mallat Z. X-Box Binding Protein-1 Dependent Plasma Cell Responses Limit the Development of Atherosclerosis. Circ Res 2017; 121:270-281. [DOI: 10.1161/circresaha.117.310884] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 06/14/2017] [Accepted: 06/15/2017] [Indexed: 02/06/2023]
Abstract
Rationale:
Diverse B cell responses and functions may be involved in atherosclerosis. Protective antibody responses, such as those against oxidized lipid epitopes, are thought to mainly derive from T cell-independent innate B cell subsets. In contrast, both pathogenic and protective roles have been associated with T cell-dependent antibodies, and their importance in both humans and mouse models is still unclear.
Objective:
To specifically target antibody production by plasma cells and determine the impact on atherosclerotic plaque development in mice with and without CD4+ T cells.
Methods and Results:
We combined a model of specific antibody deficiency, B cell-specific CD79a-
Cre
x XBP1 (X-box binding protein-1) floxed mice (XBP1-conditional knockout), with antibody-mediated depletion of CD4+ T cells. Ldlr knockout mice transplanted with XBP1-conditional knockout (or wild-type control littermate) bone marrow were fed western diet for 8 weeks with or without anti-CD4 depletion. All groups had similar levels of serum cholesterol. In Ldlr/XBP1-conditional knockout mice, serum levels of IgG, IgE, and IgM were significantly attenuated, and local antibody deposition in atherosclerotic plaque was absent. Antibody deficiency significantly accelerated atherosclerosis at both the aortic root and aortic arch. T cell and monocyte responses were not modulated, but necrotic core size was greater, even when adjusting for plaque size, and collagen deposition significantly lower. Anti-CD4 depletion in Ldlr/wild-type mice led to a decrease of serum IgG1 and IgG2c but not IgG3, as well as decreased IgM, associated with increased atherosclerosis and necrotic cores, and a decrease in plaque collagen. The combination of antibody deficiency and anti-CD4 depletion has no additive effects on aortic root atherosclerosis.
Conclusions:
The endogenous T cell-dependent humoral response can be protective. This has important implications for novel vaccine strategies for atherosclerosis and in understanding the impacts of immunotherapies used in patients at high risk for cardiovascular disease.
Collapse
Affiliation(s)
- Andrew P. Sage
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Meritxell Nus
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Jayashree Bagchi Chakraborty
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Dimitrios Tsiantoulas
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Stephen A. Newland
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Alison J. Finigan
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Leanne Masters
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Christoph J. Binder
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Ziad Mallat
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| |
Collapse
|
15
|
Shi Y, Yang CQ, Wang SW, Li W, Li J, Wang SM. Characterization of Fc gamma receptor IIb expression within abdominal aortic aneurysm. Biochem Biophys Res Commun 2017; 485:295-300. [DOI: 10.1016/j.bbrc.2017.02.088] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 02/17/2017] [Indexed: 11/28/2022]
|
16
|
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: 38] [Impact Index Per Article: 5.4] [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
|
17
|
Sage AP, Mallat Z. Readapting the adaptive immune response - therapeutic strategies for atherosclerosis. Br J Pharmacol 2017; 174:3926-3939. [PMID: 28052311 DOI: 10.1111/bph.13700] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/13/2016] [Accepted: 12/20/2016] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases remain a major global health issue, with the development of atherosclerosis as a major underlying cause. Our treatment of cardiovascular disease has improved greatly over the past three decades, but much remains to be done reduce disease burden. Current priorities include reducing atherosclerosis advancement to clinically significant stages and preventing plaque rupture or erosion. Inflammation and involvement of the adaptive immune system influences all these aspects and therefore is one focus for future therapeutic development. The atherosclerotic vascular wall is now recognized to be invaded from both sides (arterial lumen and adventitia), for better or worse, by the adaptive immune system. Atherosclerosis is also affected at several stages by adaptive immune responses, overall providing many opportunities to target these responses and to reduce disease progression. Protective influences that may be defective in diseased individuals include humoral responses to modified LDL and regulatory T cell responses. There are many strategies in development to boost these pathways in humans, including vaccine-based therapies. The effects of various existing adaptive immune targeting therapies, such as blocking critical co-stimulatory pathways or B cell depletion, on cardiovascular disease are beginning to emerge with important consequences for both autoimmune disease patients and the potential for wider use of such therapies. Entering the translation phase for adaptive immune targeting therapies is an exciting and promising prospect. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- Andrew P Sage
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK.,INSERM U970, Paris Cardiovascular Research Center, Paris, France, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
18
|
Rhoads JP, Lukens JR, Wilhelm AJ, Moore JL, Mendez-Fernandez Y, Kanneganti TD, Major AS. Oxidized Low-Density Lipoprotein Immune Complex Priming of the Nlrp3 Inflammasome Involves TLR and FcγR Cooperation and Is Dependent on CARD9. THE JOURNAL OF IMMUNOLOGY 2017; 198:2105-2114. [PMID: 28130494 DOI: 10.4049/jimmunol.1601563] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 12/20/2016] [Indexed: 11/19/2022]
Abstract
Oxidized low-density lipoprotein (oxLDL) is known to activate inflammatory responses in a variety of cells, especially macrophages and dendritic cells. Interestingly, much of the oxLDL in circulation is complexed to Abs, and these resulting immune complexes (ICs) are a prominent feature of chronic inflammatory disease, such as atherosclerosis, type-2 diabetes, systemic lupus erythematosus, and rheumatoid arthritis. Levels of oxLDL ICs often correlate with disease severity, and studies demonstrated that oxLDL ICs elicit potent inflammatory responses in macrophages. In this article, we show that bone marrow-derived dendritic cells (BMDCs) incubated with oxLDL ICs for 24 h secrete significantly more IL-1β compared with BMDCs treated with free oxLDL, whereas there was no difference in levels of TNF-α or IL-6. Treatment of BMDCs with oxLDL ICs increased expression of inflammasome-related genes Il1a, Il1b, and Nlrp3, and pretreatment with a caspase 1 inhibitor decreased IL-1β secretion in response to oxLDL ICs. This inflammasome priming was due to oxLDL IC signaling via multiple receptors, because inhibition of CD36, TLR4, and FcγR significantly decreased IL-1β secretion in response to oxLDL ICs. Signaling through these receptors converged on the adaptor protein CARD9, a component of the CARD9-Bcl10-MALT1 signalosome complex involved in NF-κB translocation. Finally, oxLDL IC-mediated IL-1β production resulted in increased Th17 polarization and cytokine secretion. Collectively, these data demonstrate that oxLDL ICs induce inflammasome activation through a separate and more robust mechanism than oxLDL alone and that these ICs may be immunomodulatory in chronic disease and not just biomarkers of severity.
Collapse
Affiliation(s)
- Jillian P Rhoads
- Tennessee Valley Healthcare System, U.S. Department of Veterans Affairs, Nashville, TN 37212.,Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232
| | - John R Lukens
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA 22908
| | - Ashley J Wilhelm
- Division of Rheumatology, Department of Medicine, Vanderbilt Medical Center, Nashville, TN 37232
| | - Jared L Moore
- Tennessee Valley Healthcare System, U.S. Department of Veterans Affairs, Nashville, TN 37212.,Division of Rheumatology, Department of Medicine, Vanderbilt Medical Center, Nashville, TN 37232
| | | | | | - Amy S Major
- Tennessee Valley Healthcare System, U.S. Department of Veterans Affairs, Nashville, TN 37212; .,Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232.,Division of Rheumatology, Department of Medicine, Vanderbilt Medical Center, Nashville, TN 37232
| |
Collapse
|
19
|
Jackson SW, Scharping NE, Jacobs HM, Wang S, Chait A, Rawlings DJ. Cutting Edge: BAFF Overexpression Reduces Atherosclerosis via TACI-Dependent B Cell Activation. THE JOURNAL OF IMMUNOLOGY 2016; 197:4529-4534. [PMID: 27837104 DOI: 10.4049/jimmunol.1601198] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 10/13/2016] [Indexed: 12/27/2022]
Abstract
Patients with systemic lupus erythematosus exhibit accelerated atherosclerosis, a chronic inflammatory disease of the arterial wall. The impact of B cells in atherosclerosis is controversial, with both protective and pathogenic roles described. For example, natural IgM binding conserved oxidized lipid epitopes protect against atherosclerosis, whereas anti-oxidized low-density lipoprotein (oxLDL) IgG likely promotes disease. Because BAFF promotes B cell class-switch recombination and humoral autoimmunity, we hypothesized that excess BAFF would accelerate atherosclerosis. In contrast, BAFF overexpression markedly reduced hypercholesterolemia and atherosclerosis in hyperlipidemic mice. BAFF-mediated atheroprotection required B cells and was associated with increased protective anti-oxLDL IgM. Surprisingly, high-titer anti-oxLDL IgM production and reduced atherosclerosis was dependent on the BAFF family receptor transmembrane activator and CAML interactor. In summary, we identified a novel role for B cell-specific, BAFF-dependent transmembrane activator and CAML interactor signals in atherosclerosis pathogenesis, of particular relevance to the use of BAFF-targeted therapies in systemic lupus erythematosus.
Collapse
Affiliation(s)
- Shaun W Jackson
- Seattle Children's Research Institute, Seattle, WA 98101.,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195
| | | | - Holly M Jacobs
- Seattle Children's Research Institute, Seattle, WA 98101
| | - Shari Wang
- Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195; and
| | - Alan Chait
- Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195; and
| | - David J Rawlings
- Seattle Children's Research Institute, Seattle, WA 98101; .,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195.,Department of Immunology, University of Washington School of Medicine, Seattle, WA 98195
| |
Collapse
|
20
|
Abstract
In the current era, one of the major factors limiting graft survival is chronic antibody-mediated rejection (ABMR), whilst patient survival is impacted by the effects of immunosuppression on susceptibility to infection, malignancy and atherosclerosis. IgG antibodies play a role in all of these processes, and many of their cellular effects are mediated by Fc gamma receptors (FcγRs). These surface receptors are expressed by most immune cells, including B cells, natural killer cells, dendritic cells and macrophages. Genetic variation in FCGR genes is likely to affect susceptibility to ABMR and to modulate the physiological functions of IgG. In this review, we discuss the potential role played by FcγRs in determining outcomes in solid organ transplantation, and how genetic polymorphisms in these receptors may contribute to variations in transplant outcome.
Collapse
Affiliation(s)
- Tomas Castro-Dopico
- Molecular Immunity Unit, Department of Medicine, MRC Laboratory of Molecular Biology, University of Cambridge, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, CB2 0QH UK
| | - Menna R. Clatworthy
- Molecular Immunity Unit, Department of Medicine, MRC Laboratory of Molecular Biology, University of Cambridge, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, CB2 0QH UK
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW Atherosclerotic cardiovascular disease confers significant morbidity and mortality in patients with systemic lupus erythematosus (SLE) and cannot be fully explained by traditional cardiovascular risk factors. Recent immunologic discoveries have outlined putative pathways in SLE that may also accelerate the development of atherosclerosis. RECENT FINDINGS Aberrant innate and adaptive immune responses implicated in lupus pathogenesis may also contribute to the development of accelerated atherosclerosis in these patients. Defective apoptosis, abnormal lipoprotein function, autoantibodies, aberrant neutrophil responses, and a dysregulated type I interferon pathway likely contribute to endothelial dysfunction. SLE macrophages have an inflammatory phenotype that may drive progression of plaque. SUMMARY Recent discoveries have placed increased emphasis on the immunology of atherosclerotic cardiovascular disease. Understanding the factors that drive the increased risk for cardiovascular disease in SLE patients may provide selective therapeutic targets for reducing inflammation and improving outcomes in atherosclerosis.
Collapse
Affiliation(s)
- Laura B. Lewandowski
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Mariana J. Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
22
|
Immune-inflammatory responses in atherosclerosis: Role of an adaptive immunity mainly driven by T and B cells. Immunobiology 2016; 221:1014-33. [PMID: 27262513 DOI: 10.1016/j.imbio.2016.05.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 05/06/2016] [Accepted: 05/23/2016] [Indexed: 01/22/2023]
Abstract
Adaptive immune response plays an important role in atherogenesis. In atherosclerosis, the proinflammatory immune response driven by Th1 is predominant but the anti-inflammatory response mediated mainly by regulatory T cells is also present. The role of Th2 and Th17 cells in atherogenesis is still debated. In the plaque, other T helper cells can be observed such as Th9 and Th22 but is little is known about their impact in atherosclerosis. Heterogeneity of CD4(+) T cell subsets presented in the plaque may suggest for plasticity of T cell that can switch the phenotype dependening on the local microenvironment and activating/blocking stimuli. Effector T cells are able to recognize self-antigens released by necrotic and apoptotic vascular cells and induce a humoral immune reaction. Tth cells resided in the germinal centers help B cells to switch the antibody class to the production of high-affinity antibodies. Humoral immunity is mediated by B cells that release antigen-specific antibodies. A variety of B cell subsets were found in human and murine atherosclerotic plaques. In mice, B1 cells could spontaneously produce atheroprotective natural IgM antibodies. Conventional B2 lymphocytes secrete either proatherogenic IgG, IgA, and IgE or atheroprotective IgG and IgM antibodies reactive with oxidation-specific epitopes on atherosclerosis-associated antigens. A small population of innate response activator (IRA) B cells, which is phenotypically intermediate between B1 and B2 cells, produces IgM but possesses proatherosclerotic properties. Finally, there is a minor subset of splenic regulatory B cells (Bregs) that protect against atherosclerotic inflammation through support of generation of Tregs and production of anti-inflammatory cytokines IL-10 and TGF-β and proapoptotic molecules.
Collapse
|
23
|
Merched AJ, Daret D, Li L, Franzl N, Sauvage-Merched M. Specific autoantigens in experimental autoimmunity-associated atherosclerosis. FASEB J 2016; 30:2123-34. [PMID: 26891734 DOI: 10.1096/fj.201500131] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 02/01/2016] [Indexed: 12/22/2022]
Abstract
Higher cardiovascular morbidity in patients with a wide range of autoimmune diseases highlights the importance of autoimmunity in promoting atherosclerosis. Our purpose was to investigate the mechanisms of accelerated atherosclerosis and identified vascular autoantigens targeted by autoimmunity. We created a mouse model of autoimmunity-associated atherosclerosis by transplanting bone marrow from FcγRIIB knockout (FcRIIB(-/-)) mice into LDL receptor knockout mice. We characterized the cellular and molecular mechanisms of atherogenesis and identified specific aortic autoantigens using serologic proteomic studies. En face lesion area analysis showed more aggressive atherosclerosis in autoimmune mice compared with control mice (0.64 ± 0.12 vs 0.32 ± 0.05 mm(2); P < 0.05, respectively). At the cellular level, FcRIIB(-/-) macrophages showed significant reduction (46-72%) in phagocytic capabilities. Proteomic analysis revealed circulating autoantibodies in autoimmune mice that targeted 25 atherosclerotic lesion proteins, including essential components of adhesion complex, cytoskeleton, and extracellular matrix, and proteins involved in critical functions and pathways. Microscopic examination of atherosclerotic plaques revealed essential colocalization of autoantibodies with endothelial cells, their adherence to basement membranes, the internal elastica lamina, and necrotic cores. The new vascular autoimmunosome may be a useful target for diagnostic and immunotherapeutic interventions in autoimmunity-associated diseases that have accelerated atherosclerosis.-Merched, A. J., Daret, D., Li, L., Franzl, N., Sauvage-Merched, M. Specific autoantigens in experimental autoimmunity-associated atherosclerosis.
Collapse
Affiliation(s)
- Aksam J Merched
- Department of Pharmaceutical Sciences, and INSERM U1053, University of Bordeaux, Bordeaux, France Department of Cell Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Danièle Daret
- Department of Cell Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Lan Li
- Department of Cell Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Nathalie Franzl
- Department of Cell Biology, Baylor College of Medicine, Houston, Texas, USA
| | | |
Collapse
|
24
|
Harmon EY, Fronhofer V, Keller RS, Feustel PJ, Zhu X, Xu H, Avram D, Jones DM, Nagarajan S, Lennartz MR. Anti-inflammatory immune skewing is atheroprotective: Apoe−/−FcγRIIb−/− mice develop fibrous carotid plaques. J Am Heart Assoc 2015; 3:e001232. [PMID: 25516435 PMCID: PMC4338708 DOI: 10.1161/jaha.114.001232] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background Stroke, caused by carotid plaque rupture, is a major cause of death in the United States. Whereas vulnerable human plaques have higher Fc receptor (FcγR) expression than their stable counterparts, how FcγR expression impacts plaque histology is unknown. We investigated the role of FcγRIIb in carotid plaque development and stability in apolipoprotein (Apo)e−/− and Apoe−/−FcγRIIb−/− double knockout (DKO) animals. Methods and Results Plaques were induced by implantation of a shear stress‐modifying cast around the carotid artery. Plaque length and stenosis were followed longitudinally using ultrasound biomicroscopy. Immune status was determined by flow cytometry, cytokine release, immunoglobulin G concentration and analysis of macrophage polarization both in plaques and in vitro. Surprisingly, DKO animals had lower plaque burden in both carotid artery and descending aorta. Plaques from Apoe−/− mice were foam‐cell rich and resembled vulnerable human specimens, whereas those from DKO mice were fibrous and histologically stable. Plaques from DKO animals expressed higher arginase 1 (Arg‐1) and lower inducible nitric oxide synthase (iNOS), indicating the presence of M2 macrophages. Analysis of blood and cervical lymph nodes revealed higher interleukin (IL)‐10, immune complexes, and regulatory T cells (Tregs) and lower IL‐12, IL‐1β, and tumor necrosis factor alpha (TNF‐α) in DKO mice. Similarly, in vitro stimulation produced higher IL‐10 and Arg‐1 and lower iNOS, IL‐1β, and TNF‐α in DKO versus Apoe−/− macrophages. These results define a systemic anti‐inflammatory phenotype. Conclusions We hypothesized that removal of FcγRIIb would exacerbate atherosclerosis and generate unstable plaques. However, we found that deletion of FcγRIIb on a congenic C57BL/6 background induces an anti‐inflammatory Treg/M2 polarization that is atheroprotective.
Collapse
Affiliation(s)
- Erin Y Harmon
- Centers for Cell Biology and Cancer Research, Albany Medical College, Albany, NY
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Xia X, Li Y, Su Q, Huang Z, Shen Y, Li W, Yu C. Inhibitory effects of Mycoepoxydiene on macrophage foam cell formation and atherosclerosis in ApoE-deficient mice. Cell Biosci 2015; 5:23. [PMID: 26045945 PMCID: PMC4455339 DOI: 10.1186/s13578-015-0017-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 05/20/2015] [Indexed: 02/02/2023] Open
Abstract
Background Mycoepoxydiene (MED) is a polyketide that can be isolated from a marine fungus and is associated with various activities, including antitumor and anti-inflammatory functions. However, its effects on atherosclerosis remain unknown. Macrophage-derived foam cells play crucial roles in the initiation and progression of atherosclerotic plaques. In this study, we investigated the effects of MED on oxidized low-density lipoprotein (ox-LDL)-induced macrophage foam cell formation and activation, and on high fat diet (HFD)-induced atherosclerosis in ApoE-deficient (ApoE−/−) mice. Results Our findings show that MED could significantly inhibit ox-LDL-induced macrophage foam cell formation and suppress the expression of lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1), which is a receptor for ox-LDL. Additionally, MED could significantly inhibit the secretion of proinflammatory cytokines, such as tumor necrosis factor (TNF-α), interleukin (IL)-6, and IL-1β. Mechanistically, MED inhibited NF-κB activation by blocking IκB-α degradation and reducing NF-κB DNA binding activity. Moreover, MED dramatically reduced the occurrence of HFD-induced atherosclerotic lesions in ApoE−/− mice. Conclusions Our study shows that MED can inhibit macrophage foam cell formation and activation by inhibiting NF-κB activation, thereby protecting ApoE−/− mice from HFD-induced atherosclerosis. Our findings suggest that MED might be a potential lead compound for the development of antiatherosclerotic therapeutics. Electronic supplementary material The online version of this article (doi:10.1186/s13578-015-0017-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaochun Xia
- The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361005 China
| | - Yang Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiang-An South Road, Xiamen, Fujian 360112 China
| | - Qiang Su
- Medical College, Xiamen University, Xiamen, China
| | - Zhengrong Huang
- The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361005 China
| | - Yuemao Shen
- School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong 250012 China
| | - Weihua Li
- The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361005 China
| | - Chundong Yu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiang-An South Road, Xiamen, Fujian 360112 China
| |
Collapse
|
26
|
Ng HP, Zhu X, Harmon EY, Lennartz MR, Nagarajan S. Reduced Atherosclerosis in apoE-inhibitory FcγRIIb-Deficient Mice Is Associated With Increased Anti-Inflammatory Responses by T Cells and Macrophages. Arterioscler Thromb Vasc Biol 2015; 35:1101-12. [PMID: 25792447 PMCID: PMC4409543 DOI: 10.1161/atvbaha.115.305290] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 02/27/2015] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Fcγ receptors (FcγRs) are classified as activating (FcγRI, III, and IV) and inhibitory (FcγRII) receptors. We have reported that deletion of activating FcγRs in apolipoprotein E (apoE) single knockout mice attenuated atherosclerosis. In this report, we investigated the hypothesis that deficiency of inhibitory FcγRIIb exacerbates atherosclerosis. APPROACH AND RESULTS ApoE-FcγRIIb double knockout mice, congenic to the C57BL/6 (apoE-FcγRIIbB6 (-/-)), were generated and atherosclerotic lesions were assessed. In contrary to our hypothesis, when compared with apoE single knockout mice, arterial lesions were significantly decreased in apoE-FcγRIIbB6 (-/-) male and female mice fed chow or high-fat diets. Chimeric mice generated by transplanting apoE-FcγRIIbB6 (-/-) marrow into apoE single knockout mice also developed reduced lesions. CD4(+) T cells from apoE-FcγRIIbB6 (-/-) mice produced higher levels of interleukin-10 and transforming growth factor-β than their apoE single knockout counterparts. As our findings conflict with a previous report using apoE-FcγRIIb129/B6 (-/-) mice on a mixed genetic background, we investigated whether strain differences contributed to the anti-inflammatory response. Macrophages from FcγRIIb129/B6 (-/-) mice on a mixed genetic background produced more interleukin-1β and MCP-1 (monocyte chemoattractant protein-1) in response to immune complexes, whereas congenic FcγRIIbB6 (-/-) mice generated more interleukin-10 and significantly less interleukin-1β. Interestingly, the expression of lupus-associated slam genes, located in proximity to fcgr2b in mouse chromosome 1, is upregulated only in mixed FcγRIIb129/B6 (-/-) mice. CONCLUSIONS Our findings demonstrate a detrimental role for FcγRIIb signaling in atherosclerosis and the contribution of anti-inflammatory cytokine responses in the attenuated lesions observed in apoE-FcγRIIbB6 (-/-) mice. As 129/sv genome-derived lupus-associated genes have been implicated in lupus phenotype in FcγRIIb129/B6 (-/-) mice, our findings suggest possible epistatic mechanism contributing to the decreased lesions.
Collapse
Affiliation(s)
- Hang Pong Ng
- From the Department of Pathology, Vascular Medicine Institute, University of Pittsburgh, PA (H.P.N., X.Z., S.N.); Department of Microbiology and Immunology (H.P.N., S.N.), University of Arkansas for Medical Sciences, Little Rock; and Center for Cell Biology and Cancer Research, Albany Medical College, NY (E.Y.H., M.R.L.)
| | - Xinmei Zhu
- From the Department of Pathology, Vascular Medicine Institute, University of Pittsburgh, PA (H.P.N., X.Z., S.N.); Department of Microbiology and Immunology (H.P.N., S.N.), University of Arkansas for Medical Sciences, Little Rock; and Center for Cell Biology and Cancer Research, Albany Medical College, NY (E.Y.H., M.R.L.)
| | - Erin Y Harmon
- From the Department of Pathology, Vascular Medicine Institute, University of Pittsburgh, PA (H.P.N., X.Z., S.N.); Department of Microbiology and Immunology (H.P.N., S.N.), University of Arkansas for Medical Sciences, Little Rock; and Center for Cell Biology and Cancer Research, Albany Medical College, NY (E.Y.H., M.R.L.)
| | - Michelle R Lennartz
- From the Department of Pathology, Vascular Medicine Institute, University of Pittsburgh, PA (H.P.N., X.Z., S.N.); Department of Microbiology and Immunology (H.P.N., S.N.), University of Arkansas for Medical Sciences, Little Rock; and Center for Cell Biology and Cancer Research, Albany Medical College, NY (E.Y.H., M.R.L.)
| | - Shanmugam Nagarajan
- From the Department of Pathology, Vascular Medicine Institute, University of Pittsburgh, PA (H.P.N., X.Z., S.N.); Department of Microbiology and Immunology (H.P.N., S.N.), University of Arkansas for Medical Sciences, Little Rock; and Center for Cell Biology and Cancer Research, Albany Medical College, NY (E.Y.H., M.R.L.).
| |
Collapse
|
27
|
Tanigaki K, Sundgren N, Khera A, Vongpatanasin W, Mineo C, Shaul PW. Fcγ receptors and ligands and cardiovascular disease. Circ Res 2015; 116:368-84. [PMID: 25593280 DOI: 10.1161/circresaha.116.302795] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Fcγ receptors (FcγRs) classically modulate intracellular signaling on binding of the Fc region of IgG in immune response cells. How FcγR and their ligands affect cardiovascular health and disease has been interrogated recently in both preclinical and clinical studies. The stimulation of activating FcγR in endothelial cells, vascular smooth muscle cells, and monocytes/macrophages causes a variety of cellular responses that may contribute to vascular disease pathogenesis. Stimulation of the lone inhibitory FγcR, FcγRIIB, also has adverse consequences in endothelial cells, antagonizing NO production and reparative mechanisms. In preclinical disease models, activating FcγRs promote atherosclerosis, whereas FcγRIIB is protective, and activating FcγRs also enhance thrombotic and nonthrombotic vascular occlusion. The FcγR ligand C-reactive protein (CRP) has undergone intense study. Although in rodents CRP does not affect atherosclerosis, it causes hypertension and insulin resistance and worsens myocardial infarction. Massive data have accumulated indicating an association between increases in circulating CRP and coronary heart disease in humans. However, Mendelian randomization studies reveal that CRP is not likely a disease mediator. CRP genetics and hypertension warrant further investigation. To date, studies of genetic variants of activating FcγRs are insufficient to implicate the receptors in coronary heart disease pathogenesis in humans. However, a link between FcγRIIB and human hypertension may be emerging. Further knowledge of the vascular biology of FcγR and their ligands will potentially enhance our understanding of cardiovascular disorders, particularly in patients whose greater predisposition for disease is not explained by traditional risk factors, such as individuals with autoimmune disorders.
Collapse
Affiliation(s)
- Keiji Tanigaki
- From the Department of Pediatrics, Center for Pulmonary and Vascular Biology (K.T., N.S., C.M., P.W.S.), and Division of Cardiology, Department of Internal Medicine (A.K., W.V.), University of Texas Southwestern Medical Center, Dallas
| | - Nathan Sundgren
- From the Department of Pediatrics, Center for Pulmonary and Vascular Biology (K.T., N.S., C.M., P.W.S.), and Division of Cardiology, Department of Internal Medicine (A.K., W.V.), University of Texas Southwestern Medical Center, Dallas
| | - Amit Khera
- From the Department of Pediatrics, Center for Pulmonary and Vascular Biology (K.T., N.S., C.M., P.W.S.), and Division of Cardiology, Department of Internal Medicine (A.K., W.V.), University of Texas Southwestern Medical Center, Dallas
| | - Wanpen Vongpatanasin
- From the Department of Pediatrics, Center for Pulmonary and Vascular Biology (K.T., N.S., C.M., P.W.S.), and Division of Cardiology, Department of Internal Medicine (A.K., W.V.), University of Texas Southwestern Medical Center, Dallas
| | - Chieko Mineo
- From the Department of Pediatrics, Center for Pulmonary and Vascular Biology (K.T., N.S., C.M., P.W.S.), and Division of Cardiology, Department of Internal Medicine (A.K., W.V.), University of Texas Southwestern Medical Center, Dallas
| | - Philip W Shaul
- From the Department of Pediatrics, Center for Pulmonary and Vascular Biology (K.T., N.S., C.M., P.W.S.), and Division of Cardiology, Department of Internal Medicine (A.K., W.V.), University of Texas Southwestern Medical Center, Dallas.
| |
Collapse
|
28
|
|
29
|
Abstract
The development of atherosclerosis is the major etiological factor causing cardiovascular disease and constitutes a lipid-induced, chronic inflammatory and autoimmune disease of the large arteries. A long-standing view of the protective role of B cells in atherosclerosis has been challenged by recent studies using B cell depletion in animal models. Whereas complete B cell deficiency increases atherosclerosis, depletion of B2 but not B1 cells reduces atherosclerosis. This has led to a re-evaluation of the multiple potential pathways by which B cells can regulate atherosclerosis, and the apparent opposing roles of B1 and B2 cells. B cells, in addition to having the unique ability to produce antibodies, are now recognized to play a number of important roles in the immune system, including cytokine production and direct regulation of T cell responses. This review summarizes current knowledge on B cell subsets and functions, and how these could distinctly influence atherosclerosis development.
Collapse
Affiliation(s)
- Andrew P Sage
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge , Cambridge , UK
| | | |
Collapse
|
30
|
Abstract
Insights into the important contribution of inflammation and immune functions in the development and progression of atherosclerosis have greatly improved our understanding of this disease. Although the role of T cells has been extensively studied for decades, only recently has the role of B cells gained more attention. Recent studies have identified differential effects of different B-cell subsets and helped to clarify the still poorly understood mechanisms by which these act. B1 cells have been shown to prevent lesion formation, whereas B2 cells have been suggested to promote it. Natural IgM antibodies, mainly derived from B1 cells, have been shown to mediate atheroprotective effects, but the functional role of other immunoglobulin classes, particularly IgG, still remains elusive. In this review, we will focus on recent insights on the role of B cells and various immunoglobulin classes and how these may mediate their effects in atherosclerotic lesion formation. Moreover, we will highlight potential therapeutic approaches focusing on B-cell depletion that could be used to translate experimental evidence to human disease.
Collapse
Affiliation(s)
- Dimitrios Tsiantoulas
- From the Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria (D.T., C.J.B.); Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (D.T., C.J.B.); and Department of Medicine, University of California San Diego, La Jolla (C.J.D., J.L.W.)
| | - Cody J Diehl
- From the Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria (D.T., C.J.B.); Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (D.T., C.J.B.); and Department of Medicine, University of California San Diego, La Jolla (C.J.D., J.L.W.)
| | - Joseph L Witztum
- From the Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria (D.T., C.J.B.); Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (D.T., C.J.B.); and Department of Medicine, University of California San Diego, La Jolla (C.J.D., J.L.W.)
| | - Christoph J Binder
- From the Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria (D.T., C.J.B.); Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (D.T., C.J.B.); and Department of Medicine, University of California San Diego, La Jolla (C.J.D., J.L.W.).
| |
Collapse
|
31
|
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: 146] [Impact Index Per Article: 14.6] [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
|
32
|
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
|
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: 204] [Impact Index Per Article: 18.5] [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
|
Lichtman AH, Binder CJ, Tsimikas S, Witztum JL. Adaptive immunity in atherogenesis: new insights and therapeutic approaches. J Clin Invest 2013; 123:27-36. [PMID: 23281407 DOI: 10.1172/jci63108] [Citation(s) in RCA: 149] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Many remarkable advances have improved our understanding of the cellular and molecular events in the pathogenesis of atherosclerosis. Chief among these is the accumulating knowledge of how the immune system contributes to all phases of atherogenesis, including well-known inflammatory reactions consequent to intimal trapping and oxidation of LDL. Advances in our understanding of the innate and adaptive responses to these events have helped to clarify the role of inflammation in atherogenesis and suggested new diagnostic modalities and novel therapeutic targets. Here we focus on recent advances in understanding how adaptive immunity affects atherogenesis.
Collapse
Affiliation(s)
- Andrew H Lichtman
- Vascular Research Division, Department of Pathology, Brigham and Women’s Hospital, and Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | | | | | |
Collapse
|
35
|
Perry HM, Bender TP, McNamara CA. B cell subsets in atherosclerosis. Front Immunol 2012; 3:373. [PMID: 23248624 PMCID: PMC3518786 DOI: 10.3389/fimmu.2012.00373] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 11/21/2012] [Indexed: 12/21/2022] Open
Abstract
Atherosclerosis, the underlying cause of heart attacks and strokes, is a chronic inflammatory disease of the artery wall. Immune cells, including lymphocytes modulate atherosclerotic lesion development through interconnected mechanisms. Elegant studies over the past decades have begun to unravel a role for B cells in atherosclerosis. Recent findings provide evidence that B cell effects on atherosclerosis may be subset-dependent. B-1a B cells have been reported to protect from atherosclerosis by secretion of natural IgM antibodies. Conventional B-2 B cells can promote atherosclerosis through less clearly defined mechanism that may involve CD4 T cells. Yet, there may be other populations of B cells within these subsets with different phenotypes altering their impact on atherosclerosis. Additionally, the role of B cell subsets in atherosclerosis may depend on their environmental niche and/or the stage of atherogenesis. This review will highlight key findings in the evolving field of B cells and atherosclerosis and touch on the potential and importance of translating these findings to human disease.
Collapse
Affiliation(s)
- Heather M Perry
- Department of Pathology, University of Virginia Charlottesville, VA, USA ; Cardiovascular Research Center, University of Virginia Health System Charlottesville, VA, USA
| | | | | |
Collapse
|
36
|
Wang X, Liu X, Kishimoto C, Yuan Z. The role of Fcγ receptors in atherosclerosis. Exp Biol Med (Maywood) 2012; 237:609-16. [PMID: 22688821 DOI: 10.1258/ebm.2012.011373] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Atherosclerosis is widely considered to be an immune-mediated process. Fcγ receptors (Fcγ Rs) contribute to the regulation of a multitude of immune and inflammatory responses and are implicated in human atherosclerotic lesions. Major cell types involved in the pathogenesis of atherosclerosis express Fcγ Rs and their proatherogenic ligands such as immune complexes and C-reactive protein, which act to activate Fcγ R signaling pathways. This review summarizes recent significant progress addressing the multifaceted roles of Fcγ Rs in atherogenesis which comes from the studies of Fcγ R-deficient animal models, clinical investigations and in vitro molecular and cellular studies. These new findings help us appreciate the emerging role of Fcγ Rs in atherosclerosis, and suggest Fcγ Rs as a potential therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Xinhong Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, China
| | | | | | | |
Collapse
|
37
|
Humoral and cellular immune responses in atherosclerosis: Spotlight on B- and T-cells. Vascul Pharmacol 2012; 56:193-203. [DOI: 10.1016/j.vph.2012.01.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 01/17/2012] [Accepted: 01/28/2012] [Indexed: 01/20/2023]
|
38
|
Ng HP, Burris RL, Nagarajan S. Attenuated atherosclerotic lesions in apoE-Fcγ-chain-deficient hyperlipidemic mouse model is associated with inhibition of Th17 cells and promotion of regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2011; 187:6082-93. [PMID: 22043015 DOI: 10.4049/jimmunol.1004133] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Though the presence of antioxidized low-density lipoprotein IgG is well documented in clinical and animal studies, the role for FcγRs to the progression of atherosclerosis has not been studied in detail. In the current study, we investigated the role for activating FcγR in the progression of atherosclerosis using apolipoprotein E (apoE)-Fcγ-chain double-knockout (DKO) mice. Relative to apoE knockout (KO) mice, arterial lesion formation was significantly decreased in apoE-Fcγ-chain DKO mice. Bone marrow chimera studies showed reduced lesions in apoE KO mice receiving the bone marrow of apoE-Fcγ-chain DKO mice. Compared to apoE KO mice, antioxidized low-density lipoprotein IgG1 (Th2) and IgG2a (Th1), IL-10, and IFN-γ secretion by activated T cells was increased in apoE-Fcγ-chain DKO mice. These findings suggest that reduced atherosclerotic lesion in apoE-Fcγ-chain DKO mice is not due to a Th1/Th2 imbalance. Interestingly, the number of Th17 cells and the secretion of IL-17 by activated CD4(+) cells were decreased in apoE-Fcγ-chain DKO mice. Notably, the number of regulatory T cells, expression of mRNA, and secretion of TGF-β and IL-10 were increased in apoE-Fcγ-chain DKO mice. Furthermore, secretions of IL-6 and STAT-3 phosphorylation essential for Th17 cell genesis were reduced in apoE-Fcγ-chain DKO mice. Importantly, decrease in Th17 cells in apoE-Fcγ-chain DKO mice was due to reduced IL-6 release by APC of apoE-Fcγ-chain DKO mice. Collectively, our data suggest that activating FcγR promotes atherosclerosis by inducing a Th17 response in the hyperlipidemic apoE KO mouse model.
Collapse
Affiliation(s)
- Hang Pong Ng
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | | | | |
Collapse
|
39
|
Affiliation(s)
- Christoph J. Binder
- From the Center for Molecular Medicine of the Austrian Academy of Sciences (C.J.B.), Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (C.J.B.); Department of Medicine, University of California, San Diego, La Jolla, CA (C.J.B., J.L.W.)
| | - Joseph L. Witztum
- From the Center for Molecular Medicine of the Austrian Academy of Sciences (C.J.B.), Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (C.J.B.); Department of Medicine, University of California, San Diego, La Jolla, CA (C.J.B., J.L.W.)
| |
Collapse
|
40
|
Lawson HA, Cady JE, Partridge C, Wolf JB, Semenkovich CF, Cheverud JM. Genetic effects at pleiotropic loci are context-dependent with consequences for the maintenance of genetic variation in populations. PLoS Genet 2011; 7:e1002256. [PMID: 21931559 PMCID: PMC3169520 DOI: 10.1371/journal.pgen.1002256] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 07/08/2011] [Indexed: 02/06/2023] Open
Abstract
Context-dependent genetic effects, including genotype-by-environment and genotype-by-sex interactions, are a potential mechanism by which genetic variation of complex traits is maintained in populations. Pleiotropic genetic effects are also thought to play an important role in evolution, reflecting functional and developmental relationships among traits. We examine context-dependent genetic effects at pleiotropic loci associated with normal variation in multiple metabolic syndrome (MetS) components (obesity, dyslipidemia, and diabetes-related traits). MetS prevalence is increasing in Western societies and, while environmental in origin, presents substantial variation in individual response. We identify 23 pleiotropic MetS quantitative trait loci (QTL) in an F16 advanced intercross between the LG/J and SM/J inbred mouse strains (Wustl:LG,SM-G16; n = 1002). Half of each family was fed a high-fat diet and half fed a low-fat diet; and additive, dominance, and parent-of-origin imprinting genotypic effects were examined in animals partitioned into sex, diet, and sex-by-diet cohorts. We examine the context-dependency of the underlying additive, dominance, and imprinting genetic effects of the traits associated with these pleiotropic QTL. Further, we examine sequence polymorphisms (SNPs) between LG/J and SM/J as well as differential expression of positional candidate genes in these regions. We show that genetic associations are different in different sex, diet, and sex-by-diet settings. We also show that over- or underdominance and ecological cross-over interactions for single phenotypes may not be common, however multidimensional synthetic phenotypes at loci with pleiotropic effects can produce situations that favor the maintenance of genetic variation in populations. Our findings have important implications for evolution and the notion of personalized medicine. We look at gene-by-diet and gene-by-sex interactions underlying natural variation in multiple metabolic traits mapping to the same regions of the genome in a mouse model. We find that the underlying genetic architecture of these traits is different in different sex and diet contexts. We further use expression data and whole-genome polymorphism data to identify compelling candidates for experimental follow-up. We use these results to examine theoretical evolutionary predictions about how variation in populations can be maintained. There has been much discussion of late on how to use evolutionary theory to inform medical genomics. Mouse models may be especially appropriate for bridging the divide between evolutionary and biomedical research, because they allow the study of the effects of natural alleles on normal variation and because human-mouse homology is well defined. Our study is unique in examining quantitative trait loci from both evolutionary and biomedical perspectives, and we highlight the complex connections of the traits comprising the metabolic syndrome and the evolutionary implications of their underlying genetic architecture. This is important for understanding disease etiology and is relevant to personalized medicine.
Collapse
Affiliation(s)
- Heather A Lawson
- Washington University in St Louis, St Louis, Missouri, United States of America.
| | | | | | | | | | | |
Collapse
|
41
|
Lennartz MR, Aggarwal A, Michaud TM, Feustel PJ, Jones DM, Brosnan MJ, Keller RS, Loegering DJ, Kreienberg PB. Ligation of macrophage Fcγ receptors recapitulates the gene expression pattern of vulnerable human carotid plaques. PLoS One 2011; 6:e21803. [PMID: 21814555 PMCID: PMC3140977 DOI: 10.1371/journal.pone.0021803] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 06/07/2011] [Indexed: 11/24/2022] Open
Abstract
Stroke is a leading cause of death in the United States. As ∼60% of strokes result from carotid plaque rupture, elucidating the mechanisms that underlie vulnerability is critical for therapeutic intervention. We tested the hypothesis that stable and vulnerable human plaques differentially express genes associated with matrix degradation. Examination established that femoral, and the distal region of carotid, plaques were histologically stable while the proximal carotid plaque regions were vulnerable. Quantitative RT-PCR was used to compare expression of 22 genes among these tissues. Distal carotid and femoral gene expression was not significantly different, permitting the distal carotid segments to be used as a paired control for their corresponding proximal regions. Analysis of the paired plaques revealed differences in 16 genes that impact plaque stability: matrix metalloproteinases (MMP, higher in vulnerable), MMP modulators (inhibitors: lower, activators: higher in vulnerable), activating Fc receptors (FcγR, higher in vulnerable) and FcγR signaling molecules (higher in vulnerable). Surprisingly, the relative expression of smooth muscle cell and macrophage markers in the three plaque types was not significantly different, suggesting that macrophage distribution and/or activation state correlates with (in)stability. Immunohistochemistry revealed that macrophages and smooth muscle cells localize to distinct and non-overlapping regions in all plaques. MMP protein localized to macrophage-rich regions. In vitro, treatment of macrophages with immune complexes, but not oxidized low density lipoprotein, C-reactive protein, or TNF-α, induced a gene expression profile similar to that of the vulnerable plaques. That ligation of FcγR recapitulates the pattern of gene expression in vulnerable plaques suggests that the FcγR → macrophage activation pathway may play a greater role in human plaque vulnerability than previously appreciated.
Collapse
Affiliation(s)
- Michelle R Lennartz
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, New York, United States of America.
| | | | | | | | | | | | | | | | | |
Collapse
|