201
|
Ley K. 2015 Russell Ross Memorial Lecture in Vascular Biology: Protective Autoimmunity in Atherosclerosis. Arterioscler Thromb Vasc Biol 2016; 36:429-38. [PMID: 26821946 PMCID: PMC4970520 DOI: 10.1161/atvbaha.115.306009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 01/18/2016] [Indexed: 01/18/2023]
Abstract
Atherosclerosis is an inflammatory disease of the arterial wall. It is accompanied by an autoimmune response against apolipoprotein B-100, the core protein of low-density lipoprotein, which manifests as CD4 T cell and antibody responses. To assess the role of the autoimmune response in atherosclerosis, the nature of the CD4 T cell response against apolipoprotein B-100 was studied with and without vaccination with major histocompatibility complex-II-restricted apolipoprotein B-100 peptides. The immunologic basis of autoimmunity in atherosclerosis is discussed in the framework of theories of adaptive immunity. Older vaccination approaches are also discussed. Vaccinating Apoe(-/-) mice with major histocompatibility complex-II-restricted apolipoprotein B-100 peptides reduces atheroma burden in the aorta by ≈40%. The protective mechanism likely includes secretion of interleukin-10. Protective autoimmunity limits atherosclerosis in mice and suggests potential for developing preventative and therapeutic vaccines for humans.
Collapse
Affiliation(s)
- Klaus Ley
- From the La Jolla Institute for Allergy & Immunology and Department of Bioengineering, UCSD, La Jolla, CA
| |
Collapse
|
202
|
Abstract
Inflammation is essential in the initial development and progression of many cardiovascular diseases involving innate and adaptive immune responses. The role of CD4(+)CD25(+)FOXP3(+) regulatory T (TREG) cells in the modulation of inflammation and immunity has received increasing attention. Given the important role of TREG cells in the induction and maintenance of immune homeostasis and tolerance, dysregulation in the generation or function of TREG cells can trigger abnormal immune responses and lead to pathology. A wealth of evidence from experimental and clinical studies has indicated that TREG cells might have an important role in protecting against cardiovascular disease, in particular atherosclerosis and abdominal aortic aneurysm. In this Review, we provide an overview of the roles of TREG cells in the pathogenesis of a number of cardiovascular diseases, including atherosclerosis, hypertension, ischaemic stroke, abdominal aortic aneurysm, Kawasaki disease, pulmonary arterial hypertension, myocardial infarction and remodelling, postischaemic neovascularization, myocarditis and dilated cardiomyopathy, and heart failure. Although the exact molecular mechanisms underlying the cardioprotective effects of TREG cells are still to be elucidated, targeted therapies with TREG cells might provide a promising and novel future approach to the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Xiao Meng
- The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan, Shandong 250012, China
| | - Jianmin Yang
- The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan, Shandong 250012, China
| | - Mei Dong
- The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan, Shandong 250012, China
| | - Kai Zhang
- The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan, Shandong 250012, China
| | - Eric Tu
- Mucosal Immunology Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Qi Gao
- Department of Clinical Laboratory, Shandong Provincial Hospital affiliated to Shandong University, 324 Jingwu Weiqi Road, Jinan 250021, China
| | - Wanjun Chen
- Mucosal Immunology Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan, Shandong 250012, China
| | - Yun Zhang
- The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan, Shandong 250012, China
| |
Collapse
|
203
|
Babaev VR, Ding L, Zhang Y, May JM, Lin PC, Fazio S, Linton MF. Macrophage IKKα Deficiency Suppresses Akt Phosphorylation, Reduces Cell Survival, and Decreases Early Atherosclerosis. Arterioscler Thromb Vasc Biol 2016; 36:598-607. [PMID: 26848161 DOI: 10.1161/atvbaha.115.306931] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 01/17/2015] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The IκB kinase (IKK) is an enzyme complex that initiates the nuclear factor κB transcription factor cascade, which is important in regulating multiple cellular responses. IKKα is directly associated with 2 major prosurvival pathways, PI3K/Akt and nuclear factor κB, but its role in cell survival is not clear. Macrophages play critical roles in the pathogenesis of atherosclerosis, yet the impact of IKKα signaling on macrophage survival and atherogenesis remains unclear. APPROACH AND RESULTS Here, we demonstrate that genetic IKKα deficiency, as well as pharmacological inhibition of IKK, in mouse macrophages significantly reduces Akt S(473) phosphorylation, which is accompanied by suppression of mTOR complex 2 signaling. Moreover, IKKα null macrophages treated with lipotoxic palmitic acid exhibited early exhaustion of Akt signaling compared with wild-type cells. This was accompanied by a dramatic decrease in the resistance of IKKα(-/-) monocytes and macrophages to different proapoptotic stimuli compared with wild-type cells. In vivo, IKKα deficiency increased macrophage apoptosis in atherosclerotic lesions and decreased early atherosclerosis in both female and male low-density lipoprotein receptor (LDLR)(-/-) mice reconstituted with IKKα(-/-) hematopoietic cells and fed with the Western diet for 8 weeks compared with control LDLR(-/-) mice transplanted with wild-type cells. CONCLUSIONS Hematopoietic IKKα deficiency in mouse suppresses Akt signaling, compromising monocyte/macrophage survival and this decreases early atherosclerosis.
Collapse
Affiliation(s)
- Vladimir R Babaev
- From the Atherosclerosis Research Unit, Department of Medicine (V.R.B., L.D., Y.Z., J.M.M., M.R.F.L.) and Pharmacology (M.R.F.L.), Vanderbilt University Medical Center, Nashville, TN; Center for Cancer Research, National Cancer Institute, Frederick, MD (P.C.L.); and Department of Medicine, Center of Preventive Cardiology, Oregon Health & Science University, Portland, OR (S.F.).
| | - Lei Ding
- From the Atherosclerosis Research Unit, Department of Medicine (V.R.B., L.D., Y.Z., J.M.M., M.R.F.L.) and Pharmacology (M.R.F.L.), Vanderbilt University Medical Center, Nashville, TN; Center for Cancer Research, National Cancer Institute, Frederick, MD (P.C.L.); and Department of Medicine, Center of Preventive Cardiology, Oregon Health & Science University, Portland, OR (S.F.)
| | - Youmin Zhang
- From the Atherosclerosis Research Unit, Department of Medicine (V.R.B., L.D., Y.Z., J.M.M., M.R.F.L.) and Pharmacology (M.R.F.L.), Vanderbilt University Medical Center, Nashville, TN; Center for Cancer Research, National Cancer Institute, Frederick, MD (P.C.L.); and Department of Medicine, Center of Preventive Cardiology, Oregon Health & Science University, Portland, OR (S.F.)
| | - James M May
- From the Atherosclerosis Research Unit, Department of Medicine (V.R.B., L.D., Y.Z., J.M.M., M.R.F.L.) and Pharmacology (M.R.F.L.), Vanderbilt University Medical Center, Nashville, TN; Center for Cancer Research, National Cancer Institute, Frederick, MD (P.C.L.); and Department of Medicine, Center of Preventive Cardiology, Oregon Health & Science University, Portland, OR (S.F.)
| | - P Charles Lin
- From the Atherosclerosis Research Unit, Department of Medicine (V.R.B., L.D., Y.Z., J.M.M., M.R.F.L.) and Pharmacology (M.R.F.L.), Vanderbilt University Medical Center, Nashville, TN; Center for Cancer Research, National Cancer Institute, Frederick, MD (P.C.L.); and Department of Medicine, Center of Preventive Cardiology, Oregon Health & Science University, Portland, OR (S.F.)
| | - Sergio Fazio
- From the Atherosclerosis Research Unit, Department of Medicine (V.R.B., L.D., Y.Z., J.M.M., M.R.F.L.) and Pharmacology (M.R.F.L.), Vanderbilt University Medical Center, Nashville, TN; Center for Cancer Research, National Cancer Institute, Frederick, MD (P.C.L.); and Department of Medicine, Center of Preventive Cardiology, Oregon Health & Science University, Portland, OR (S.F.)
| | - MacRae F Linton
- From the Atherosclerosis Research Unit, Department of Medicine (V.R.B., L.D., Y.Z., J.M.M., M.R.F.L.) and Pharmacology (M.R.F.L.), Vanderbilt University Medical Center, Nashville, TN; Center for Cancer Research, National Cancer Institute, Frederick, MD (P.C.L.); and Department of Medicine, Center of Preventive Cardiology, Oregon Health & Science University, Portland, OR (S.F.)
| |
Collapse
|
204
|
Li Y, Kanellakis P, Hosseini H, Cao A, Deswaerte V, Tipping P, Toh BH, Bobik A, Kyaw T. A CD1d-dependent lipid antagonist to NKT cells ameliorates atherosclerosis in ApoE −/−mice by reducing lesion necrosis and inflammation. Cardiovasc Res 2016; 109:305-317. [DOI: 10.1093/cvr/cvv259] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
205
|
Natorska J, Marek G, Sadowski J, Undas A. Presence of B cells within aortic valves in patients with aortic stenosis: Relation to severity of the disease. J Cardiol 2016; 67:80-5. [DOI: 10.1016/j.jjcc.2015.05.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 04/03/2015] [Accepted: 05/07/2015] [Indexed: 11/29/2022]
|
206
|
Ding R, Gao W, He Z, Wang H, Conrad CD, Dinney CM, Yuan X, Wu F, Ma L, Wu Z, Liang C. Overrepresentation of Th1- and Th17-like Follicular Helper T Cells in Coronary Artery Disease. J Cardiovasc Transl Res 2015; 8:503-5. [PMID: 26597829 DOI: 10.1007/s12265-015-9662-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 11/10/2015] [Indexed: 10/22/2022]
Abstract
T cells and B cells play substantial roles in the process of coronary artery disease (CAD). Here, we examined the role of circulating follicular helper T (Tfh) cells in CAD. Compared to non-CAD controls, CAD patients had increased levels of circulating Tfh. Also, circulating Tfh in CAD patients exhibited increased frequencies of Th1- and Th17-like phenotypes and aberrant cytokine expressions. Coculture experiments with B cells showed that Tfh from CAD patients were more potent at inducing antibody production from B cells, enhancing plasmablast differentiation and suppressing B10 cell differentiation. Importantly, we found that the skewing of circulating Tfh toward the Th1/Th17-like cells was directly correlated with B cell inflammation and low density lipoprotein level in CAD patients. Together, our data demonstrated a skewing of blood Tfh composition in CAD patients, which resulted in significant changes in B cell inflammation.
Collapse
Affiliation(s)
- Ru Ding
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Wenwu Gao
- Department of Orthopedics, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Zhiqing He
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Haibing Wang
- Ganbu Ward, No. 401 Hospital of PLA, Qingdao, Shandong, China
| | | | | | - Xiaozou Yuan
- Research Center, BGC Biotechnology, Chicago, IL, USA
| | - Feng Wu
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Lan Ma
- Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Zonggui Wu
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China.
| | - Chun Liang
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China.
| |
Collapse
|
207
|
Centa M, Gruber S, Nilsson D, Polyzos KA, Johansson DK, Hansson GK, Ketelhuth DFJ, Binder CJ, Malin S. Atherosclerosis Susceptibility in Mice Is Independent of the V1 Immunoglobulin Heavy Chain Gene. Arterioscler Thromb Vasc Biol 2015; 36:25-36. [PMID: 26564818 PMCID: PMC4684249 DOI: 10.1161/atvbaha.115.305990] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 11/04/2015] [Indexed: 12/02/2022]
Abstract
Supplemental Digital Content is available in the text. The V1 (VHS107.1.42) immunoglobulin heavy chain gene is thought to be critical in producing IgM natural antibodies of the T15-idiotype that protect against both atherosclerosis and infection from Streptococcus pneumoniae. Our aim was to determine whether genetic loss of the V1 gene increased atherosclerotic plaque burden in vivo because of a reduction in the T15-idiotype or other atheroprotective antibodies.
Collapse
Affiliation(s)
- Monica Centa
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden (M.C., D.N., K.A.P., D.K.J., G.K.H., D.F.J.K., S.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (S.G., C.J.B.); and Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (S.G., C.J.B.)
| | - Sabrina Gruber
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden (M.C., D.N., K.A.P., D.K.J., G.K.H., D.F.J.K., S.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (S.G., C.J.B.); and Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (S.G., C.J.B.)
| | - Daniel Nilsson
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden (M.C., D.N., K.A.P., D.K.J., G.K.H., D.F.J.K., S.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (S.G., C.J.B.); and Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (S.G., C.J.B.)
| | - Konstantinos A Polyzos
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden (M.C., D.N., K.A.P., D.K.J., G.K.H., D.F.J.K., S.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (S.G., C.J.B.); and Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (S.G., C.J.B.)
| | - Daniel K Johansson
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden (M.C., D.N., K.A.P., D.K.J., G.K.H., D.F.J.K., S.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (S.G., C.J.B.); and Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (S.G., C.J.B.)
| | - Göran K Hansson
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden (M.C., D.N., K.A.P., D.K.J., G.K.H., D.F.J.K., S.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (S.G., C.J.B.); and Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (S.G., C.J.B.)
| | - Daniel F J Ketelhuth
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden (M.C., D.N., K.A.P., D.K.J., G.K.H., D.F.J.K., S.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (S.G., C.J.B.); and Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (S.G., C.J.B.)
| | - Christoph J Binder
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden (M.C., D.N., K.A.P., D.K.J., G.K.H., D.F.J.K., S.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (S.G., C.J.B.); and Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (S.G., C.J.B.)
| | - Stephen Malin
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden (M.C., D.N., K.A.P., D.K.J., G.K.H., D.F.J.K., S.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (S.G., C.J.B.); and Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria (S.G., C.J.B.).
| |
Collapse
|
208
|
Abstract
The immune reactions that regulate atherosclerotic plaque inflammation involve chemokines, lipid mediators and costimulatory molecules. Chemokines are a family of chemotactic cytokines that mediate immune cell recruitment and control cell homeostasis and activation of different immune cell types and subsets. Chemokine production and activation of chemokine receptors form a positive feedback mechanism to recruit monocytes, neutrophils and lymphocytes into the atherosclerotic plaque. In addition, chemokine signalling affects immune cell mobilization from the bone marrow. Targeting several of the chemokines and/or chemokine receptors reduces experimental atherosclerosis, whereas specific chemokine pathways appear to be involved in plaque regression. Leukotrienes are lipid mediators that are formed locally in atherosclerotic lesions from arachidonic acid. Leukotrienes mediate immune cell recruitment and activation within the plaque as well as smooth muscle cell proliferation and endothelial dysfunction. Antileukotrienes decrease experimental atherosclerosis, and recent observational data suggest beneficial clinical effects of leukotriene receptor antagonism in cardiovascular disease prevention. By contrast, other lipid mediators, such as lipoxins and metabolites of omega-3 fatty acids, have been associated with the resolution of inflammation. Costimulatory molecules play a central role in fine-tuning immunological reactions and mediate crosstalk between innate and adaptive immunity in atherosclerosis. Targeting these interactions is a promising approach for the treatment of atherosclerosis, but immunological side effects are still a concern. In summary, targeting chemokines, leukotriene receptors and costimulatory molecules could represent potential therapeutic strategies to control atherosclerotic plaque inflammation.
Collapse
Affiliation(s)
- M Bäck
- Translational Cardiology, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - C Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Munich, Germany.,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands
| | - E Lutgens
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Munich, Germany.,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany.,Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
209
|
Le Borgne M, Caligiuri G, Nicoletti A. Once Upon a Time: The Adaptive Immune Response in Atherosclerosis--a Fairy Tale No More. Mol Med 2015; 21 Suppl 1:S13-8. [PMID: 26605642 DOI: 10.2119/molmed.2015.00027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 02/03/2015] [Indexed: 01/06/2023] Open
Abstract
Extensive research has been carried out to decipher the function of the adaptive immune response in atherosclerosis, with the expectation that it will pave the road for the design of immunomodulatory therapies that will prevent or reverse the progression of the disease. All this work has led to the concept that some T- and B-cell subsets are proatherogenic, whereas others are atheroprotective. In addition to the immune response occurring in the spleen and lymph nodes, it has been shown that lymphoid neo-genesis takes place in the adventitia of atherosclerotic vessels, leading to the formation of tertiary lymphoid organs where an adaptive immune response can be mounted. Whereas the mechanisms orchestrating the formation of these organs are becoming better understood, their impact on atherosclerosis progression remains unclear. Several potential therapeutic strategies against atherosclerosis, such as protective vaccination against atherosclerosis antigens or inhibiting the activation of proatherogenic B cells, have been proposed based on our improving knowledge of the role of the immune system in atherosclerosis. These strategies have shown success in preclinical studies, giving hope that they will lead to clinical applications.
Collapse
Affiliation(s)
- Marie Le Borgne
- Unité 1148, Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital Xavier Bichat, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Paris, France; Département Hospitalo-Universitaire DHU FIRE, Paris, France
| | - Giuseppina Caligiuri
- Unité 1148, Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital Xavier Bichat, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Paris, France; Département Hospitalo-Universitaire DHU FIRE, Paris, France
| | - Antonino Nicoletti
- Unité 1148, Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital Xavier Bichat, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Paris, France; Département Hospitalo-Universitaire DHU FIRE, Paris, France
| |
Collapse
|
210
|
Lupieri A, Smirnova N, Malet N, Gayral S, Laffargue M. PI3K signaling in arterial diseases: Non redundant functions of the PI3K isoforms. Adv Biol Regul 2015; 59:4-18. [PMID: 26238239 DOI: 10.1016/j.jbior.2015.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 06/15/2015] [Accepted: 06/15/2015] [Indexed: 06/04/2023]
Abstract
Cardiovascular diseases are the most common cause of death around the world. This includes atherosclerosis and the adverse effects of its treatment, such as restenosis and thrombotic complications. The development of these arterial pathologies requires a series of highly-intertwined interactions between immune and arterial cells, leading to specific inflammatory and fibroproliferative cellular responses. In the last few years, the study of phosphoinositide 3-kinase (PI3K) functions has become an attractive area of investigation in the field of arterial diseases, especially since inhibitors of specific PI3K isoforms have been developed. The PI3K family includes 8 members divided into classes I, II or III depending on their substrate specificity. Although some of the different isoforms are responsible for the production of the same 3-phosphoinositides, they each have specific, non-redundant functions as a result of differences in expression levels in different cell types, activation mechanisms and specific subcellular locations. This review will focus on the functions of the different PI3K isoforms that are suspected as having protective or deleterious effects in both the various immune cells and types of cell found in the arterial wall. It will also discuss our current understanding in the context of which PI3K isoform(s) should be targeted for future therapeutic interventions to prevent or treat arterial diseases.
Collapse
Affiliation(s)
- Adrien Lupieri
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France
| | - Natalia Smirnova
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France
| | - Nicole Malet
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France
| | - Stéphanie Gayral
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France
| | - Muriel Laffargue
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France.
| |
Collapse
|
211
|
Sage AP, Nus M, Baker LL, Finigan AJ, Masters LM, Mallat Z. Regulatory B Cell–Specific Interleukin-10 Is Dispensable for Atherosclerosis Development in Mice. Arterioscler Thromb Vasc Biol 2015; 35:1770-3. [DOI: 10.1161/atvbaha.115.305568] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 06/05/2015] [Indexed: 12/24/2022]
Affiliation(s)
- Andrew P. Sage
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S, M.S, L.L.B., A.J.F., L.M.M., Z.M.); and Institut National de la Sante et de la Recherche Medicale, Paris, France (Z.M.)
| | - Meritxell Nus
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S, M.S, L.L.B., A.J.F., L.M.M., Z.M.); and Institut National de la Sante et de la Recherche Medicale, Paris, France (Z.M.)
| | - Lauren L. Baker
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S, M.S, L.L.B., A.J.F., L.M.M., Z.M.); and Institut National de la Sante et de la Recherche Medicale, Paris, France (Z.M.)
| | - Alison J. Finigan
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S, M.S, L.L.B., A.J.F., L.M.M., Z.M.); and Institut National de la Sante et de la Recherche Medicale, Paris, France (Z.M.)
| | - Leanne M. Masters
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S, M.S, L.L.B., A.J.F., L.M.M., Z.M.); and Institut National de la Sante et de la Recherche Medicale, Paris, France (Z.M.)
| | - Ziad Mallat
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S, M.S, L.L.B., A.J.F., L.M.M., Z.M.); and Institut National de la Sante et de la Recherche Medicale, Paris, France (Z.M.)
| |
Collapse
|
212
|
Rosenfeld SM, Perry HM, Gonen A, Prohaska TA, Srikakulapu P, Grewal S, Das D, McSkimming C, Taylor AM, Tsimikas S, Bender TP, Witztum JL, McNamara CA. B-1b Cells Secrete Atheroprotective IgM and Attenuate Atherosclerosis. Circ Res 2015; 117:e28-39. [PMID: 26082558 DOI: 10.1161/circresaha.117.306044] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 06/16/2015] [Indexed: 01/28/2023]
Abstract
RATIONALE B cells contribute to atherosclerosis through subset-specific mechanisms. Whereas some controversy exists about the role of B-2 cells, B-1a cells are atheroprotective because of secretion of atheroprotective IgM antibodies independent of antigen. B-1b cells, a unique subset of B-1 cells that respond specifically to T-cell-independent antigens, have not been studied within the context of atherosclerosis. OBJECTIVE To determine whether B-1b cells produce atheroprotective IgM antibodies and function to protect against diet-induced atherosclerosis. METHODS AND RESULTS We demonstrate that B-1b cells are sufficient to produce IgM antibodies against oxidation-specific epitopes on low-density lipoprotein both in vitro and in vivo. In addition, we demonstrate that B-1b cells provide atheroprotection after adoptive transfer into B- and T-cell deficient (Rag1(-/-)Apoe(-/-)) hosts. We implicate inhibitor of differentiation 3 (Id3) in the regulation of B-1b cells as B-cell-specific Id3 knockout mice (Id3(BKO)Apoe(-/-)) have increased numbers of B-1b cells systemically, increased titers of oxidation-specific epitope-reactive IgM antibodies, and significantly reduced diet-induced atherosclerosis when compared with Id3(WT)Apoe(-/-) controls. Finally, we report that the presence of a homozygous single nucleotide polymorphism in ID3 in humans that attenuates Id3 function is associated with an increased percentage of circulating B-1 cells and anti-malondialdehyde-low-density lipoprotein IgM suggesting clinical relevance. CONCLUSIONS These results provide novel evidence that B-1b cells produce atheroprotective oxidation-specific epitope-reactive IgM antibodies and protect against atherosclerosis in mice and suggest that similar mechanisms may occur in humans.
Collapse
Affiliation(s)
- Sam M Rosenfeld
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Heather M Perry
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Ayelet Gonen
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Thomas A Prohaska
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Prasad Srikakulapu
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Sukhdeep Grewal
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Deepanjana Das
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Chantel McSkimming
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Angela M Taylor
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Sotirios Tsimikas
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Timothy P Bender
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Joseph L Witztum
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Coleen A McNamara
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla.
| |
Collapse
|
213
|
Abstract
Coronary artery disease (CAD) is the leading cause of death in the United States. Although CAD was formerly considered a lipid accumulation-mediated disease, it has now been clearly shown to involve an ongoing inflammatory response. Advances in basic science research have established the crucial role of inflammation in mediating all stages of CAD. Today, there is convincing evidence that multiple interrelated immune mechanisms interact with metabolic risk factors to initiate, promote, and ultimately activate lesions in the coronary arteries. This review aims to provide current evidence pertaining to the role of inflammation in the pathogenesis of CAD and discusses the impact of inflammatory markers and their modification on clinical outcomes.
Collapse
|
214
|
Hosseini H, Li Y, Kanellakis P, Tay C, Cao A, Tipping P, Bobik A, Toh BH, Kyaw T. Phosphatidylserine liposomes mimic apoptotic cells to attenuate atherosclerosis by expanding polyreactive IgM producing B1a lymphocytes. Cardiovasc Res 2015; 106:443-452. [DOI: 10.1093/cvr/cvv037] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
215
|
Macrophages and immune cells in atherosclerosis: recent advances and novel concepts. Basic Res Cardiol 2015; 110:34. [PMID: 25947006 DOI: 10.1007/s00395-015-0491-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 04/17/2015] [Accepted: 04/30/2015] [Indexed: 01/13/2023]
Abstract
Atherosclerotic lesion-related thrombosis is the major cause of myocardial infarction and stroke, which together constitute the leading cause of mortality worldwide. The inflammatory response is considered as a predominant driving force in atherosclerotic plaque formation, growth and progression towards instability and rupture. Notably, accumulation of macrophages in the intima and emergence of a pro-inflammatory milieu are a characteristic feature of plaque progression, and these processes can be modulated by adaptive immune responses. Recently, novel evidences of onsite proliferation of macrophages in lesions and transdifferentiation of smooth muscle cells to macrophages have challenged the prevalent paradigm that macrophage accumulation mostly relies on recruitment of circulating monocytes to plaques. Furthermore, previously unrecognized roles of inflammatory cell subsets such as plasmacytoid dendritic cells, innate response activator B cells or CD8(+) T cells in atherosclerosis have emerged, as well as novel mechanisms by which regulatory T cells or natural killer T cells contribute to lesion formation. Here, we review and discuss these recent advances in our understanding of inflammatory processes in atherosclerosis.
Collapse
|
216
|
Magen E, Mishal J, Vardy D. Selective IgE deficiency and cardiovascular diseases. Allergy Asthma Proc 2015; 36:225-9. [PMID: 25976439 DOI: 10.2500/aap.2015.36.3825] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Selective immunoglobulin E (IgE) deficiency (IgED) is defined as serum levels of IgE more than or equal to 2 kIU/L and is associated with immune dysregulation and autoimmunity. This study aimed to investigate a prevalence of atherosclerotic cardiovascular disease (ASCVD) in population with IgED. Within the electronic patient record (EPR) database of Leumit Health Care Services (LHS) in Israel, data capture was performed using IBM Cognos 10.1.1 BI Report Studio software. The case samples were drawn from the full study population (n = 18,487), having any allergy-related symptoms and/or those requesting antiallergy medications and performed serum total IgE measurement during 2012 at LHS. All subjects aged more than or equal to 40 years old, with serum total IgE less than 2 kIU/L were included in case group. Control group was randomly sampled from the remained subjects, with a case-control ratio of 10 controls for each case (1:10). The comorbid cardiovascular diseases during less than or equal to 10 years before serum total IgE testing were identified and retrieved using specific International Classification of Diseases, 9th Revision, Clinical Modification diagnostic codes. There were 103 in case and 1030 subjects in control group. Compared with control group patients, the case group had significantly more arterial hypertension [34 (37.7%) versus 187 (18.2%), p < 0.001], ischemic heart disease (IHD) [26 (25.2%) versus 87 (8.4%), p < 0.001], carotid stenosis [5 (4.9%) versus 7 (0.7%), p = 0.003], cerebrovascular disease (CVD) [3 (2.9%) versus 5 (0.5%), p = 0.029], and peripheral vascular disease (PVD) [4 (3.9%) versus 9 (0.9%), p = 0.024]. IgED is associated with higher prevalence of arterial hypertension and ASCVD.
Collapse
Affiliation(s)
- Eli Magen
- Leumit Health Services, Barzilai Medical Center, Ben-Gurion University of Negev, Ashkelon, Israel
| | | | | |
Collapse
|
217
|
Fairweather D. Sex differences in inflammation during atherosclerosis. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2015; 8:49-59. [PMID: 25983559 PMCID: PMC4405090 DOI: 10.4137/cmc.s17068] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 02/02/2015] [Accepted: 02/09/2015] [Indexed: 12/17/2022]
Abstract
Atherosclerosis is the leading cause of death in the United States and worldwide, yet more men die from atherosclerosis than women, and at a younger age. Women, on the other hand, mainly develop atherosclerosis following menopause, and particularly if they have one or more autoimmune diseases, suggesting that the immune mechanisms that increase disease in men are different from those in women. The key processes in the pathogenesis of atherosclerosis are vascular inflammation, lipid accumulation, intimal thickening and fibrosis, remodeling, and plaque rupture or erosion leading to myocardial infarction and ischemia. Evidence indicates that sex hormones alter the immune response during atherosclerosis, resulting in different disease phenotypes according to sex. Women, for example, respond to infection and damage with increased antibody and autoantibody responses, while men have elevated innate immune activation. This review describes current knowledge regarding sex differences in the inflammatory immune response during atherosclerosis. Understanding sex differences is critical for improving individualized medicine.
Collapse
Affiliation(s)
- DeLisa Fairweather
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
218
|
Ilhan F, Kalkanli ST. Atherosclerosis and the role of immune cells. World J Clin Cases 2015; 3:345-352. [PMID: 25879006 PMCID: PMC4391004 DOI: 10.12998/wjcc.v3.i4.345] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 06/30/2014] [Accepted: 01/20/2015] [Indexed: 02/05/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease arising from lipids, specifically low-density lipoproteins, and leukocytes. Following the activation of endothelium with the expression of adhesion molecules and monocytes, inflammatory cytokines from macrophages, and plasmacytoid dendritic cells, high levels of interferon (IFN)-α and β are generated upon the activation of toll-like receptor-9, and T-cells, especially the ones with Th1 profile, produce pro-inflammatory mediators such as IFN-γ and upregulate macrophages to adhere to the endothelium and migrate into the intima. This review presents an exhaustive account for the role of immune cells in the atherosclerosis.
Collapse
|
219
|
Sterile inflammation in the spleen during atherosclerosis provides oxidation-specific epitopes that induce a protective B-cell response. Proc Natl Acad Sci U S A 2015; 112:E2030-8. [PMID: 25848033 DOI: 10.1073/pnas.1421227112] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The B-cell response in atherosclerosis is directed toward oxidation-specific epitopes such as phosphorylcholine (PC) that arise during disease-driven oxidation of self-antigens. PC-bearing antigens have been used to induce atheroprotective antibodies against modified low-density lipoproteins (oxLDL), leading to plaque reduction. Previous studies have found that B-cell transfer from aged atherosclerotic mice confers protection to young mice, but the mechanism is unknown. Here, we dissected the atheroprotective response in the spleen and found an ongoing germinal center reaction, accumulation of antibody-forming cells, and inflammasome activation in apolipoprotein E-deficient mice (Apoe(-/-)). Specific B-cell clone expansion involved the heavy chain variable region (Vh) 5 and Vh7 B-cell receptor families that harbor anti-PC reactivity. oxLDL also accumulated in the spleen. To investigate whether protection could be induced by self-antigens alone, we injected apoptotic cells that carry the same oxidation-specific epitopes as oxLDL. This treatment reduced serum cholesterol and inhibited the development of atherosclerosis in a B-cell-dependent manner. Thus, we conclude that the spleen harbors a protective B-cell response that is initiated in atherosclerosis through sterile inflammation. These data highlight the importance of the spleen in atherosclerosis-associated immunity.
Collapse
|
220
|
Zarzycka B, Nicolaes GAF, Lutgens E. Targeting the adaptive immune system: new strategies in the treatment of atherosclerosis. Expert Rev Clin Pharmacol 2015; 8:297-313. [PMID: 25843158 DOI: 10.1586/17512433.2015.1025052] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Atherosclerosis is a lipid-driven chronic inflammatory disease of the arterial wall. Current treatment of atherosclerosis is focused on limiting its risk factors, such as hyperlipidemia or hypertension. However, treatments that target the inflammatory nature of atherosclerosis are still under development. Discovery of novel targets involved in the inflammation of the arterial wall creates opportunities to design new therapeutics that successfully modulate atherosclerosis. Here, we review drug targets that have proven to play pivotal roles in the adaptive immune system in atherosclerosis, and we discuss their potential as novel therapeutics.
Collapse
Affiliation(s)
- Barbara Zarzycka
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands
| | | | | |
Collapse
|
221
|
Inflammatory mediators in vascular disease: identifying promising targets for intracranial aneurysm research. Mediators Inflamm 2015; 2015:896283. [PMID: 25922566 PMCID: PMC4397479 DOI: 10.1155/2015/896283] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 03/18/2015] [Accepted: 03/19/2015] [Indexed: 12/21/2022] Open
Abstract
Inflammatory processes are implicated in many diseases of the vasculature and have been shown to play a key role in the formation of intracranial aneurysms (IAs). Although the specific mechanisms underlying these processes have been thoroughly investigated in related pathologies, such as atherosclerosis, there remains a paucity of information regarding the immunopathology of IA. Cells such as macrophages and lymphocytes and their effector molecules have been suggested to be players in IA, but their specific interactions and the role of other components of the inflammatory response have yet to be determined. Drawing parallels between the pathogenesis of IA and other vascular disorders could provide a roadmap for developing a mechanistic understanding of the immunopathology of IA and uncovering useful targets for therapeutic intervention. Future research should address the presence and function of leukocyte subsets, mechanisms of leukocyte recruitment and activation, and the role of damage-associated molecular patterns in IA.
Collapse
|
222
|
Virtue A, Mai J, Wang H, Yang X. Lymphocytes and Atherosclerosis. Atherosclerosis 2015. [DOI: 10.1002/9781118828533.ch13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
223
|
van Dijk RA, Duinisveld AJF, Schaapherder AF, Mulder-Stapel A, Hamming JF, Kuiper J, de Boer OJ, van der Wal AC, Kolodgie FD, Virmani R, Lindeman JHN. A change in inflammatory footprint precedes plaque instability: a systematic evaluation of cellular aspects of the adaptive immune response in human atherosclerosis. J Am Heart Assoc 2015; 4:jah3876. [PMID: 25814626 PMCID: PMC4579929 DOI: 10.1161/jaha.114.001403] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Background Experimental studies characterize adaptive immune response as a critical factor in the progression and complications of atherosclerosis. Yet, it is unclear whether these observations translate to the human situation. This study systematically evaluates cellular components of the adaptive immune response in a biobank of human aortas covering the full spectrum of atherosclerotic disease. Methods and Results A systematic analysis was performed on 114 well‐characterized perirenal aortic specimens with immunostaining for T‐cell subsets (CD3/4/8/45RA/45RO/FoxP3) and the Th1/non‐Th1/Th17 ratio (CD4+T‐bet+/CD4+T‐bet−/CD4+/interleukin‐17+ double staining). CD20 and CD138 were used to identify B cells and plasma cells, while B‐cell maturation was evaluated by AID/CD21 staining and expression of lymphoid homeostatic CXCL13. Scattered CD4 and CD8 cells with a T memory subtype were found in normal aorta and early, nonprogressive lesions. The total number of T cells increases in progressive atherosclerotic lesions (≈1:5 CD4/CD8 T‐cell ratio). A further increase in medial and adventitial T cells is found upon progression to vulnerable lesions. This critical stage is further hallmarked by de novo formation of adventitial lymphoidlike structures containing B cells and plasma cells, a process accompanied by transient expression of CXCL13. A dramatic reduction of T‐cell subsets, disappearance of lymphoid structures, and loss of CXCL13 expression characterize postruptured lesions. FoxP3 and Th17 T cells were minimally present throughout the atherosclerotic process. Conclusions Transient CXCL13 expression, restricted presence of B cells in human atherosclerosis, along with formation of nonfunctional extranodal lymphoid structures in the phase preceding plaque rupture, indicates a “critical” change in the inflammatory footprint before and during plaque destabilization.
Collapse
Affiliation(s)
- R A van Dijk
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands (D., D., M.S., H., L.)
| | - A J F Duinisveld
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands (D., D., M.S., H., L.)
| | - A F Schaapherder
- Department of Transplantation Surgery, Leiden University Medical Center, Leiden, The Netherlands (S.)
| | - A Mulder-Stapel
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands (D., D., M.S., H., L.)
| | - J F Hamming
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands (D., D., M.S., H., L.)
| | - J Kuiper
- Gorlaeus Laboratories, Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands (K.)
| | - O J de Boer
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands (B., W.)
| | - A C van der Wal
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands (B., W.)
| | - F D Kolodgie
- CVPath Institute Inc., Gaithersburg, MD (K., V.)
| | - R Virmani
- CVPath Institute Inc., Gaithersburg, MD (K., V.)
| | - J H N Lindeman
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands (D., D., M.S., H., L.)
| |
Collapse
|
224
|
Affiliation(s)
- Catherine C Hedrick
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA.
| |
Collapse
|
225
|
Patel J, McNeill E, Douglas G, Hale AB, de Bono J, Lee R, Iqbal AJ, Regan-Komito D, Stylianou E, Greaves DR, Channon KM. RGS1 regulates myeloid cell accumulation in atherosclerosis and aortic aneurysm rupture through altered chemokine signalling. Nat Commun 2015; 6:6614. [PMID: 25782711 PMCID: PMC4374153 DOI: 10.1038/ncomms7614] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 02/12/2015] [Indexed: 12/31/2022] Open
Abstract
Chemokine signalling drives monocyte recruitment in atherosclerosis and aortic aneurysms. The mechanisms that lead to retention and accumulation of macrophages in the vascular wall remain unclear. Regulator of G-Protein Signalling-1 (RGS1) deactivates G-protein signalling, reducing the response to sustained chemokine stimulation. Here we show that Rgs1 is upregulated in atherosclerotic plaque and aortic aneurysms. Rgs1 reduces macrophage chemotaxis and desensitizes chemokine receptor signalling. In early atherosclerotic lesions, Rgs1 regulates macrophage accumulation and is required for the formation and rupture of Angiotensin II-induced aortic aneurysms, through effects on leukocyte retention. Collectively, these data reveal a role for Rgs1 in leukocyte trafficking and vascular inflammation and identify Rgs1, and inhibition of chemokine receptor signalling as potential therapeutic targets in vascular disease.
Collapse
Affiliation(s)
- Jyoti Patel
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Eileen McNeill
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Gillian Douglas
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Ashley B. Hale
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Joseph de Bono
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Regent Lee
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Asif J. Iqbal
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Daniel Regan-Komito
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | | | - David R. Greaves
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Keith M. Channon
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| |
Collapse
|
226
|
Khoo LHB, Thiam CH, Soh SY, Angeli V. Splenic extrafollicular reactions and BM plasma cells sustain IgM response associated with hypercholesterolemia. Eur J Immunol 2015; 45:1300-12. [DOI: 10.1002/eji.201344347] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 01/14/2015] [Accepted: 01/29/2015] [Indexed: 11/06/2022]
Affiliation(s)
- Lawrence Han Boon Khoo
- Department of Microbiology; Immunology Programme; National University of Singapore; Singapore
- Singapore Immunology Network; Agency for Science; Technology and Research; Biopolis Singapore
| | - Chung Hwee Thiam
- Department of Microbiology; Immunology Programme; National University of Singapore; Singapore
| | - Serena Ying Soh
- Department of Microbiology; Immunology Programme; National University of Singapore; Singapore
| | - Véronique Angeli
- Department of Microbiology; Immunology Programme; National University of Singapore; Singapore
| |
Collapse
|
227
|
Ammirati E, Moroni F, Magnoni M, Camici PG. The role of T and B cells in human atherosclerosis and atherothrombosis. Clin Exp Immunol 2015; 179:173-87. [PMID: 25352024 DOI: 10.1111/cei.12477] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2014] [Indexed: 01/05/2023] Open
Abstract
Far from being merely a passive cholesterol accumulation within the arterial wall, the development of atherosclerosis is currently known to imply both inflammation and immune effector mechanisms. Adaptive immunity has been implicated in the process of disease initiation and progression interwined with traditional cardiovascular risk factors. Although the body of knowledge regarding the correlation between atherosclerosis and immunity in humans is growing rapidly, a relevant proportion of it derives from studies carried out in animal models of cardiovascular disease (CVD). However, while the mouse is a well-suited model, the results obtained therein are not fully transferrable to the human setting due to intrinsic genomic and environmental differences. In the present review, we will discuss mainly human findings, obtained either by examination of post-mortem and surgical atherosclerotic material or through the analysis of the immunological profile of peripheral blood cells. In particular, we will discuss the findings supporting a pro-atherogenic role of T cell subsets, such as effector memory T cells or the potential protective function of regulatory T cells. Recent studies suggest that traditional T cell-driven B2 cell responses appear to be atherogenic, while innate B1 cells appear to exert a protective action through the secretion of naturally occurring antibodies. The insights into the immune pathogenesis of atherosclerosis can provide new targets in the quest for novel therapeutic targets to abate CVD morbidity and mortality.
Collapse
Affiliation(s)
- E Ammirati
- Cardiothoracic Department, San Raffaele Scientific Institute and Vita-Salute University, Milan, Italy; Cardiovascular and Thoracic Department, AO Niguarda Ca' Granda, Milan, Italy
| | | | | | | |
Collapse
|
228
|
Clement M, Guedj K, Andreata F, Morvan M, Bey L, Khallou-Laschet J, Gaston AT, Delbosc S, Alsac JM, Bruneval P, Deschildre C, Le Borgne M, Castier Y, Kim HJ, Cantor H, Michel JB, Caligiuri G, Nicoletti A. Control of the T Follicular Helper–Germinal Center B-Cell Axis by CD8
+
Regulatory T Cells Limits Atherosclerosis and Tertiary Lymphoid Organ Development. Circulation 2015; 131:560-70. [PMID: 25552357 DOI: 10.1161/circulationaha.114.010988] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
The atheromodulating activity of B cells during the development of atherosclerosis is well documented, but the mechanisms by which these cells are regulated have not been investigated.
Methods and Results—
Here, we analyzed the contribution of Qa-1–restricted CD8
+
regulatory T cells to the control of the T follicular helper–germinal center B-cell axis during atherogenesis. Genetic disruption of CD8
+
regulatory T cell function in atherosclerosis-prone apolipoprotein E knockout mice resulted in overactivation of this axis in secondary lymphoid organs, led to the increased development of tertiary lymphoid organs in the aorta, and enhanced disease development. In contrast, restoring control of the T follicular helper–germinal center B-cell axis by blocking the ICOS-ICOSL pathway reduced the development of atherosclerosis and the formation of tertiary lymphoid organs. Moreover, analyses of human atherosclerotic aneurysmal arteries by flow cytometry, gene expression analysis, and immunofluorescence confirmed the presence of T follicular helper cells within tertiary lymphoid organs.
Conclusions—
This study is the first to demonstrate that the T follicular helper–germinal center B-cell axis is proatherogenic and that CD8
+
regulatory T cells control the germinal center reaction in both secondary and tertiary lymphoid organs. Therefore, disrupting this axis represents an innovative therapeutic approach.
Collapse
Affiliation(s)
- Marc Clement
- From Unité 1148, Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital X Bichat, Paris, France (M.C., K.G., F.A., M.M., J.-.K.L., A.-T.G., S.D., C.D., M.L.B., Y.C., J.-B.M., G.C., A.N.); Université Denis Diderot, Paris VII, Paris, France (M.C., K.G., F.A., L.B., J.-K.L., M.L.B., A.N.); Hôpital Européen Georges Pompidou, AP-HP, Faculté de Médecine René Descartes, Université Paris 5, Paris, France (J.-M.A., P.B.); and Department of Pathology, Harvard Medical School, Boston, MA (H.-J.K., H.C.)
| | - Kevin Guedj
- From Unité 1148, Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital X Bichat, Paris, France (M.C., K.G., F.A., M.M., J.-.K.L., A.-T.G., S.D., C.D., M.L.B., Y.C., J.-B.M., G.C., A.N.); Université Denis Diderot, Paris VII, Paris, France (M.C., K.G., F.A., L.B., J.-K.L., M.L.B., A.N.); Hôpital Européen Georges Pompidou, AP-HP, Faculté de Médecine René Descartes, Université Paris 5, Paris, France (J.-M.A., P.B.); and Department of Pathology, Harvard Medical School, Boston, MA (H.-J.K., H.C.)
| | - Francesco Andreata
- From Unité 1148, Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital X Bichat, Paris, France (M.C., K.G., F.A., M.M., J.-.K.L., A.-T.G., S.D., C.D., M.L.B., Y.C., J.-B.M., G.C., A.N.); Université Denis Diderot, Paris VII, Paris, France (M.C., K.G., F.A., L.B., J.-K.L., M.L.B., A.N.); Hôpital Européen Georges Pompidou, AP-HP, Faculté de Médecine René Descartes, Université Paris 5, Paris, France (J.-M.A., P.B.); and Department of Pathology, Harvard Medical School, Boston, MA (H.-J.K., H.C.)
| | - Marion Morvan
- From Unité 1148, Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital X Bichat, Paris, France (M.C., K.G., F.A., M.M., J.-.K.L., A.-T.G., S.D., C.D., M.L.B., Y.C., J.-B.M., G.C., A.N.); Université Denis Diderot, Paris VII, Paris, France (M.C., K.G., F.A., L.B., J.-K.L., M.L.B., A.N.); Hôpital Européen Georges Pompidou, AP-HP, Faculté de Médecine René Descartes, Université Paris 5, Paris, France (J.-M.A., P.B.); and Department of Pathology, Harvard Medical School, Boston, MA (H.-J.K., H.C.)
| | - Laetitia Bey
- From Unité 1148, Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital X Bichat, Paris, France (M.C., K.G., F.A., M.M., J.-.K.L., A.-T.G., S.D., C.D., M.L.B., Y.C., J.-B.M., G.C., A.N.); Université Denis Diderot, Paris VII, Paris, France (M.C., K.G., F.A., L.B., J.-K.L., M.L.B., A.N.); Hôpital Européen Georges Pompidou, AP-HP, Faculté de Médecine René Descartes, Université Paris 5, Paris, France (J.-M.A., P.B.); and Department of Pathology, Harvard Medical School, Boston, MA (H.-J.K., H.C.)
| | - Jamila Khallou-Laschet
- From Unité 1148, Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital X Bichat, Paris, France (M.C., K.G., F.A., M.M., J.-.K.L., A.-T.G., S.D., C.D., M.L.B., Y.C., J.-B.M., G.C., A.N.); Université Denis Diderot, Paris VII, Paris, France (M.C., K.G., F.A., L.B., J.-K.L., M.L.B., A.N.); Hôpital Européen Georges Pompidou, AP-HP, Faculté de Médecine René Descartes, Université Paris 5, Paris, France (J.-M.A., P.B.); and Department of Pathology, Harvard Medical School, Boston, MA (H.-J.K., H.C.)
| | - Anh-Thu Gaston
- From Unité 1148, Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital X Bichat, Paris, France (M.C., K.G., F.A., M.M., J.-.K.L., A.-T.G., S.D., C.D., M.L.B., Y.C., J.-B.M., G.C., A.N.); Université Denis Diderot, Paris VII, Paris, France (M.C., K.G., F.A., L.B., J.-K.L., M.L.B., A.N.); Hôpital Européen Georges Pompidou, AP-HP, Faculté de Médecine René Descartes, Université Paris 5, Paris, France (J.-M.A., P.B.); and Department of Pathology, Harvard Medical School, Boston, MA (H.-J.K., H.C.)
| | - Sandrine Delbosc
- From Unité 1148, Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital X Bichat, Paris, France (M.C., K.G., F.A., M.M., J.-.K.L., A.-T.G., S.D., C.D., M.L.B., Y.C., J.-B.M., G.C., A.N.); Université Denis Diderot, Paris VII, Paris, France (M.C., K.G., F.A., L.B., J.-K.L., M.L.B., A.N.); Hôpital Européen Georges Pompidou, AP-HP, Faculté de Médecine René Descartes, Université Paris 5, Paris, France (J.-M.A., P.B.); and Department of Pathology, Harvard Medical School, Boston, MA (H.-J.K., H.C.)
| | - Jean-Marc Alsac
- From Unité 1148, Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital X Bichat, Paris, France (M.C., K.G., F.A., M.M., J.-.K.L., A.-T.G., S.D., C.D., M.L.B., Y.C., J.-B.M., G.C., A.N.); Université Denis Diderot, Paris VII, Paris, France (M.C., K.G., F.A., L.B., J.-K.L., M.L.B., A.N.); Hôpital Européen Georges Pompidou, AP-HP, Faculté de Médecine René Descartes, Université Paris 5, Paris, France (J.-M.A., P.B.); and Department of Pathology, Harvard Medical School, Boston, MA (H.-J.K., H.C.)
| | - Patrick Bruneval
- From Unité 1148, Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital X Bichat, Paris, France (M.C., K.G., F.A., M.M., J.-.K.L., A.-T.G., S.D., C.D., M.L.B., Y.C., J.-B.M., G.C., A.N.); Université Denis Diderot, Paris VII, Paris, France (M.C., K.G., F.A., L.B., J.-K.L., M.L.B., A.N.); Hôpital Européen Georges Pompidou, AP-HP, Faculté de Médecine René Descartes, Université Paris 5, Paris, France (J.-M.A., P.B.); and Department of Pathology, Harvard Medical School, Boston, MA (H.-J.K., H.C.)
| | - Catherine Deschildre
- From Unité 1148, Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital X Bichat, Paris, France (M.C., K.G., F.A., M.M., J.-.K.L., A.-T.G., S.D., C.D., M.L.B., Y.C., J.-B.M., G.C., A.N.); Université Denis Diderot, Paris VII, Paris, France (M.C., K.G., F.A., L.B., J.-K.L., M.L.B., A.N.); Hôpital Européen Georges Pompidou, AP-HP, Faculté de Médecine René Descartes, Université Paris 5, Paris, France (J.-M.A., P.B.); and Department of Pathology, Harvard Medical School, Boston, MA (H.-J.K., H.C.)
| | - Marie Le Borgne
- From Unité 1148, Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital X Bichat, Paris, France (M.C., K.G., F.A., M.M., J.-.K.L., A.-T.G., S.D., C.D., M.L.B., Y.C., J.-B.M., G.C., A.N.); Université Denis Diderot, Paris VII, Paris, France (M.C., K.G., F.A., L.B., J.-K.L., M.L.B., A.N.); Hôpital Européen Georges Pompidou, AP-HP, Faculté de Médecine René Descartes, Université Paris 5, Paris, France (J.-M.A., P.B.); and Department of Pathology, Harvard Medical School, Boston, MA (H.-J.K., H.C.)
| | - Yves Castier
- From Unité 1148, Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital X Bichat, Paris, France (M.C., K.G., F.A., M.M., J.-.K.L., A.-T.G., S.D., C.D., M.L.B., Y.C., J.-B.M., G.C., A.N.); Université Denis Diderot, Paris VII, Paris, France (M.C., K.G., F.A., L.B., J.-K.L., M.L.B., A.N.); Hôpital Européen Georges Pompidou, AP-HP, Faculté de Médecine René Descartes, Université Paris 5, Paris, France (J.-M.A., P.B.); and Department of Pathology, Harvard Medical School, Boston, MA (H.-J.K., H.C.)
| | - Hye-Jung Kim
- From Unité 1148, Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital X Bichat, Paris, France (M.C., K.G., F.A., M.M., J.-.K.L., A.-T.G., S.D., C.D., M.L.B., Y.C., J.-B.M., G.C., A.N.); Université Denis Diderot, Paris VII, Paris, France (M.C., K.G., F.A., L.B., J.-K.L., M.L.B., A.N.); Hôpital Européen Georges Pompidou, AP-HP, Faculté de Médecine René Descartes, Université Paris 5, Paris, France (J.-M.A., P.B.); and Department of Pathology, Harvard Medical School, Boston, MA (H.-J.K., H.C.)
| | - Harvey Cantor
- From Unité 1148, Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital X Bichat, Paris, France (M.C., K.G., F.A., M.M., J.-.K.L., A.-T.G., S.D., C.D., M.L.B., Y.C., J.-B.M., G.C., A.N.); Université Denis Diderot, Paris VII, Paris, France (M.C., K.G., F.A., L.B., J.-K.L., M.L.B., A.N.); Hôpital Européen Georges Pompidou, AP-HP, Faculté de Médecine René Descartes, Université Paris 5, Paris, France (J.-M.A., P.B.); and Department of Pathology, Harvard Medical School, Boston, MA (H.-J.K., H.C.)
| | - Jean-Baptiste Michel
- From Unité 1148, Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital X Bichat, Paris, France (M.C., K.G., F.A., M.M., J.-.K.L., A.-T.G., S.D., C.D., M.L.B., Y.C., J.-B.M., G.C., A.N.); Université Denis Diderot, Paris VII, Paris, France (M.C., K.G., F.A., L.B., J.-K.L., M.L.B., A.N.); Hôpital Européen Georges Pompidou, AP-HP, Faculté de Médecine René Descartes, Université Paris 5, Paris, France (J.-M.A., P.B.); and Department of Pathology, Harvard Medical School, Boston, MA (H.-J.K., H.C.)
| | - Giuseppina Caligiuri
- From Unité 1148, Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital X Bichat, Paris, France (M.C., K.G., F.A., M.M., J.-.K.L., A.-T.G., S.D., C.D., M.L.B., Y.C., J.-B.M., G.C., A.N.); Université Denis Diderot, Paris VII, Paris, France (M.C., K.G., F.A., L.B., J.-K.L., M.L.B., A.N.); Hôpital Européen Georges Pompidou, AP-HP, Faculté de Médecine René Descartes, Université Paris 5, Paris, France (J.-M.A., P.B.); and Department of Pathology, Harvard Medical School, Boston, MA (H.-J.K., H.C.)
| | - Antonino Nicoletti
- From Unité 1148, Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital X Bichat, Paris, France (M.C., K.G., F.A., M.M., J.-.K.L., A.-T.G., S.D., C.D., M.L.B., Y.C., J.-B.M., G.C., A.N.); Université Denis Diderot, Paris VII, Paris, France (M.C., K.G., F.A., L.B., J.-K.L., M.L.B., A.N.); Hôpital Européen Georges Pompidou, AP-HP, Faculté de Médecine René Descartes, Université Paris 5, Paris, France (J.-M.A., P.B.); and Department of Pathology, Harvard Medical School, Boston, MA (H.-J.K., H.C.).
| |
Collapse
|
229
|
Hansson GK. How to repeat a success and control a bad influence. Circulation 2015; 131:525-7. [PMID: 25552358 DOI: 10.1161/circulationaha.114.014560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Göran K Hansson
- From Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
230
|
Huan T, Rong J, Tanriverdi K, Meng Q, Bhattacharya A, McManus DD, Joehanes R, Assimes TL, McPherson R, Samani NJ, Erdmann J, Schunkert H, Courchesne P, Munson PJ, Johnson AD, O'Donnell CJ, Zhang B, Larson MG, Freedman JE, Levy D, Yang X. Dissecting the roles of microRNAs in coronary heart disease via integrative genomic analyses. Arterioscler Thromb Vasc Biol 2015; 35:1011-21. [PMID: 25657313 DOI: 10.1161/atvbaha.114.305176] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The roles of microRNAs (miRNAs) in coronary heart disease (CHD) have not been well characterized. This study sought to systematically characterize the complex genomic architecture of CHD by integrating whole blood miRNA and mRNA expression with genetic variation in 186 CHD cases and 186 controls. APPROACH AND RESULTS At false discovery rate <0.2, 15 miRNAs were differentially expressed between CHD cases and controls. To explore regulatory mechanisms, we integrated miRNA and mRNA expression with genome-wide genotype data to investigate miRNA and mRNA associations and relationships of genetic variation with miRNAs. We identified a large number of correlated miRNA-mRNA pairs and genetic loci that seem to regulate miRNA levels. Subsequently, we explored the relationships of these complex molecular associations with CHD status. We identified a large difference in miRNA-mRNA associations between CHD cases and controls, as demonstrated by a significantly higher proportion of inversely correlated miRNA-mRNA pairs in cases versus controls (80% versus 30%; P<1×10(-16)), suggesting a genome-wide shift in the regulatory structure of the transcriptome in CHD. The differentially coexpressed miRNA-mRNA pairs showed enrichment for CHD risk genetic variants affecting both miRNA and mRNA expression levels, implicating a putatively causal role in CHD. Furthermore, 3 miRNAs (miR-1275, miR-365a-3p, and miR-150-5p) were associated with an mRNA coexpression module that was causally linked to CHD and reflected the dysregulation of B-cell centered immune function. CONCLUSIONS Our results provide novel evidence that miRNAs are important regulators of biological processes involved in CHD via genetic control and via their tight coexpression with mRNAs.
Collapse
Affiliation(s)
- Tianxiao Huan
- From the National Heart, Lung, and Blood Institute's Framingham Heart Study, MA (T.H., R.J., P.C., A.D.J., C.J.O., D.L.); The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (T.H., R.J., P.C., D.L.); Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (A.D.J., C.J.O.); Department of Mathematics and Statistics, Boston University, MA (J.R., M.G.L.); Department of Medicine, University of Massachusetts Medical School, Worcester (K.T., D.D.M., J.E.F.); Department of Integrative Biology and Physiology, University of California, Los Angeles (Q.M., A.B., X.Y.); Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD (R.J., P.J.M); Department of Medicine, Harvard Medical School, Harvard University, Boston, MA (R.J.); Department of Medicine, Stanford University School of Medicine, Palo Alto, CA (T.L.A.); Departments of Medicine and Biochemistry, University of Ottawa, Ottawa, Ontario, Canada (R.M.); Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom (N.J.S.); National Institute for Health Research (NIHR) Leicester Cardiovascular Biomedical Research Unit, Leicester, United Kingdom; Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany (J.E.); DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (J.E.); Deutsches Herzzentrum München, Technische Universität München, München, Germany (H.S.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S.); and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (B.Z.)
| | - Jian Rong
- From the National Heart, Lung, and Blood Institute's Framingham Heart Study, MA (T.H., R.J., P.C., A.D.J., C.J.O., D.L.); The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (T.H., R.J., P.C., D.L.); Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (A.D.J., C.J.O.); Department of Mathematics and Statistics, Boston University, MA (J.R., M.G.L.); Department of Medicine, University of Massachusetts Medical School, Worcester (K.T., D.D.M., J.E.F.); Department of Integrative Biology and Physiology, University of California, Los Angeles (Q.M., A.B., X.Y.); Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD (R.J., P.J.M); Department of Medicine, Harvard Medical School, Harvard University, Boston, MA (R.J.); Department of Medicine, Stanford University School of Medicine, Palo Alto, CA (T.L.A.); Departments of Medicine and Biochemistry, University of Ottawa, Ottawa, Ontario, Canada (R.M.); Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom (N.J.S.); National Institute for Health Research (NIHR) Leicester Cardiovascular Biomedical Research Unit, Leicester, United Kingdom; Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany (J.E.); DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (J.E.); Deutsches Herzzentrum München, Technische Universität München, München, Germany (H.S.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S.); and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (B.Z.)
| | - Kahraman Tanriverdi
- From the National Heart, Lung, and Blood Institute's Framingham Heart Study, MA (T.H., R.J., P.C., A.D.J., C.J.O., D.L.); The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (T.H., R.J., P.C., D.L.); Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (A.D.J., C.J.O.); Department of Mathematics and Statistics, Boston University, MA (J.R., M.G.L.); Department of Medicine, University of Massachusetts Medical School, Worcester (K.T., D.D.M., J.E.F.); Department of Integrative Biology and Physiology, University of California, Los Angeles (Q.M., A.B., X.Y.); Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD (R.J., P.J.M); Department of Medicine, Harvard Medical School, Harvard University, Boston, MA (R.J.); Department of Medicine, Stanford University School of Medicine, Palo Alto, CA (T.L.A.); Departments of Medicine and Biochemistry, University of Ottawa, Ottawa, Ontario, Canada (R.M.); Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom (N.J.S.); National Institute for Health Research (NIHR) Leicester Cardiovascular Biomedical Research Unit, Leicester, United Kingdom; Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany (J.E.); DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (J.E.); Deutsches Herzzentrum München, Technische Universität München, München, Germany (H.S.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S.); and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (B.Z.)
| | - Qingying Meng
- From the National Heart, Lung, and Blood Institute's Framingham Heart Study, MA (T.H., R.J., P.C., A.D.J., C.J.O., D.L.); The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (T.H., R.J., P.C., D.L.); Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (A.D.J., C.J.O.); Department of Mathematics and Statistics, Boston University, MA (J.R., M.G.L.); Department of Medicine, University of Massachusetts Medical School, Worcester (K.T., D.D.M., J.E.F.); Department of Integrative Biology and Physiology, University of California, Los Angeles (Q.M., A.B., X.Y.); Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD (R.J., P.J.M); Department of Medicine, Harvard Medical School, Harvard University, Boston, MA (R.J.); Department of Medicine, Stanford University School of Medicine, Palo Alto, CA (T.L.A.); Departments of Medicine and Biochemistry, University of Ottawa, Ottawa, Ontario, Canada (R.M.); Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom (N.J.S.); National Institute for Health Research (NIHR) Leicester Cardiovascular Biomedical Research Unit, Leicester, United Kingdom; Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany (J.E.); DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (J.E.); Deutsches Herzzentrum München, Technische Universität München, München, Germany (H.S.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S.); and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (B.Z.)
| | - Anindya Bhattacharya
- From the National Heart, Lung, and Blood Institute's Framingham Heart Study, MA (T.H., R.J., P.C., A.D.J., C.J.O., D.L.); The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (T.H., R.J., P.C., D.L.); Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (A.D.J., C.J.O.); Department of Mathematics and Statistics, Boston University, MA (J.R., M.G.L.); Department of Medicine, University of Massachusetts Medical School, Worcester (K.T., D.D.M., J.E.F.); Department of Integrative Biology and Physiology, University of California, Los Angeles (Q.M., A.B., X.Y.); Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD (R.J., P.J.M); Department of Medicine, Harvard Medical School, Harvard University, Boston, MA (R.J.); Department of Medicine, Stanford University School of Medicine, Palo Alto, CA (T.L.A.); Departments of Medicine and Biochemistry, University of Ottawa, Ottawa, Ontario, Canada (R.M.); Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom (N.J.S.); National Institute for Health Research (NIHR) Leicester Cardiovascular Biomedical Research Unit, Leicester, United Kingdom; Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany (J.E.); DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (J.E.); Deutsches Herzzentrum München, Technische Universität München, München, Germany (H.S.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S.); and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (B.Z.)
| | - David D McManus
- From the National Heart, Lung, and Blood Institute's Framingham Heart Study, MA (T.H., R.J., P.C., A.D.J., C.J.O., D.L.); The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (T.H., R.J., P.C., D.L.); Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (A.D.J., C.J.O.); Department of Mathematics and Statistics, Boston University, MA (J.R., M.G.L.); Department of Medicine, University of Massachusetts Medical School, Worcester (K.T., D.D.M., J.E.F.); Department of Integrative Biology and Physiology, University of California, Los Angeles (Q.M., A.B., X.Y.); Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD (R.J., P.J.M); Department of Medicine, Harvard Medical School, Harvard University, Boston, MA (R.J.); Department of Medicine, Stanford University School of Medicine, Palo Alto, CA (T.L.A.); Departments of Medicine and Biochemistry, University of Ottawa, Ottawa, Ontario, Canada (R.M.); Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom (N.J.S.); National Institute for Health Research (NIHR) Leicester Cardiovascular Biomedical Research Unit, Leicester, United Kingdom; Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany (J.E.); DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (J.E.); Deutsches Herzzentrum München, Technische Universität München, München, Germany (H.S.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S.); and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (B.Z.)
| | - Roby Joehanes
- From the National Heart, Lung, and Blood Institute's Framingham Heart Study, MA (T.H., R.J., P.C., A.D.J., C.J.O., D.L.); The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (T.H., R.J., P.C., D.L.); Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (A.D.J., C.J.O.); Department of Mathematics and Statistics, Boston University, MA (J.R., M.G.L.); Department of Medicine, University of Massachusetts Medical School, Worcester (K.T., D.D.M., J.E.F.); Department of Integrative Biology and Physiology, University of California, Los Angeles (Q.M., A.B., X.Y.); Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD (R.J., P.J.M); Department of Medicine, Harvard Medical School, Harvard University, Boston, MA (R.J.); Department of Medicine, Stanford University School of Medicine, Palo Alto, CA (T.L.A.); Departments of Medicine and Biochemistry, University of Ottawa, Ottawa, Ontario, Canada (R.M.); Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom (N.J.S.); National Institute for Health Research (NIHR) Leicester Cardiovascular Biomedical Research Unit, Leicester, United Kingdom; Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany (J.E.); DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (J.E.); Deutsches Herzzentrum München, Technische Universität München, München, Germany (H.S.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S.); and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (B.Z.)
| | - Themistocles L Assimes
- From the National Heart, Lung, and Blood Institute's Framingham Heart Study, MA (T.H., R.J., P.C., A.D.J., C.J.O., D.L.); The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (T.H., R.J., P.C., D.L.); Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (A.D.J., C.J.O.); Department of Mathematics and Statistics, Boston University, MA (J.R., M.G.L.); Department of Medicine, University of Massachusetts Medical School, Worcester (K.T., D.D.M., J.E.F.); Department of Integrative Biology and Physiology, University of California, Los Angeles (Q.M., A.B., X.Y.); Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD (R.J., P.J.M); Department of Medicine, Harvard Medical School, Harvard University, Boston, MA (R.J.); Department of Medicine, Stanford University School of Medicine, Palo Alto, CA (T.L.A.); Departments of Medicine and Biochemistry, University of Ottawa, Ottawa, Ontario, Canada (R.M.); Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom (N.J.S.); National Institute for Health Research (NIHR) Leicester Cardiovascular Biomedical Research Unit, Leicester, United Kingdom; Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany (J.E.); DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (J.E.); Deutsches Herzzentrum München, Technische Universität München, München, Germany (H.S.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S.); and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (B.Z.)
| | - Ruth McPherson
- From the National Heart, Lung, and Blood Institute's Framingham Heart Study, MA (T.H., R.J., P.C., A.D.J., C.J.O., D.L.); The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (T.H., R.J., P.C., D.L.); Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (A.D.J., C.J.O.); Department of Mathematics and Statistics, Boston University, MA (J.R., M.G.L.); Department of Medicine, University of Massachusetts Medical School, Worcester (K.T., D.D.M., J.E.F.); Department of Integrative Biology and Physiology, University of California, Los Angeles (Q.M., A.B., X.Y.); Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD (R.J., P.J.M); Department of Medicine, Harvard Medical School, Harvard University, Boston, MA (R.J.); Department of Medicine, Stanford University School of Medicine, Palo Alto, CA (T.L.A.); Departments of Medicine and Biochemistry, University of Ottawa, Ottawa, Ontario, Canada (R.M.); Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom (N.J.S.); National Institute for Health Research (NIHR) Leicester Cardiovascular Biomedical Research Unit, Leicester, United Kingdom; Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany (J.E.); DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (J.E.); Deutsches Herzzentrum München, Technische Universität München, München, Germany (H.S.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S.); and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (B.Z.)
| | - Nilesh J Samani
- From the National Heart, Lung, and Blood Institute's Framingham Heart Study, MA (T.H., R.J., P.C., A.D.J., C.J.O., D.L.); The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (T.H., R.J., P.C., D.L.); Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (A.D.J., C.J.O.); Department of Mathematics and Statistics, Boston University, MA (J.R., M.G.L.); Department of Medicine, University of Massachusetts Medical School, Worcester (K.T., D.D.M., J.E.F.); Department of Integrative Biology and Physiology, University of California, Los Angeles (Q.M., A.B., X.Y.); Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD (R.J., P.J.M); Department of Medicine, Harvard Medical School, Harvard University, Boston, MA (R.J.); Department of Medicine, Stanford University School of Medicine, Palo Alto, CA (T.L.A.); Departments of Medicine and Biochemistry, University of Ottawa, Ottawa, Ontario, Canada (R.M.); Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom (N.J.S.); National Institute for Health Research (NIHR) Leicester Cardiovascular Biomedical Research Unit, Leicester, United Kingdom; Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany (J.E.); DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (J.E.); Deutsches Herzzentrum München, Technische Universität München, München, Germany (H.S.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S.); and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (B.Z.)
| | - Jeanette Erdmann
- From the National Heart, Lung, and Blood Institute's Framingham Heart Study, MA (T.H., R.J., P.C., A.D.J., C.J.O., D.L.); The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (T.H., R.J., P.C., D.L.); Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (A.D.J., C.J.O.); Department of Mathematics and Statistics, Boston University, MA (J.R., M.G.L.); Department of Medicine, University of Massachusetts Medical School, Worcester (K.T., D.D.M., J.E.F.); Department of Integrative Biology and Physiology, University of California, Los Angeles (Q.M., A.B., X.Y.); Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD (R.J., P.J.M); Department of Medicine, Harvard Medical School, Harvard University, Boston, MA (R.J.); Department of Medicine, Stanford University School of Medicine, Palo Alto, CA (T.L.A.); Departments of Medicine and Biochemistry, University of Ottawa, Ottawa, Ontario, Canada (R.M.); Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom (N.J.S.); National Institute for Health Research (NIHR) Leicester Cardiovascular Biomedical Research Unit, Leicester, United Kingdom; Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany (J.E.); DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (J.E.); Deutsches Herzzentrum München, Technische Universität München, München, Germany (H.S.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S.); and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (B.Z.)
| | - Heribert Schunkert
- From the National Heart, Lung, and Blood Institute's Framingham Heart Study, MA (T.H., R.J., P.C., A.D.J., C.J.O., D.L.); The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (T.H., R.J., P.C., D.L.); Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (A.D.J., C.J.O.); Department of Mathematics and Statistics, Boston University, MA (J.R., M.G.L.); Department of Medicine, University of Massachusetts Medical School, Worcester (K.T., D.D.M., J.E.F.); Department of Integrative Biology and Physiology, University of California, Los Angeles (Q.M., A.B., X.Y.); Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD (R.J., P.J.M); Department of Medicine, Harvard Medical School, Harvard University, Boston, MA (R.J.); Department of Medicine, Stanford University School of Medicine, Palo Alto, CA (T.L.A.); Departments of Medicine and Biochemistry, University of Ottawa, Ottawa, Ontario, Canada (R.M.); Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom (N.J.S.); National Institute for Health Research (NIHR) Leicester Cardiovascular Biomedical Research Unit, Leicester, United Kingdom; Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany (J.E.); DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (J.E.); Deutsches Herzzentrum München, Technische Universität München, München, Germany (H.S.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S.); and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (B.Z.)
| | - Paul Courchesne
- From the National Heart, Lung, and Blood Institute's Framingham Heart Study, MA (T.H., R.J., P.C., A.D.J., C.J.O., D.L.); The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (T.H., R.J., P.C., D.L.); Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (A.D.J., C.J.O.); Department of Mathematics and Statistics, Boston University, MA (J.R., M.G.L.); Department of Medicine, University of Massachusetts Medical School, Worcester (K.T., D.D.M., J.E.F.); Department of Integrative Biology and Physiology, University of California, Los Angeles (Q.M., A.B., X.Y.); Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD (R.J., P.J.M); Department of Medicine, Harvard Medical School, Harvard University, Boston, MA (R.J.); Department of Medicine, Stanford University School of Medicine, Palo Alto, CA (T.L.A.); Departments of Medicine and Biochemistry, University of Ottawa, Ottawa, Ontario, Canada (R.M.); Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom (N.J.S.); National Institute for Health Research (NIHR) Leicester Cardiovascular Biomedical Research Unit, Leicester, United Kingdom; Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany (J.E.); DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (J.E.); Deutsches Herzzentrum München, Technische Universität München, München, Germany (H.S.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S.); and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (B.Z.)
| | - Peter J Munson
- From the National Heart, Lung, and Blood Institute's Framingham Heart Study, MA (T.H., R.J., P.C., A.D.J., C.J.O., D.L.); The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (T.H., R.J., P.C., D.L.); Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (A.D.J., C.J.O.); Department of Mathematics and Statistics, Boston University, MA (J.R., M.G.L.); Department of Medicine, University of Massachusetts Medical School, Worcester (K.T., D.D.M., J.E.F.); Department of Integrative Biology and Physiology, University of California, Los Angeles (Q.M., A.B., X.Y.); Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD (R.J., P.J.M); Department of Medicine, Harvard Medical School, Harvard University, Boston, MA (R.J.); Department of Medicine, Stanford University School of Medicine, Palo Alto, CA (T.L.A.); Departments of Medicine and Biochemistry, University of Ottawa, Ottawa, Ontario, Canada (R.M.); Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom (N.J.S.); National Institute for Health Research (NIHR) Leicester Cardiovascular Biomedical Research Unit, Leicester, United Kingdom; Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany (J.E.); DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (J.E.); Deutsches Herzzentrum München, Technische Universität München, München, Germany (H.S.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S.); and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (B.Z.)
| | - Andrew D Johnson
- From the National Heart, Lung, and Blood Institute's Framingham Heart Study, MA (T.H., R.J., P.C., A.D.J., C.J.O., D.L.); The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (T.H., R.J., P.C., D.L.); Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (A.D.J., C.J.O.); Department of Mathematics and Statistics, Boston University, MA (J.R., M.G.L.); Department of Medicine, University of Massachusetts Medical School, Worcester (K.T., D.D.M., J.E.F.); Department of Integrative Biology and Physiology, University of California, Los Angeles (Q.M., A.B., X.Y.); Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD (R.J., P.J.M); Department of Medicine, Harvard Medical School, Harvard University, Boston, MA (R.J.); Department of Medicine, Stanford University School of Medicine, Palo Alto, CA (T.L.A.); Departments of Medicine and Biochemistry, University of Ottawa, Ottawa, Ontario, Canada (R.M.); Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom (N.J.S.); National Institute for Health Research (NIHR) Leicester Cardiovascular Biomedical Research Unit, Leicester, United Kingdom; Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany (J.E.); DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (J.E.); Deutsches Herzzentrum München, Technische Universität München, München, Germany (H.S.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S.); and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (B.Z.)
| | - Christopher J O'Donnell
- From the National Heart, Lung, and Blood Institute's Framingham Heart Study, MA (T.H., R.J., P.C., A.D.J., C.J.O., D.L.); The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (T.H., R.J., P.C., D.L.); Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (A.D.J., C.J.O.); Department of Mathematics and Statistics, Boston University, MA (J.R., M.G.L.); Department of Medicine, University of Massachusetts Medical School, Worcester (K.T., D.D.M., J.E.F.); Department of Integrative Biology and Physiology, University of California, Los Angeles (Q.M., A.B., X.Y.); Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD (R.J., P.J.M); Department of Medicine, Harvard Medical School, Harvard University, Boston, MA (R.J.); Department of Medicine, Stanford University School of Medicine, Palo Alto, CA (T.L.A.); Departments of Medicine and Biochemistry, University of Ottawa, Ottawa, Ontario, Canada (R.M.); Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom (N.J.S.); National Institute for Health Research (NIHR) Leicester Cardiovascular Biomedical Research Unit, Leicester, United Kingdom; Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany (J.E.); DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (J.E.); Deutsches Herzzentrum München, Technische Universität München, München, Germany (H.S.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S.); and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (B.Z.)
| | - Bin Zhang
- From the National Heart, Lung, and Blood Institute's Framingham Heart Study, MA (T.H., R.J., P.C., A.D.J., C.J.O., D.L.); The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (T.H., R.J., P.C., D.L.); Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (A.D.J., C.J.O.); Department of Mathematics and Statistics, Boston University, MA (J.R., M.G.L.); Department of Medicine, University of Massachusetts Medical School, Worcester (K.T., D.D.M., J.E.F.); Department of Integrative Biology and Physiology, University of California, Los Angeles (Q.M., A.B., X.Y.); Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD (R.J., P.J.M); Department of Medicine, Harvard Medical School, Harvard University, Boston, MA (R.J.); Department of Medicine, Stanford University School of Medicine, Palo Alto, CA (T.L.A.); Departments of Medicine and Biochemistry, University of Ottawa, Ottawa, Ontario, Canada (R.M.); Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom (N.J.S.); National Institute for Health Research (NIHR) Leicester Cardiovascular Biomedical Research Unit, Leicester, United Kingdom; Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany (J.E.); DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (J.E.); Deutsches Herzzentrum München, Technische Universität München, München, Germany (H.S.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S.); and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (B.Z.)
| | - Martin G Larson
- From the National Heart, Lung, and Blood Institute's Framingham Heart Study, MA (T.H., R.J., P.C., A.D.J., C.J.O., D.L.); The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (T.H., R.J., P.C., D.L.); Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (A.D.J., C.J.O.); Department of Mathematics and Statistics, Boston University, MA (J.R., M.G.L.); Department of Medicine, University of Massachusetts Medical School, Worcester (K.T., D.D.M., J.E.F.); Department of Integrative Biology and Physiology, University of California, Los Angeles (Q.M., A.B., X.Y.); Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD (R.J., P.J.M); Department of Medicine, Harvard Medical School, Harvard University, Boston, MA (R.J.); Department of Medicine, Stanford University School of Medicine, Palo Alto, CA (T.L.A.); Departments of Medicine and Biochemistry, University of Ottawa, Ottawa, Ontario, Canada (R.M.); Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom (N.J.S.); National Institute for Health Research (NIHR) Leicester Cardiovascular Biomedical Research Unit, Leicester, United Kingdom; Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany (J.E.); DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (J.E.); Deutsches Herzzentrum München, Technische Universität München, München, Germany (H.S.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S.); and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (B.Z.)
| | - Jane E Freedman
- From the National Heart, Lung, and Blood Institute's Framingham Heart Study, MA (T.H., R.J., P.C., A.D.J., C.J.O., D.L.); The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (T.H., R.J., P.C., D.L.); Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (A.D.J., C.J.O.); Department of Mathematics and Statistics, Boston University, MA (J.R., M.G.L.); Department of Medicine, University of Massachusetts Medical School, Worcester (K.T., D.D.M., J.E.F.); Department of Integrative Biology and Physiology, University of California, Los Angeles (Q.M., A.B., X.Y.); Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD (R.J., P.J.M); Department of Medicine, Harvard Medical School, Harvard University, Boston, MA (R.J.); Department of Medicine, Stanford University School of Medicine, Palo Alto, CA (T.L.A.); Departments of Medicine and Biochemistry, University of Ottawa, Ottawa, Ontario, Canada (R.M.); Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom (N.J.S.); National Institute for Health Research (NIHR) Leicester Cardiovascular Biomedical Research Unit, Leicester, United Kingdom; Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany (J.E.); DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (J.E.); Deutsches Herzzentrum München, Technische Universität München, München, Germany (H.S.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S.); and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (B.Z.).
| | - Daniel Levy
- From the National Heart, Lung, and Blood Institute's Framingham Heart Study, MA (T.H., R.J., P.C., A.D.J., C.J.O., D.L.); The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (T.H., R.J., P.C., D.L.); Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (A.D.J., C.J.O.); Department of Mathematics and Statistics, Boston University, MA (J.R., M.G.L.); Department of Medicine, University of Massachusetts Medical School, Worcester (K.T., D.D.M., J.E.F.); Department of Integrative Biology and Physiology, University of California, Los Angeles (Q.M., A.B., X.Y.); Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD (R.J., P.J.M); Department of Medicine, Harvard Medical School, Harvard University, Boston, MA (R.J.); Department of Medicine, Stanford University School of Medicine, Palo Alto, CA (T.L.A.); Departments of Medicine and Biochemistry, University of Ottawa, Ottawa, Ontario, Canada (R.M.); Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom (N.J.S.); National Institute for Health Research (NIHR) Leicester Cardiovascular Biomedical Research Unit, Leicester, United Kingdom; Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany (J.E.); DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (J.E.); Deutsches Herzzentrum München, Technische Universität München, München, Germany (H.S.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S.); and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (B.Z.)
| | - Xia Yang
- From the National Heart, Lung, and Blood Institute's Framingham Heart Study, MA (T.H., R.J., P.C., A.D.J., C.J.O., D.L.); The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (T.H., R.J., P.C., D.L.); Cardiovascular Epidemiology and Human Genomics Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (A.D.J., C.J.O.); Department of Mathematics and Statistics, Boston University, MA (J.R., M.G.L.); Department of Medicine, University of Massachusetts Medical School, Worcester (K.T., D.D.M., J.E.F.); Department of Integrative Biology and Physiology, University of California, Los Angeles (Q.M., A.B., X.Y.); Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD (R.J., P.J.M); Department of Medicine, Harvard Medical School, Harvard University, Boston, MA (R.J.); Department of Medicine, Stanford University School of Medicine, Palo Alto, CA (T.L.A.); Departments of Medicine and Biochemistry, University of Ottawa, Ottawa, Ontario, Canada (R.M.); Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom (N.J.S.); National Institute for Health Research (NIHR) Leicester Cardiovascular Biomedical Research Unit, Leicester, United Kingdom; Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany (J.E.); DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany (J.E.); Deutsches Herzzentrum München, Technische Universität München, München, Germany (H.S.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S.); and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (B.Z.)
| |
Collapse
|
231
|
Role of vascular inflammation in coronary artery disease: potential of anti-inflammatory drugs in the prevention of atherothrombosis. Inflammation and anti-inflammatory drugs in coronary artery disease. Am J Cardiovasc Drugs 2015; 15:1-11. [PMID: 25369900 DOI: 10.1007/s40256-014-0094-z] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Coronary artery disease (CAD) and acute myocardial infarction (AMI) are inflammatory pathologies, involving interleukins (ILs), such as IL-1β, IL-6 and tumor necrosis factor (TNF)-α, and acute phase proteins production, such as for C reactive protein (CRP). The process begins with retention of low-density lipoprotein (LDL) and its oxidation inside the intima, with the formation of the "foam cells." Toll-like receptors and inflamassomes participate in atherosclerosis formation, as well as in the activation of the complement system. In addition to innate immunity, adaptive immunity is also associated with atherosclerosis through antigen-presenting cells, T and B lymphocytes. AMI also increases the expression of some ILs and promotes macrophage and lymphocyte accumulation. Reperfusion increases the expression of anti-inflammatory ILs (such as IL-10) and generates oxygen free radicals. Although CAD and AMI are inflammatory disorders, the only drugs with anti-inflammatory effect so far widely used in ischemic heart disease are aspirin and statins. Some immunomodulatory or immunosuppressive promising therapies, such as cyclosporine and colchicine, may have benefits in CAD. Methotrexate also has potential cardioprotective anti-inflammatory effects, through increased adenosine levels. The TETHYS trial (The Effects of mETHotrexate Therapy on ST Segment Elevation MYocardial InfarctionS trial) will evaluate low-dose methotrexate in ST elevation AMI. The CIRT (Cardiovascular Inflammation Reduction Trial), in turn, will evaluate low-dose methotrexate in patients with a high prevalence of subclinical vascular inflammation. The CANTOS (The Canakinumab Antiinflammatory Thrombosis Outcomes Study) will evaluate canakinumab in patients with CAD and persistently elevated CRP. The blockage of other potential targets, such as the IL-6 receptor, CC2 chemokine receptor and CD20, could bring benefits in CAD.
Collapse
|
232
|
Shaikh SR, Haas KM, Beck MA, Teague H. The effects of diet-induced obesity on B cell function. Clin Exp Immunol 2015; 179:90-9. [PMID: 25169121 DOI: 10.1111/cei.12444] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2014] [Indexed: 12/12/2022] Open
Abstract
B-1 and B-2 B cell subsets carry out a diverse array of functions that range broadly from responding to innate stimuli, antigen presentation, cytokine secretion and antibody production. In this review, we first cover the functional roles of the major murine B cell subsets. We then highlight emerging evidence, primarily in preclinical rodent studies, to show that select B cell subsets are a therapeutic target in obesity and its associated co-morbidities. High fat diets promote accumulation of select murine B cell phenotypes in visceral adipose tissue. As a consequence, B cells exacerbate inflammation and thereby insulin sensitivity through the production of autoantibodies and via cross-talk with select adipose resident macrophages, CD4(+) and CD8(+) T cells. In contrast, interleukin (IL)-10-secreting regulatory B cells counteract the proinflammatory profile and improve glucose sensitivity. We subsequently review data from rodent studies that show pharmacological supplementation of obesogenic diets with long chain n-3 polyunsaturated fatty acids or specialized pro-resolving lipid mediators synthesized from endogenous n-3 polyunsaturated fatty acids boost B cell activation and antibody production. This may have potential benefits for improving inflammation in addition to combating the increased risk of viral infection that is an associated complication of obesity and type II diabetes. Finally, we propose potential underlying mechanisms throughout the review by which B cell activity could be differentially regulated in response to high fat diets.
Collapse
Affiliation(s)
- S R Shaikh
- Department of Biochemistry and Molecular Biology, East Carolina Diabetes and Obesity Institute, East Carolina Heart Institute, Brody School of Medicine, East Carolina University, Greenville, NC, USA; Department of Microbiology and Immunology, East Carolina Diabetes and Obesity Institute, East Carolina Heart Institute, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | | | | | | |
Collapse
|
233
|
Mellak S, Ait-Oufella H, Esposito B, Loyer X, Poirier M, Tedder TF, Tedgui A, Mallat Z, Potteaux S. Angiotensin II mobilizes spleen monocytes to promote the development of abdominal aortic aneurysm in Apoe-/- mice. Arterioscler Thromb Vasc Biol 2014; 35:378-88. [PMID: 25524776 DOI: 10.1161/atvbaha.114.304389] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Abdominal aortic aneurysm (AAA) is widespread among elderly people and results in progressive expansion and rupture of the aorta with high mortality. Macrophages, which are the main population observed within the site of aneurysm, are thought to derive from circulating monocytes although no direct evidence has been provided to date. In this study, we were particularly interested in understanding the trafficking behavior of monocyte subsets in AAA and their role in disease pathogenesis. APPROACH AND RESULTS Using bone marrow transplantation in Apoe(-/-) mice, we showed that circulating monocytes give rise to abdominal aortic macrophages in hypercholesterolemic mice submitted to angiotensin II (AngII). Detailed monitoring of monocyte compartmentalization revealed that lymphocyte antigen 6C(high) and lymphocyte antigen 6C(low) monocytes transiently increase in blood early after AngII infusion and differentially infiltrate the abdominal aorta. The splenic reservoir accounted for the mobilization of the 2 monocyte subsets after 3 days of AngII infusion. Spleen removal or lymphocyte deficiency in Apoe(-/-) Rag2(-/-) mice similarly impaired early monocyte increase in blood in response to AngII and protected against AAA development, independently of blood pressure. Reconstitution of Apoe(-/-) Rag2(-/-) mice with total splenocytes but not with B-cell-depleted splenocytes restored monocyte mobilization in response to AngII and enhanced susceptibility to AAA. CONCLUSIONS Taken together, the data show that lymphocyte antigen 6C(high) and lymphocyte antigen 6C(low) monocytes are mobilized from the spleen in response to AngII. Intriguingly, the process is dependent on the presence of B cells and significantly contributes to the development of AAA and the occurrence of aortic rupture.
Collapse
Affiliation(s)
- Safa Mellak
- From the INSERM Unit UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (S.M., H.A.-O., B.E., X.L., M.P., A.T., Z.M., S.P.); Réanimation Médicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Université Pierre-et-Marie Curie, Université Pierre-et-Marie Curie, Paris, France (H.A.-O.); Department of Immunology, Duke University Medical Center, Durham, NC (T.F.T.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.)
| | - Hafid Ait-Oufella
- From the INSERM Unit UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (S.M., H.A.-O., B.E., X.L., M.P., A.T., Z.M., S.P.); Réanimation Médicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Université Pierre-et-Marie Curie, Université Pierre-et-Marie Curie, Paris, France (H.A.-O.); Department of Immunology, Duke University Medical Center, Durham, NC (T.F.T.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.)
| | - Bruno Esposito
- From the INSERM Unit UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (S.M., H.A.-O., B.E., X.L., M.P., A.T., Z.M., S.P.); Réanimation Médicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Université Pierre-et-Marie Curie, Université Pierre-et-Marie Curie, Paris, France (H.A.-O.); Department of Immunology, Duke University Medical Center, Durham, NC (T.F.T.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.)
| | - Xavier Loyer
- From the INSERM Unit UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (S.M., H.A.-O., B.E., X.L., M.P., A.T., Z.M., S.P.); Réanimation Médicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Université Pierre-et-Marie Curie, Université Pierre-et-Marie Curie, Paris, France (H.A.-O.); Department of Immunology, Duke University Medical Center, Durham, NC (T.F.T.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.)
| | - Maxime Poirier
- From the INSERM Unit UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (S.M., H.A.-O., B.E., X.L., M.P., A.T., Z.M., S.P.); Réanimation Médicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Université Pierre-et-Marie Curie, Université Pierre-et-Marie Curie, Paris, France (H.A.-O.); Department of Immunology, Duke University Medical Center, Durham, NC (T.F.T.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.)
| | - Thomas F Tedder
- From the INSERM Unit UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (S.M., H.A.-O., B.E., X.L., M.P., A.T., Z.M., S.P.); Réanimation Médicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Université Pierre-et-Marie Curie, Université Pierre-et-Marie Curie, Paris, France (H.A.-O.); Department of Immunology, Duke University Medical Center, Durham, NC (T.F.T.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.)
| | - Alain Tedgui
- From the INSERM Unit UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (S.M., H.A.-O., B.E., X.L., M.P., A.T., Z.M., S.P.); Réanimation Médicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Université Pierre-et-Marie Curie, Université Pierre-et-Marie Curie, Paris, France (H.A.-O.); Department of Immunology, Duke University Medical Center, Durham, NC (T.F.T.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.)
| | - Ziad Mallat
- From the INSERM Unit UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (S.M., H.A.-O., B.E., X.L., M.P., A.T., Z.M., S.P.); Réanimation Médicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Université Pierre-et-Marie Curie, Université Pierre-et-Marie Curie, Paris, France (H.A.-O.); Department of Immunology, Duke University Medical Center, Durham, NC (T.F.T.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.)
| | - Stéphane Potteaux
- From the INSERM Unit UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (S.M., H.A.-O., B.E., X.L., M.P., A.T., Z.M., S.P.); Réanimation Médicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Université Pierre-et-Marie Curie, Université Pierre-et-Marie Curie, Paris, France (H.A.-O.); Department of Immunology, Duke University Medical Center, Durham, NC (T.F.T.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.).
| |
Collapse
|
234
|
Shah PK, Chyu KY, Dimayuga PC, Nilsson J. Vaccine for Atherosclerosis. J Am Coll Cardiol 2014; 64:2779-91. [DOI: 10.1016/j.jacc.2014.10.018] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 10/08/2014] [Accepted: 10/10/2014] [Indexed: 11/25/2022]
|
235
|
Barsalou J, Bradley TJ, Silverman ED. Cardiovascular risk in pediatric-onset rheumatological diseases. Arthritis Res Ther 2014; 15:212. [PMID: 23731870 PMCID: PMC3672705 DOI: 10.1186/ar4212] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular morbidity and mortality are becoming major health concerns for adults with inflammatory rheumatic diseases. The enhanced atherogenesis in this patient population is promoted by the exposure to traditional risk factors as well as nontraditional cardiovascular insults, such as corticosteroid therapy, chronic inflammation and autoantibodies. Despite definite differences between many adult-onset and pediatric-onset rheumatologic diseases, it is extremely likely that atherosclerosis will become the leading cause of morbidity and mortality in this pediatric patient population. Because cardiovascular events are rare at this young age, surrogate measures of atherosclerosis must be used. The three major noninvasive vascular measures of early atherosclerosis--namely, flow-mediated dilatation, carotid intima-media thickness and pulse wave velocity--can be performed easily on children. Few studies have explored the prevalence of cardiovascular risk factors and even fewer have used the surrogate vascular measures to document signs of early atherosclerosis in children with pediatric-onset rheumatic diseases. The objective of this review is to provide an overview on cardiovascular risk and early atherosclerosis in pediatric-onset systemic lupus erythematosus, juvenile idiopathic arthritis and juvenile dermatomyositis patients, and to review cardiovascular preventive strategies that should be considered in this population.
Collapse
|
236
|
Li Y, To K, Kanellakis P, Hosseini H, Deswaerte V, Tipping P, Smyth MJ, Toh BH, Bobik A, Kyaw T. CD4+ natural killer T cells potently augment aortic root atherosclerosis by perforin- and granzyme B-dependent cytotoxicity. Circ Res 2014; 116:245-54. [PMID: 25398236 DOI: 10.1161/circresaha.116.304734] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE CD4(+) natural killer T (NKT) cells augment atherosclerosis in apolipoprotein E-deficient (ApoE)(-/-) mice but their mechanisms of action are unknown. OBJECTIVES We investigated the roles of bystander T, B, and NK cells; NKT cell-derived interferon-γ, interleukin (IL)-4, and IL-21 cytokines; and NKT cell-derived perforin and granzyme B cytotoxins in promoting CD4(+) NKT cell atherogenicity. METHODS AND RESULTS Transfer of CD4(+) NKT cells into T- and B-cell-deficient ApoE(-/-)Rag2(-/-) mice augmented aortic root atherosclerosis by ≈75% that was ≈30% of lesions in ApoE(-/-) mice; macrophage accumulation similarly increased. Transferred NKT cells were identified in the liver and atherosclerotic lesions of recipient mice. Transfer of CD4(+) NKT cells into T-, B-cell-deficient, and NK cell-deficient ApoE(-/-)Rag2(-/-)γC(-/-) mice also augmented atherosclerosis. These data indicate that CD4(+) NKT cells can exert proatherogenic effects independent of other lymphocytes. To investigate the role of NKT cell-derived interferon-γ, IL-4, and IL-21 cytokines and perforin and granzyme B cytotoxins, CD4(+) NKT cells from mice deficient in these molecules were transferred into NKT cell-deficient ApoE(-/-)Jα18(-/-) mice. CD4(+) NKT cells deficient in IL-4, interferon-γ, or IL-21 augmented atherosclerosis in ApoE(-/-)Jα18(-/-) mice by ≈95%, ≈80%, and ≈70%, respectively. Transfer of CD4(+) NKT cells deficient in perforin or granzyme B failed to augment atherosclerosis. Apoptotic cells, necrotic cores, and proinflammatory VCAM-1 (vascular cell adhesion molecule) and MCP-1 (monocyte chemotactic protein) were reduced in mice receiving perforin-deficient NKT cells. CD4(+) NKT cells are twice as potent as CD4(+) T cells in promoting atherosclerosis. CONCLUSIONS CD4(+) NKT cells potently promote atherosclerosis by perforin and granzyme B-dependent apoptosis that increases postapoptotic necrosis and inflammation.
Collapse
Affiliation(s)
- Yi Li
- From the BakerIDI Heart and Diabetes Institute, Melbourne, Australia (L.Y., K.T., P.K., H.H., V.D., A.B., T.K.); Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School (L.Y., K.T., P.T., B.-H.T., T.K.) and Department of Immunology, Central Clinical School, Faculty of Medicine Nursing and Health Sciences (A.B.), Monash University, Melbourne, Australia; Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia (M.J.S.); and School of Medicine, University of Queensland, Herston, Queensland, Australia (M.J.S.)
| | - Kelly To
- From the BakerIDI Heart and Diabetes Institute, Melbourne, Australia (L.Y., K.T., P.K., H.H., V.D., A.B., T.K.); Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School (L.Y., K.T., P.T., B.-H.T., T.K.) and Department of Immunology, Central Clinical School, Faculty of Medicine Nursing and Health Sciences (A.B.), Monash University, Melbourne, Australia; Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia (M.J.S.); and School of Medicine, University of Queensland, Herston, Queensland, Australia (M.J.S.)
| | - Peter Kanellakis
- From the BakerIDI Heart and Diabetes Institute, Melbourne, Australia (L.Y., K.T., P.K., H.H., V.D., A.B., T.K.); Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School (L.Y., K.T., P.T., B.-H.T., T.K.) and Department of Immunology, Central Clinical School, Faculty of Medicine Nursing and Health Sciences (A.B.), Monash University, Melbourne, Australia; Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia (M.J.S.); and School of Medicine, University of Queensland, Herston, Queensland, Australia (M.J.S.)
| | - Hamid Hosseini
- From the BakerIDI Heart and Diabetes Institute, Melbourne, Australia (L.Y., K.T., P.K., H.H., V.D., A.B., T.K.); Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School (L.Y., K.T., P.T., B.-H.T., T.K.) and Department of Immunology, Central Clinical School, Faculty of Medicine Nursing and Health Sciences (A.B.), Monash University, Melbourne, Australia; Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia (M.J.S.); and School of Medicine, University of Queensland, Herston, Queensland, Australia (M.J.S.)
| | - Virginie Deswaerte
- From the BakerIDI Heart and Diabetes Institute, Melbourne, Australia (L.Y., K.T., P.K., H.H., V.D., A.B., T.K.); Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School (L.Y., K.T., P.T., B.-H.T., T.K.) and Department of Immunology, Central Clinical School, Faculty of Medicine Nursing and Health Sciences (A.B.), Monash University, Melbourne, Australia; Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia (M.J.S.); and School of Medicine, University of Queensland, Herston, Queensland, Australia (M.J.S.)
| | - Peter Tipping
- From the BakerIDI Heart and Diabetes Institute, Melbourne, Australia (L.Y., K.T., P.K., H.H., V.D., A.B., T.K.); Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School (L.Y., K.T., P.T., B.-H.T., T.K.) and Department of Immunology, Central Clinical School, Faculty of Medicine Nursing and Health Sciences (A.B.), Monash University, Melbourne, Australia; Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia (M.J.S.); and School of Medicine, University of Queensland, Herston, Queensland, Australia (M.J.S.)
| | - Mark J Smyth
- From the BakerIDI Heart and Diabetes Institute, Melbourne, Australia (L.Y., K.T., P.K., H.H., V.D., A.B., T.K.); Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School (L.Y., K.T., P.T., B.-H.T., T.K.) and Department of Immunology, Central Clinical School, Faculty of Medicine Nursing and Health Sciences (A.B.), Monash University, Melbourne, Australia; Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia (M.J.S.); and School of Medicine, University of Queensland, Herston, Queensland, Australia (M.J.S.)
| | - Ban-Hock Toh
- From the BakerIDI Heart and Diabetes Institute, Melbourne, Australia (L.Y., K.T., P.K., H.H., V.D., A.B., T.K.); Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School (L.Y., K.T., P.T., B.-H.T., T.K.) and Department of Immunology, Central Clinical School, Faculty of Medicine Nursing and Health Sciences (A.B.), Monash University, Melbourne, Australia; Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia (M.J.S.); and School of Medicine, University of Queensland, Herston, Queensland, Australia (M.J.S.)
| | - Alexander Bobik
- From the BakerIDI Heart and Diabetes Institute, Melbourne, Australia (L.Y., K.T., P.K., H.H., V.D., A.B., T.K.); Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School (L.Y., K.T., P.T., B.-H.T., T.K.) and Department of Immunology, Central Clinical School, Faculty of Medicine Nursing and Health Sciences (A.B.), Monash University, Melbourne, Australia; Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia (M.J.S.); and School of Medicine, University of Queensland, Herston, Queensland, Australia (M.J.S.)
| | - Tin Kyaw
- From the BakerIDI Heart and Diabetes Institute, Melbourne, Australia (L.Y., K.T., P.K., H.H., V.D., A.B., T.K.); Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School (L.Y., K.T., P.T., B.-H.T., T.K.) and Department of Immunology, Central Clinical School, Faculty of Medicine Nursing and Health Sciences (A.B.), Monash University, Melbourne, Australia; Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia (M.J.S.); and School of Medicine, University of Queensland, Herston, Queensland, Australia (M.J.S.).
| |
Collapse
|
237
|
Tsiantoulas D, Sage AP, Mallat Z, Binder CJ. Targeting B cells in atherosclerosis: closing the gap from bench to bedside. Arterioscler Thromb Vasc Biol 2014; 35:296-302. [PMID: 25359862 DOI: 10.1161/atvbaha.114.303569] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Atherosclerotic plaque formation is strongly influenced by different arms of the immune system, including B lymphocytes. B cells are divided into 2 main families: the B1 and the B2 cells. B1 cells are atheroprotective mainly via the production of natural IgM antibodies that bind oxidized low-density lipoprotein and apoptotic cells. B2 cells, which include follicular and marginal zone B cells, are suggested to be proatherogenic. Antibody-mediated depletion of B cells has become a valuable treatment option for certain autoimmune diseases, such as systemic lupus erythematosus and rheumatoid arthritis that are also characterized by the development of premature atherosclerosis. Thus, B cells represent a novel interesting target for therapeutic modulation of the atherosclerotic disease process. Here, we discuss the effect of different of B-cell subsets in experimental atherosclerosis, their mechanism of action as well as potential ways to exploit these findings for the treatment of human disease.
Collapse
Affiliation(s)
- Dimitrios Tsiantoulas
- From the Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria (D.T., C.J.B.); and Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S., Z.M.)
| | - Andrew P Sage
- From the Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria (D.T., C.J.B.); and Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S., Z.M.)
| | - Ziad Mallat
- From the Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria (D.T., C.J.B.); and Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S., Z.M.)
| | - Christoph J Binder
- From the Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria (D.T., C.J.B.); and Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S., Z.M.).
| |
Collapse
|
238
|
Hsue PY, Scherzer R, Grunfeld C, Imboden J, Wu Y, Del Puerto G, Nitta E, Shigenaga J, Schnell Heringer A, Ganz P, Graf J. Depletion of B-cells with rituximab improves endothelial function and reduces inflammation among individuals with rheumatoid arthritis. J Am Heart Assoc 2014; 3:e001267. [PMID: 25336464 PMCID: PMC4323827 DOI: 10.1161/jaha.114.001267] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Individuals with rheumatoid arthritis (RA) are at increased risk for cardiovascular disease, partly due to systemic inflammation and endothelial dysfunction. B‐cells play an important pathogenic role in the inflammatory process that drives RA disease activity. Rituximab, a chimeric murine/human monoclonal antibody that depletes B‐cells, is an effective therapy for RA. The purpose of this study was to determine whether B‐cell depletion with rituximab reduces systemic inflammation and improves macrovascular (brachial artery flow‐mediated dilation, FMD) and microvascular (reactive hyperemia) endothelial function in RA patients. Methods and Results RA patients received a single course of rituximab (1000 mg IV infusion at baseline and on day 15). FMD, reactive hyperemia, inflammatory markers, and clinical assessments were performed at baseline, week 12, and week 24. Twenty patients (95% female, median age 54 years) completed the study. Following treatment, FMD improved from a baseline of 4.5±0.4% to 6.4±0.6% at 12 weeks (mean±SE; P<0.0001), followed by a decline at week 24; a similar pattern was observed for hyperemic velocity. Significant decreases in RA disease scores, high‐sensitivity C‐reactive protein, erythrocyte sedimentation rate, and circulating CD19+ B‐cells were sustained through week 24. Cholesterol and triglycerides became significantly although modestly elevated during the study. Conclusions Depletion of B‐cells with rituximab improved macrovascular and microvascular endothelial function and reduced systemic inflammation, despite modest elevation in lipids. Given these results, rituximab should be evaluated in the future for its possible role in reducing excess cardiovascular risk in RA. Clinical Trial Registration URL http://ClinicalTrials.gov. Unique identifier: NCT00844714.
Collapse
Affiliation(s)
- Priscilla Y Hsue
- Division of Cardiology, Department of Medicine, San Francisco General Hospital, University of California, San Francisco, CA (P.Y.H., Y.W., E.N., A.S.H., P.G.)
| | - Rebecca Scherzer
- San Francisco Veterans Affairs Medical Center, Department of Medicine UCSF, University of California, San Francisco, CA (R.S., C.G., J.S.)
| | - Carl Grunfeld
- San Francisco Veterans Affairs Medical Center, Department of Medicine UCSF, University of California, San Francisco, CA (R.S., C.G., J.S.)
| | - John Imboden
- Division of Rheumatology, Department of Medicine, San Francisco General Hospital, University of California, San Francisco, CA (J.I., G.P., J.G.)
| | - Yuaner Wu
- Division of Cardiology, Department of Medicine, San Francisco General Hospital, University of California, San Francisco, CA (P.Y.H., Y.W., E.N., A.S.H., P.G.)
| | - Gus Del Puerto
- Division of Rheumatology, Department of Medicine, San Francisco General Hospital, University of California, San Francisco, CA (J.I., G.P., J.G.)
| | - Elaine Nitta
- Division of Cardiology, Department of Medicine, San Francisco General Hospital, University of California, San Francisco, CA (P.Y.H., Y.W., E.N., A.S.H., P.G.)
| | - Judy Shigenaga
- San Francisco Veterans Affairs Medical Center, Department of Medicine UCSF, University of California, San Francisco, CA (R.S., C.G., J.S.)
| | - Amanda Schnell Heringer
- Division of Cardiology, Department of Medicine, San Francisco General Hospital, University of California, San Francisco, CA (P.Y.H., Y.W., E.N., A.S.H., P.G.)
| | - Peter Ganz
- Division of Cardiology, Department of Medicine, San Francisco General Hospital, University of California, San Francisco, CA (P.Y.H., Y.W., E.N., A.S.H., P.G.)
| | - Jonathan Graf
- Division of Rheumatology, Department of Medicine, San Francisco General Hospital, University of California, San Francisco, CA (J.I., G.P., J.G.)
| |
Collapse
|
239
|
|
240
|
Abstract
Adaptive immunity is involved in the pathogenesis of atherosclerosis, but the recruitment of T and B lymphocytes to atherosclerotic lesions is not as well studied as that of monocytes. In this review, we summarize the current understanding of the role of lymphocyte subsets in the pathogenesis of atherosclerosis and discuss chemokines and chemokine receptors involved in lymphocyte homing to atherosclerotic lesions. We review evidence for involvement of the chemokines CCL5, CCL19, CCL21, CXCL10, and CXCL16 and macrophage migration inhibitory factor in lymphocyte homing in atherosclerosis. Also, we review the role of their receptors CCR5, CCR6, CCR7, CXCR3, CXCR6, and CXCR2/CXCR4 and the role of the L-selectin in mouse models of atherosclerosis.
Collapse
Affiliation(s)
- Jie Li
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA
| | - Klaus Ley
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA.
| |
Collapse
|
241
|
Strissel KJ, Denis GV, Nikolajczyk BS. Immune regulators of inflammation in obesity-associated type 2 diabetes and coronary artery disease. Curr Opin Endocrinol Diabetes Obes 2014; 21:330-8. [PMID: 25106001 PMCID: PMC4251956 DOI: 10.1097/med.0000000000000085] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW To summarize current work identifying inflammatory components that underlie associations between obesity-associated type 2 diabetes and coronary artery disease. RECENT FINDINGS Recent studies implicate immune cells as drivers of pathogenic inflammation in human type 2 diabetes. Inflammatory lymphocytes characterize unhealthy adipose tissue, but regional adipose volume, primarily visceral and pericardial fat, also predict severity and risk for obesity-associated coronary artery disease. Having a greater understanding of shared characteristics between inflammatory cells from different adipose tissue depots and a more accessible tissue, such as blood, will facilitate progress toward clinical translation of our appreciation of obesity as an inflammatory disease. SUMMARY Obesity predisposes inflammation and metabolic dysfunction through multiple mechanisms, but these mechanisms remain understudied in humans. Studies of obese patients have identified disproportionate impacts of specific T cell subsets in metabolic diseases like type 2 diabetes. On the basis of demonstration that adipose tissue inflammation is depot-specific, analysis of adiposity by waist-to-hip ratio or MRI will increase interpretive value of lymphocyte-focused studies and aid clinicians in determining which obese individuals are at highest risk for coronary artery disease. New tools to combat obesity-associated coronary artery disease and other comorbidities will stem from identification of immune cell-mediated inflammatory networks that are amenable to pharmacological interventions.
Collapse
Affiliation(s)
- Katherine J. Strissel
- Department of Medicine, Boston University School of Medicine, Boston MA, USA
- Cancer Research Center, Boston University School of Medicine, Boston MA, USA
| | - Gerald V. Denis
- Department of Medicine, Boston University School of Medicine, Boston MA, USA
- Cancer Research Center, Boston University School of Medicine, Boston MA, USA
| | - Barbara S. Nikolajczyk
- Department of Medicine, Boston University School of Medicine, Boston MA, USA
- Department of Microbiology, Boston University School of Medicine, Boston MA, USA
| |
Collapse
|
242
|
Sage AP, Murphy D, Maffia P, Masters LM, Sabir SR, Baker LL, Cambrook H, Finigan AJ, Ait-Oufella H, Grassia G, Harrison JE, Ludewig B, Reith W, Hansson GK, Reizis B, Hugues S, Mallat Z. MHC Class II-restricted antigen presentation by plasmacytoid dendritic cells drives proatherogenic T cell immunity. Circulation 2014; 130:1363-73. [PMID: 25223984 DOI: 10.1161/circulationaha.114.011090] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Plasmacytoid dendritic cells (pDCs) bridge innate and adaptive immune responses and are important regulators of immuno-inflammatory diseases. However, their role in atherosclerosis remains elusive. METHODS AND RESULTS Here, we used genetic approaches to investigate the role of pDCs in atherosclerosis. Selective pDC deficiency in vivo was achieved using CD11c-Cre × Tcf4(-/flox) bone marrow transplanted into Ldlr(-/-) mice. Compared with control Ldlr(-/-) chimeric mice, CD11c-Cre × Tcf4(-/flox) mice had reduced atherosclerosis levels. To begin to understand the mechanisms by which pDCs regulate atherosclerosis, we studied chimeric Ldlr(-/-) mice with selective MHCII deficiency on pDCs. Significantly, these mice also developed reduced atherosclerosis compared with controls without reductions in pDC numbers or changes in conventional DCs. MHCII-deficient pDCs showed defective stimulation of apolipoprotein B100-specific CD4(+) T cells in response to native low-density lipoprotein, whereas production of interferon-α was not affected. Finally, the atheroprotective effect of selective MHCII deficiency in pDCs was associated with significant reductions of proatherogenic T cell-derived interferon-γ and lesional T cell infiltration, and was abrogated in CD4(+) T cell-depleted animals. CONCLUSIONS This study supports a proatherogenic role for pDCs in murine atherosclerosis and identifies a critical role for MHCII-restricted antigen presentation by pDCs in driving proatherogenic T cell immunity.
Collapse
Affiliation(s)
- Andrew P Sage
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Deirdre Murphy
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Pasquale Maffia
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Leanne M Masters
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Suleman R Sabir
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Lauren L Baker
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Helen Cambrook
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Alison J Finigan
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Hafid Ait-Oufella
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Gianluca Grassia
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - James E Harrison
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Burkhard Ludewig
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Walter Reith
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Göran K Hansson
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Boris Reizis
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Stéphanie Hugues
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.)
| | - Ziad Mallat
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom (A.P.S., D.M., L.M.M., L.L.B., A.J.F., J.E.H., Z.M.); Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M., S.R.S., H.C., G.G.); Institut National de la Santé et de la Recherche Médicale, Unit 970, Paris Cardiovascular Research Center, Paris, France (H.A., Z.M.); the Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M., G.G.); Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland (B.L.); the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland (W.R.); Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden (G.K.H.); the Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY (B.R.); and the Department of Pathology, University of Geneva Medical School, CH-1211 Geneva, Switzerland (S.H.).
| |
Collapse
|
243
|
B2 cells suppress experimental abdominal aortic aneurysms. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:3130-41. [PMID: 25194661 DOI: 10.1016/j.ajpath.2014.07.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 06/16/2014] [Accepted: 07/10/2014] [Indexed: 12/11/2022]
Abstract
Recent reports of rupture in patients with abdominal aortic aneurysm (AAA) receiving B-cell depletion therapy highlight the importance of understanding the role of B cells (B1 and B2 subsets) in the development of AAA. We hypothesized that B2 cells aggravate experimental aneurysm formation. The IHC staining revealed infiltration of B cells in the aorta of wild-type (C57BL/6) mice at day 7 after elastase perfusion and persisted through day 21. Quantification of immune cell types using flow cytometry at day 14 showed significantly greater infiltration of mononuclear cells, including B cells (B2: 93% of total B cells) and T cells in elastase-perfused aortas compared with saline-perfused or normal aortas. muMT (mature B-cell deficient) mice were prone to AAA formation similar to wild-type mice in two different experimental AAA models. Contradicting our hypothesis, adoptive transfer of B2 cells suppressed AAA formation (102.0% ± 7.3% versus 75.2% ± 5.5%; P < 0.05) with concomitant increase in the splenic regulatory T cell (0.24% ± 0.03% versus 0.92% ± 0.23%; P < 0.05) and decrease in aortic infiltration of mononuclear cells. Our data suggest that B2 cells constitute the largest population of B cells in experimental AAA. Furthermore, B2 cells, in the absence of other B-cell subsets, increase splenic regulatory T-cell population and suppress AAA formation.
Collapse
|
244
|
Turunen SP, Kummu O, Wang C, Harila K, Mattila R, Sahlman M, Pussinen PJ, Hörkkö S. Immunization with malondialdehyde-modified low-density lipoprotein (LDL) reduces atherosclerosis in LDL receptor-deficient mice challenged with Porphyromonas gingivalis. Innate Immun 2014; 21:370-85. [PMID: 25134521 DOI: 10.1177/1753425914542444] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 06/06/2014] [Indexed: 01/31/2023] Open
Abstract
Periodontal infections increase the risk of atherosclerotic vascular disease via partly unresolved mechanisms. Of the natural IgM Abs that recognize molecular mimicry on bacterial epitopes and modified lipid and protein structures, IgM directed against oxidized low-density lipoprotein (LDL) is associated with atheroprotective properties. Here, the effect of natural immune responses to malondialdehyde-modified LDL (MDA-LDL) in conferring protection against atherosclerosis, which was accelerated by the major periodontopathogen Porphyromonas gingivalis, was investigated. LDL receptor-deficient (LDLR(-/-)) mice were immunized with mouse MDA-LDL without adjuvant before topical application challenge with live P. gingivalis. Atherosclerosis was analyzed after a high-fat diet, and plasma IgG and IgM Ab levels were measured throughout the study, and the secretion of IL-5, IL-10 and IFN-γ in splenocytes stimulated with MDA-LDL was determined. LDLR(-/-) mice immunized with MDA-LDL had elevated IgM and IgG levels to MDA-LDL compared with saline-treated controls. MDA-LDL immunization diminished aortic lipid depositions after challenge with P. gingivalis compared with mice receiving only P. gingivalis challenge. Immunization of LDLR(-/-) mice with homologous MDA-LDL stimulated the production of IL-5, implicating general activation of B-1 cells. Immune responses to MDA-LDL protected from the P. gingivalis-accelerated atherosclerosis. Thus, the linkage between bacterial infectious burden and atherogenesis is suggested to be modulated via natural IgM directed against cross-reactive epitopes on bacteria and modified LDL.
Collapse
Affiliation(s)
- S Pauliina Turunen
- Department of Medical Microbiology and Immunology, Institute of Diagnostics, University of Oulu, Finland and Medical Research Center Oulu, Oulu, Finland NordLab Oulu, Oulu University Hospital, Oulu, Finland
| | - Outi Kummu
- Department of Medical Microbiology and Immunology, Institute of Diagnostics, University of Oulu, Finland and Medical Research Center Oulu, Oulu, Finland NordLab Oulu, Oulu University Hospital, Oulu, Finland
| | - Chunguang Wang
- Department of Medical Microbiology and Immunology, Institute of Diagnostics, University of Oulu, Finland and Medical Research Center Oulu, Oulu, Finland
| | - Kirsi Harila
- Department of Medical Microbiology and Immunology, Institute of Diagnostics, University of Oulu, Finland and Medical Research Center Oulu, Oulu, Finland
| | - Riikka Mattila
- Department of Medical Microbiology and Immunology, Institute of Diagnostics, University of Oulu, Finland and Medical Research Center Oulu, Oulu, Finland
| | - Marjo Sahlman
- Department of Medical Microbiology and Immunology, Institute of Diagnostics, University of Oulu, Finland and Medical Research Center Oulu, Oulu, Finland
| | | | - Sohvi Hörkkö
- Department of Medical Microbiology and Immunology, Institute of Diagnostics, University of Oulu, Finland and Medical Research Center Oulu, Oulu, Finland NordLab Oulu, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
245
|
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
|
246
|
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
|
247
|
Gjurich BN, Taghavie-Moghadam PL, Ley K, Galkina EV. L-selectin deficiency decreases aortic B1a and Breg subsets and promotes atherosclerosis. Thromb Haemost 2014; 112:803-11. [PMID: 24989887 DOI: 10.1160/th13-10-0865] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 04/29/2014] [Indexed: 12/18/2022]
Abstract
There is a significant recruitment of leucocytes into aortas during atherogenesis. L-selectin regulates leucocyte migration into secondary lymphoid and peripheral tissues and was proposed to play a role in leucocyte homing into aortas. Here, we determine the role of L-selectin in atherosclerosis. L-selectin-deficient Apoe-/- (Sell-/-Apoe-/-) mice had a 74% increase in plaque burden compared to Apoe-/- mice fed a chow diet for 50 weeks. Elevated atherosclerosis was accompanied by increased aortic leucocyte content, but a 50% reduction in aortic B cells despite elevated B cell counts in the blood. Follicular B cells represented 65%, whereas B1a and regulatory B cells (Breg) comprised 5% of aortic B cells. B1a and Breg cell subsets were reduced in Sell-/-Apoe-/- aortas with accompanied two-fold decrease in aortic T15 antibody and 1.2-fold decrease of interleukin-10 (IL-10) levels. L-selectin was required for B1 cell homing to the atherosclerotic aorta, as demonstrated by a 1.5-fold decrease in the migration of Sell-/-Apoe-/- vs Apoe-/- cells. Notably, we found a 1.6-fold increase in CD68hi macrophages in Sell-/-Apoe-/- compared to Apoe-/- aortas, despite comparable blood monocyte numbers and L-selectin-dependent aortic homing. L-selectin had no effect on neutrophil migration into aorta, but led to elevated blood neutrophil numbers, suggesting a potential involvement of neutrophils in atherogenesis of Sell-/-Apoe-/- mice. Thus, L-selectin deficiency increases peripheral blood neutrophil and lymphocyte numbers, decreases aortic B1a and Breg populations, T15 antibody and IL-10 levels, and increases aortic macrophage content of Sell-/-Apoe-/- mice. Altogether, these data provide evidence for an overall atheroprotective role of L-selectin.
Collapse
Affiliation(s)
| | | | | | - Elena V Galkina
- Elena V. Galkina, PhD, Associate Professor, Dept. Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Road, Norfolk, VA 23507-1696, USA, Tel.: +1 757 446 5019, Fax: +1 757 624 2255, E-mail:
| |
Collapse
|
248
|
Ait-Oufella H, Sage AP, Mallat Z, Tedgui A. Adaptive (T and B cells) immunity and control by dendritic cells in atherosclerosis. Circ Res 2014; 114:1640-60. [PMID: 24812352 DOI: 10.1161/circresaha.114.302761] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Chronic inflammation in response to lipoprotein accumulation in the arterial wall is central in the development of atherosclerosis. Both innate and adaptive immunity are involved in this process. Adaptive immune responses develop against an array of potential antigens presented to effector T lymphocytes by antigen-presenting cells, especially dendritic cells. Functional analysis of the role of different T-cell subsets identified the Th1 responses as proatherogenic, whereas regulatory T-cell responses exert antiatherogenic activities. The effect of Th2 and Th17 responses is still debated. Atherosclerosis is also associated with B-cell activation. Recent evidence established that conventional B-2 cells promote atherosclerosis. In contrast, innate B-1 B cells offer protection through secretion of natural IgM antibodies. This review discusses the recent development in our understanding of the role of T- and B-cell subsets in atherosclerosis and addresses the role of dendritic cell subpopulations in the control of adaptive immunity.
Collapse
Affiliation(s)
- Hafid Ait-Oufella
- From INSERM UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (H.A.-O., Z.M., A.T.); Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Paris, France (H.A.-O.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S., Z.M.)
| | | | | | | |
Collapse
|
249
|
Llorente-Cortés V, de Gonzalo-Calvo D, Orbe J, Páramo JA, Badimon L. Signature of subclinical femoral artery atherosclerosis in peripheral blood mononuclear cells. Eur J Clin Invest 2014; 44:539-48. [PMID: 24716741 DOI: 10.1111/eci.12267] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 04/07/2014] [Indexed: 12/15/2022]
Abstract
BACKGROUND Peripheral arterial disease is a relevant public health problem associated with increased risk of morbimortality. Most of the patients with this condition are asymptomatic. Therefore, the development of accessible biochemical markers seems to be necessary to anticipate diagnosis. Our hypothesis is that asymptomatic subjects with objectively confirmed femoral artery atherosclerosis could be distinguished from control subjects by gene expression analysis in peripheral blood mononuclear cells (PBMC). MATERIALS AND METHODS A total of 37 asymptomatic males over 50 years old were recruited at the University Clinic of Navarra (Spain). Nineteen participants were free from atherosclerotic vascular disease and 18 participants presented subclinical femoral artery atherosclerosis defined by means of Doppler ultrasound. PBMC were isolated from blood and the RNA extracted. A panel of atherosclerotic-related genes were evaluated by Taqman low-density array. RESULTS In univariate logistic regression models, we found a direct relationship between IL4, ITGAM and TLR2 expression levels in PBMC and femoral atherosclerosis, even when the models were adjusted for age and hypertension prevalence. Multivariate logistic regression models showed that elevated IL4 expression levels were intimately associated with subclinical femoral atherosclerosis after adjusting for the same potential confounders. CONCLUSIONS Current data suggest that gene expression in PBMC, in particular IL4 expression, could be a useful tool in the diagnosis of femoral artery atherosclerosis in asymptomatic patients. Furthermore, in patients with no differences in cardiovascular risk factors except for hypertension, the results point to the immune and inflammatory deregulation as a feature of subclinical peripheral atherosclerosis.
Collapse
|
250
|
Mohanta SK, Yin C, Peng L, Srikakulapu P, Bontha V, Hu D, Weih F, Weber C, Gerdes N, Habenicht AJ. Artery Tertiary Lymphoid Organs Contribute to Innate and Adaptive Immune Responses in Advanced Mouse Atherosclerosis. Circ Res 2014; 114:1772-87. [PMID: 24855201 DOI: 10.1161/circresaha.114.301137] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tertiary lymphoid organs emerge in tissues in response to nonresolving inflammation. Recent research characterized artery tertiary lymphoid organs in the aorta adventitia of aged apolipoprotein E–deficient mice. The atherosclerosis-associated lymphocyte aggregates are organized into distinct compartments, including separate T-cell areas harboring conventional, monocyte-derived, lymphoid, and plasmacytoid dendritic cells, as well as activated T-cell effectors and memory cells; B-cell follicles containing follicular dendritic cells in activated germinal centers; and peripheral niches of plasma cells. Artery tertiary lymphoid organs show marked neoangiogenesis, aberrant lymphangiogenesis, and extensive induction of high endothelial venules. Moreover, newly formed lymph node–like conduits connect the external lamina with high endothelial venules in T-cell areas and also extend into germinal centers. Mouse artery tertiary lymphoid organs recruit large numbers of naïve T cells and harbor lymphocyte subsets with opposing activities, including CD4
+
and CD8
+
effector and memory T cells, natural and induced CD4
+
regulatory T cells, and memory B cells at different stages of differentiation. These data suggest that artery tertiary lymphoid organs participate in primary immune responses and organize T- and B-cell autoimmune responses in advanced atherosclerosis. In this review, we discuss the novel concept that pro- and antiatherogenic immune responses toward unknown arterial wall–derived autoantigens may be organized by artery tertiary lymphoid organs and that disruption of the balance between pro- and antiatherogenic immune cell subsets may trigger clinically overt atherosclerosis.
Collapse
Affiliation(s)
- Sarajo Kumar Mohanta
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Changjun Yin
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Li Peng
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Prasad Srikakulapu
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Vineela Bontha
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Desheng Hu
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Falk Weih
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Christian Weber
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Norbert Gerdes
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Andreas J.R. Habenicht
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| |
Collapse
|