151
|
Fischer JW. Role of hyaluronan in atherosclerosis: Current knowledge and open questions. Matrix Biol 2018; 78-79:324-336. [PMID: 29510229 DOI: 10.1016/j.matbio.2018.03.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/20/2018] [Accepted: 03/01/2018] [Indexed: 12/21/2022]
Abstract
Hyaluronan (HA), HA synthases (HAS) and HA receptors are expressed during the progression of atherosclerotic plaques. HA is thought to promote the activated phenotype of local vascular smooth muscle cells characterized by increased migration, proliferation and matrix synthesis. Furthermore, HA may modulate the immune response by increasing macrophage retention and by promoting the polarization of Th1 cells that enhance macrophage driven inflammation as well. The pro-atherosclerotic functions of HA are opposed by the presence of HA in the glycocalyx where it critically contributes to anti-thrombotic and anti-inflammatory function of the glycocalyx. Patients with atherosclerosis often are affected by comorbidities among them diabetes mellitus type 2 and inflammatory comorbidities. Diabetes mellitus type 2 likely has close interrelations to HA synthesis in atherosclerosis because the activity and transcription of HA synthases are sensitive to the intracellular glucose metabolism, which determines the substrate availability and the posttranslational modifications of HA synthases. The pro-inflammatory comorbidities aggravate the course of atherosclerosis and will affect the expression of the genes related to HA biosynthesis, -degradation, HA-matrix assembly or signaling. One example being the induction of HAS3 by interleukin-1β and other cytokines. Furthermore complications of atherosclerosis such as the healing after myocardial infarction also involve HA responses.
Collapse
Affiliation(s)
- Jens W Fischer
- Institut für Pharmakologie und Klinische Pharmakologie, University Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany; CARID, Cardiovascular Research Institute Düsseldorf, University Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
152
|
Teixeira V, Tam LS. Novel Insights in Systemic Lupus Erythematosus and Atherosclerosis. Front Med (Lausanne) 2018; 4:262. [PMID: 29435447 PMCID: PMC5796914 DOI: 10.3389/fmed.2017.00262] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/28/2017] [Indexed: 01/22/2023] Open
Abstract
Introduction The systemic inflammatory nature of systemic lupus erythematosus (SLE) is well patent not only in the diverse clinical manifestations of the disease but also in the increased risk of premature atherosclerosis and cardiovascular events (CVE), making SLE one of the most complex diseases to study and manage in clinical practice. Aim To travel from old aspects to modern insights on the physiopathology, new molecular biomarkers, imaging methods of atherosclerosis assessment, and the potential treatments of atherosclerosis in SLE. Methods We conducted a literature search using PubMed database and performed a critical review. Conclusion/discussion Several developments have taken place in the understanding of the relationship between SLE and premature atherosclerosis. Nevertheless, cardiovascular diseases are still the major cause of reduced life expectancy in SLE and the main cause of death. The lack of standardization methods for the imaging assessment of atherosclerosis in SLE and the multifactorial nature of the disease are well patriated in the difficulty of achieving consistent and reproducible results among studies that focus in cardiovascular risk assessment and prediction. A raising number of molecular biomarkers of atherosclerosis have been proposed, but the combination of several biomarkers and risk factors may better estimate cardiovascular disease risk. Moreover, the development of effective therapies to prevent progression of atherosclerosis and CVE shall address systemic inflammation.
Collapse
Affiliation(s)
- Vítor Teixeira
- Rheumatology Department, Centro Hospitalar de Lisboa Norte, EPE, Hospital de Santa Maria, Lisbon, Portugal
| | - Lai-Shan Tam
- Department of Medicine and Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| |
Collapse
|
153
|
Wolf D, Stachon P, Bode C, Zirlik A. Inflammatory mechanisms in atherosclerosis. Hamostaseologie 2017; 34:63-71. [DOI: 10.5482/hamo-13-09-0050] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 12/05/2013] [Indexed: 01/13/2023] Open
Abstract
SummaryThroughout the last two decades inflammation has been recognized as the central mechanism underlying atherogenesis. A multitude of basic science work demonstrates the pivotal role of inflammatory processes during every step of atherosclerotic plaque formation: From initiation via propagation to complication.This review describes some of the key mechanisms involved with a particular focus on the diverse group of inflammatory cells and their subsets that distinctly contribute to atherogenic and anti-atherogenic phenomena. Furthermore, we summarize the controlling action of a tight network of co-stimulatory molecules and cytokines orchestrating the inflammatory and anti-inflammatory effector functions. Finally, the current status of clinical trials evaluating anti-inflammatory/ immune-modulatory treatment strategies is summarized and an outlook for future therapeutic implications is provided.
Collapse
|
154
|
Abstract
Atherosclerosis is initiated by cholesterol entry into arteries that triggers chronic immune-inflammatory lesions in the vessels. Early lesions are clinically insignificant but advanced complex lesions and vulnerable rupture prone lesions impact on quality of life and can be life threatening. Rupture of vulnerable atherosclerotic lesions initiates thrombotic occlusion of vital arteries precipitating heart attacks and strokes that remain major killers globally despite therapeutic use of statins to lower blood cholesterol levels. Conventional B2 cells are proatherogenic whereas peritoneal Bla cells are atheroprotective. Depletion of B2 cells by administration of mAb to CD20 or to BAFF receptor or in BAFF receptor-deficient mice ameliorates atherosclerosis. B2 cells may promote atherosclerosis by production of IgG, secretion of proinflammatory cytokine TNFα and activation of CD4 T cells. Together these B2 cell mechanisms contribute to generation of rupture-prone vulnerable atherosclerotic plaques characterised by large necrotic cores. In contrast, peritoneal Bla cells protect against atherosclerosis by secretion of natural IgM that scavenges apoptotic cells and oxidised LDL and reduces necrotic cores in atherosclerotic lesions. These atheroprotective effects can be further increased by stimulating Bla cells by administration of apoptotic cells, liposomes of phosphatidylserine abundant on surfaces of apoptotic cell, by mAb to TIM1, a phosphatidylserine receptor expressed by B1a cells and by TLR4-MyD88 activation. Experimental studies of atherosclerosis in mouse models indicate that reductions in atherogenic B2 cells and/or activation of atheroprotective B1a cells protects against atherosclerosis development, findings which have potential for clinical translation to reduce risks of deaths from heart attacks and strokes.
Collapse
Affiliation(s)
- Tin Kyaw
- a Australia and Baker IDI Heart and Diabetes Institute , Victoria , Australia.,b Department of Medicine , Southern Clinical School, Monash University , Victoria , Australia , and
| | - Peter Tipping
- b Department of Medicine , Southern Clinical School, Monash University , Victoria , Australia , and
| | - Alex Bobik
- a Australia and Baker IDI Heart and Diabetes Institute , Victoria , Australia.,c Department of Immunology , Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University , Victoria , Australia
| | - Ban-Hock Toh
- b Department of Medicine , Southern Clinical School, Monash University , Victoria , Australia , and
| |
Collapse
|
155
|
Fonseca FAH, Izar MC, Maugeri IML, Berwanger O, Damiani LP, Pinto IM, Szarf G, França CN, Bianco HT, Moreira FT, Caixeta A, Alves CMR, Soriano Lopes A, Klassen A, Tavares MFM, Fonseca HA, Carvalho ACC. Effects of four antiplatelet/statin combined strategies on immune and inflammatory responses in patients with acute myocardial infarction undergoing pharmacoinvasive strategy: Design and rationale of the B and T Types of Lymphocytes Evaluation in Acute Myocardial Infarction (BATTLE-AMI) study: study protocol for a randomized controlled trial. Trials 2017; 18:601. [PMID: 29258572 PMCID: PMC5735810 DOI: 10.1186/s13063-017-2361-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 11/17/2017] [Indexed: 02/07/2023] Open
Abstract
Background Early reperfusion of the occluded coronary artery during acute myocardial infarction is considered crucial for reduction of infarcted mass and recovery of ventricular function. Effective microcirculation and the balance between protective and harmful lymphocytes may have roles in reperfusion injury and may affect final ventricular remodeling. Methods/design BATTLE-AMI is an open-label, randomized trial comparing the effects of four therapeutic strategies (rosuvastatin/ticagrelor, rosuvastatin/clopidogrel, simvastatin plus ezetimibe/ticagrelor, or simvastatin plus ezetimibe/clopidogrel) on infarcted mass and left ventricular ejection fraction (LVEF) (blinded endpoints) in patients with ST-segment elevation myocardial infarction submitted to fibrinolytic therapy before coronary angiogram (pharmacoinvasive strategy). All patients (n = 300, 75 per arm) will be followed up for six months. The effects of treatment on subsets of B and T lymphocytes will be determined by flow-cytometry/ELISPOT and will be correlated with the infarcted mass, LVEF, and microcirculation perfusion obtained by cardiac magnetic resonance imaging. The primary hypothesis is that the combined rosuvastatin/ticagrelor therapy will be superior to other therapies (particularly for the comparison with simvastatin plus ezetimibe/clopidogrel) for the achievement of better LVEF at 30 days (primary endpoint) and smaller infarcted mass (secondary endpoint) at 30 days and six months. The trial will also evaluate the improvement in the immune/inflammatory responses mediated by B and T lymphocytes. Omics field (metabolomics and proteomics) will help to understand these responses by molecular events. Discussion BATTLE-AMI is aimed to (1) evaluate the role of subsets of lymphocytes on microcirculation improvement and (2) show how the choice of statin/antiplatelet therapy may affect cardiac remodeling after acute myocardial infarction with ST elevation. Trial registration ClinicalTrials.gov, NCT02428374. Registered on 28 September 2014. Electronic supplementary material The online version of this article (doi:10.1186/s13063-017-2361-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Francisco A H Fonseca
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil.
| | - Maria Cristina Izar
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | - Ieda M L Maugeri
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | - Otavio Berwanger
- Hospital do Coração, Rua Desembargador Eliseu Guilherme, 147, São Paulo, Brazil
| | - Lucas P Damiani
- Hospital do Coração, Rua Desembargador Eliseu Guilherme, 147, São Paulo, Brazil
| | - Ibraim M Pinto
- Instituto Dante Pazzanese de Cardiologia, Avenida Dante Pazzanese 500, São Paulo, Brazil
| | - Gilberto Szarf
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | - Carolina N França
- Universidade Santo Amaro, Rua Professor Enéas de Siqueira 340, São Paulo, Brazil
| | - Henrique T Bianco
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | - Flavio T Moreira
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | - Adriano Caixeta
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | - Claudia M R Alves
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | - Aline Soriano Lopes
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | - Aline Klassen
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | - Marina F M Tavares
- Universidade de São Paulo, Avenida Professor Lineu Prestes, 748, São Paulo, Brazil
| | - Henrique A Fonseca
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | - Antonio C C Carvalho
- Universidade Federal de São Paulo, Rua Loefgren 1350, 04040-001, São Paulo, SP, Brazil
| | | |
Collapse
|
156
|
Nus M, Tsiantoulas D, Mallat Z. Plan B (-cell) in atherosclerosis. Eur J Pharmacol 2017; 816:76-81. [PMID: 28882560 DOI: 10.1016/j.ejphar.2017.09.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 08/31/2017] [Accepted: 09/01/2017] [Indexed: 11/18/2022]
Abstract
Atherosclerosis is a leading cause of death worldwide. It is a complex chronic inflammatory disease involving interactions between vascular, circulating and immune cells. B cells play an important role in chronic inflammation producing antibodies and regulating T and natural killer (NKT) cell activation. The role of B cells in atherosclerosis is complex, with atherogenic and protective roles assigned for distinct B cell subsets. Drugs that deplete B cells or modulate their functions are now used in the treatment of various autoimmune diseases in humans. Here, we briefly review the roles of B cell subsets in atherogenesis, and emphasize the potential impact of B cell targeted therapies on the cardiovascular risk of treated patients. Developing more B cell subset-specific therapies would lead to more effective treatments with enhanced safety profile.
Collapse
Affiliation(s)
- Meritxell Nus
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Dimitrios Tsiantoulas
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Ziad Mallat
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK; Institut National de la Santé et de la Recherche Médicale, U970 Paris, France.
| |
Collapse
|
157
|
Blair P, Leib C, Goddard M, Rosser E, Park I, Nilsson AH, Nilsson J, Strom A, Cross A, Cole J, Mauri C, Monaco C. B regulatory cells are increased in hypercholesterolaemic mice and protect from lesion development via IL-10. Thromb Haemost 2017; 114:835-47. [DOI: 10.1160/th14-12-1084] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 05/07/2015] [Indexed: 11/05/2022]
Abstract
SummaryWhilst innate B1-B cells are atheroprotective, adaptive B2-B cells are considered pro-atherogenic. Different subsets of B regulatory cells (Breg) have been described. In experimental arthritis and lupus-like disease, Breg are contained within the CD21hiCD23hiCD24hi B cell pool. The existence and role of Breg in vascular disease is not known. We sought to investigate the existence, identity and location of Breg in vascular disease. The representation of B2-B cell subsets in the spleens and lymph nodes (LNs) of Apolipoprotein E-/- (ApoE-/-) mice compared to controls was characterised by flow cytometry. Additionally, we utilised a model of neointima formation based on the placement of a perivascular collar around the carotid artery in ApoE-/- mice to ascertain whether B cells and B cell subsets confer protection against lesion development. Adoptive transfer of B cells was performed from wild type or genetically modified mice. We showed that CD21hiCD23hiCD24hi B cells are unexpectedly increased in the draining LNs of ApoE-/- mice. Adoptive transfer of LN-derived B2-B cells or purified CD21hiCD23hiCD24hi B cells to syngeneic mice reduced lesion size and inflammation without changing serum cholesterol levels. Follicular B2-B cells did not confer protection. IL-10 blockade or transfer of IL10-deficient B cells prevented LN-derived B cell-mediated protection. This is the first identification of a specific LN-derived B2-Breg subset that confers IL-10 mediated protection from neointima formation. This may open the way for immune modulatory approaches in cardiovascular disease.
Collapse
|
158
|
Homann S, Grandoch M, Kiene LS, Podsvyadek Y, Feldmann K, Rabausch B, Nagy N, Lehr S, Kretschmer I, Oberhuber A, Bollyky P, Fischer JW. Hyaluronan synthase 3 promotes plaque inflammation and atheroprogression. Matrix Biol 2017; 66:67-80. [PMID: 28987865 DOI: 10.1016/j.matbio.2017.09.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 09/18/2017] [Accepted: 09/19/2017] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Hyaluronan (HA) is a prominent component of the provisional extracellular matrix (ECM) present in the neointima of atherosclerotic plaques. Here the role of HA synthase 3 (HAS3) in atheroprogression was studied. APPROACH AND RESULTS It is demonstrated here that HAS isoenzymes 1, -2 and -3 are expressed in human atherosclerotic plaques of the carotid artery. In Apolipoprotein E (Apoe)-deficient mice Has3 expression is increased early during lesion formation when macrophages enter atherosclerotic plaques. Importantly, HAS3 expression in vascular smooth muscle cells (VSMC) was found to be regulated by interleukin 1 β (IL-1β) in an NFkB dependent manner and blocking antibodies to IL-1β abrogate Has3 expression in VSMC by activated macrophages. Has3/Apoe double deficient mice developed less atherosclerosis characterized by decreased Th1-cell responses, decreased IL-12 release, and decreased macrophage-driven inflammation. CONCLUSIONS Inhibition of HAS3-dependent synthesis of HA dampens systemic Th1 cell polarization and reduces plaque inflammation. These data suggest that HAS3 might be a promising therapeutic target in atherosclerosis. Moreover, because HAS3 is regulated by IL-1β, our results suggest that therapeutic anti-IL-1β antibodies, recently tested in human clinical trials (CANTOS), may exert their beneficial effects on inflammation in post-myocardial infarction patients in part via effects on HAS3. TOC categorybasic study TOC subcategoryarteriosclerosis.
Collapse
Affiliation(s)
- Susanne Homann
- Institut für Pharmakologie und Klinische Pharmakologie, University Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Maria Grandoch
- Institut für Pharmakologie und Klinische Pharmakologie, University Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Lena S Kiene
- Institut für Pharmakologie und Klinische Pharmakologie, University Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Yanina Podsvyadek
- Institut für Pharmakologie und Klinische Pharmakologie, University Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Kathrin Feldmann
- Institut für Pharmakologie und Klinische Pharmakologie, University Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Berit Rabausch
- Institut für Pharmakologie und Klinische Pharmakologie, University Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Nadine Nagy
- Institut für Pharmakologie und Klinische Pharmakologie, University Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany; Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford Immunology, Stanford, USA
| | - Stefan Lehr
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, Düsseldorf, Germany
| | - Inga Kretschmer
- Institut für Pharmakologie und Klinische Pharmakologie, University Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Alexander Oberhuber
- Department of Vascular and Endovascular Surgery, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Paul Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford Immunology, Stanford, USA
| | - Jens W Fischer
- Institut für Pharmakologie und Klinische Pharmakologie, University Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany; CARID, Cardiovascular Research Institute Düsseldorf, University Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
159
|
Ding R, Gao W, He Z, Wu F, Chu Y, Wu J, Ma L, Liang C. Circulating CD4 +CXCR5 + T cells contribute to proinflammatory responses in multiple ways in coronary artery disease. Int Immunopharmacol 2017; 52:318-323. [PMID: 28985621 DOI: 10.1016/j.intimp.2017.09.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 09/23/2017] [Accepted: 09/27/2017] [Indexed: 01/18/2023]
Abstract
Coronary artery disease (CAD) is a common subtype of cardiovascular disease. The major contributing event is atherosclerosis, which is a progressive inflammatory condition resulting in the thickening of the arterial wall and the formation of atheromatous plaques. Recent evidence suggests that circulating CD4+CXCR5+ T cells can contribute to inflammatory reactions. In this study, the frequency, phenotype, and function of circulating CD4+CXCR5+ T cells in CAD patients were examined. Data showed that circulating CD4+CXCR5+ T cells in CAD patients were enriched with a PD-1+CCR7- subset, which was previously identified as the most potent in B cell help. The CD4+CXCR5+ T cells in CAD patients also secreted significantly higher levels of IFN-γ, IL-17A, and IL-21 than those from healthy controls. Depleting the PD-1+ population significantly reduced the cytokine secretion. Interestingly, the CD4+CXCR5+PD-1- T cells significantly upregulated PD-1 following anti-CD3/CD28 or SEB stimulation. CD4+CXCR5+ T cells from CAD patients also demonstrated more potent capacity to stimulate B cell inflammation than those from healthy individuals. The phosphorylation of STAT1 and STAT3 were significantly higher in B cells incubated with CD4+CXCR5+ T cells from CAD than controls. The IL-6 and IFN-γ expression were also significantly higher in B cells incubated with CD4+CXCR5+ T cells from CAD. Together, this study demonstrated that CAD patients presented a highly activated CD4+CXCR5+ T cell subset that could contribute to proinflammatory responses in multiple ways. The possibility of using CD4+CXCR5+ T cells as a therapeutic target should therefore be examined in CAD patients.
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
| | - Feng Wu
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Yang Chu
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Jie Wu
- Medical Department, The Maternal and Child Health Hospital of Jinan City, Jinan, Shandong 250001, China
| | - Lan Ma
- Department of Cardiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| | - Chun Liang
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China.
| |
Collapse
|
160
|
Meeuwsen JAL, van Duijvenvoorde A, Gohar A, Kozma MO, van de Weg SM, Gijsberts CM, Haitjema S, Björkbacka H, Fredrikson GN, de Borst GJ, den Ruijter HM, Pasterkamp G, Binder CJ, Hoefer IE, de Jager SCA. High Levels of (Un)Switched Memory B Cells Are Associated With Better Outcome in Patients With Advanced Atherosclerotic Disease. J Am Heart Assoc 2017; 6:e005747. [PMID: 28882820 PMCID: PMC5634255 DOI: 10.1161/jaha.117.005747] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 07/07/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Atherosclerosis is an inflammatory lipid disorder and the main underlying pathology of acute ischemic events. Despite a vast amount of data from murine atherosclerosis models, evidence of B-cell involvement in human atherosclerotic disease is limited. We therefore investigated the association of circulating B-cell subtypes with the occurrence of secondary cardiovascular events in advanced atherosclerotic disease. METHODS AND RESULTS This cohort study consists of 168 patients who were included in the Athero-Express biobank between 2009 and 2011. Before surgery, peripheral blood mononuclear cells were isolated and stored in liquid nitrogen. After gentle thawing of the peripheral blood mononuclear cells, different B-cell subtypes including naïve, (un)switched memory, and CD27+CD43+ B1-like B cells, were analyzed by flow cytometry. Univariable and multivariable Cox proportional hazard models were used to analyze associations between B-cell subtypes, circulating antibodies and secondary cardiovascular manifestations during the 3-year follow-up period. Mean age was 70.1±9.6 years, males represented 62.8% of the population, and 54 patients had secondary manifestations during follow-up. High numbers of unswitched memory cells were protective against secondary outcome (hazard ratio, 0.30 [95% CI, 0.13-0.69]; P<0.01). Similar results were obtained for the switched memory cells that also showed to be protective against secondary outcome (hazard ratio, 0.33 [95% CI, 0.14-0.77]; P=0.01). CONCLUSIONS A high number of (un)switched memory B cells is associated with better outcome following carotid artery endarterectomy. These findings suggest a potential role for B-cell subsets in prediction and prevention of secondary cardiovascular events in patients with atherosclerosis.
Collapse
Affiliation(s)
- John A L Meeuwsen
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Amerik van Duijvenvoorde
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Aisha Gohar
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Maria O Kozma
- Department of Laboratory Medicine, Medical University of Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Sander M van de Weg
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Crystel M Gijsberts
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Saskia Haitjema
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Harry Björkbacka
- Department of Clinical Sciences, Skåne University Hospital Malmö, Lund University, Malmö, Sweden
| | - Gunilla N Fredrikson
- Department of Clinical Sciences, Skåne University Hospital Malmö, Lund University, Malmö, Sweden
| | - Gert J de Borst
- Department of Vascular Surgery, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Hester M den Ruijter
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Gerard Pasterkamp
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
- Laboratory for Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Imo E Hoefer
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
- Laboratory for Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Saskia C A de Jager
- Laboratory for Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
161
|
Sánchez-Trujillo L, Vázquez-Garza E, Castillo EC, García-Rivas G, Torre-Amione G. Role of Adaptive Immunity in the Development and Progression of Heart Failure: New Evidence. Arch Med Res 2017; 48:1-11. [PMID: 28577862 DOI: 10.1016/j.arcmed.2016.12.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 12/13/2016] [Indexed: 12/18/2022]
Abstract
Heart failure (HF) is considered the endpoint of a variety of cardiac diseases, which are the leading cause of death in adults and considered a growing pandemic worldwide. Independent of the initial form of cardiac injury, there is evidence linking the involvement of the immune system. In HF there is evidence of the participation of TH1, and TH17 cells, which account for sustained pathological chronic inflammation, cell migration, and the induction of specific pathological phenotypes of mononuclear cells. Of equal or even higher relevance are the B lymphocyte activation mechanisms that include production of pro-inflammatory cytokines, chemokines, and cardiac autoantibodies with or without activation of the complement proteins. Both of these unbalanced T- and B-cell pathways of the adaptive immune system are associated with cardiomyocyte death and tissue remodeling by fibrosis leading to a dysfunctional heart. At this time, therapy with neutralizing antibodies and the use of anti-cytokine immunomodulators to counteract the immune system effects have reached a plateau of mixed results in clinical trials. Nevertheless, recent evidence showed promising results in animal models that suggest that modulation of the adaptive immune system cells more than some of their effector molecules could have benefits in HF patients. This review summarizes the role of the adaptive immunity cells in HF, considering the sustained activation of adaptive immune system as a potential contributor to disease progression in humans and experimental models where its regulation provides a new therapeutic target.
Collapse
Affiliation(s)
- Luis Sánchez-Trujillo
- Cátedra de Cardiología y Medicina Vascular, Escuela de Medicina, Tecnologico de Monterrey, Monterrey, México; Hospital General de Zona No. 4, Instituto Mexicano del Seguro Social, México City, Mexico
| | - Eduardo Vázquez-Garza
- Cátedra de Cardiología y Medicina Vascular, Escuela de Medicina, Tecnologico de Monterrey, Monterrey, México
| | - Elena C Castillo
- Cátedra de Cardiología y Medicina Vascular, Escuela de Medicina, Tecnologico de Monterrey, Monterrey, México
| | - Gerardo García-Rivas
- Cátedra de Cardiología y Medicina Vascular, Escuela de Medicina, Tecnologico de Monterrey, Monterrey, México; Centro de Investigación Biomédica, Hospital Zambrano Hellion, Tecnologico de Monterrey, San Pedro Garza-García, México.
| | - Guillermo Torre-Amione
- Cátedra de Cardiología y Medicina Vascular, Escuela de Medicina, Tecnologico de Monterrey, Monterrey, México; Centro de Investigación Biomédica, Hospital Zambrano Hellion, Tecnologico de Monterrey, San Pedro Garza-García, México; Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, Texas
| |
Collapse
|
162
|
Gu XL, He H, Lin L, Luo GX, Wen YF, Xiang DC, Qiu J. Tim-1+B cells suppress T cell interferon-gamma production and promote Foxp3 expression, but have impaired regulatory function in coronary artery disease. APMIS 2017; 125:872-879. [PMID: 28736945 DOI: 10.1111/apm.12729] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 05/03/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Xiao-Long Gu
- Department of Cardiology; Guangzhou Liuhuaqiao Hospital; Guangzhou China
| | - Huan He
- Department of Anesthesiology; Guangzhou Liuhuaqiao Hospital; Guangzhou China
| | - Lin Lin
- Department of Cardiology; Guangzhou Liuhuaqiao Hospital; Guangzhou China
| | - Guo-Xin Luo
- Department of Ultrasound; Guangzhou Liuhuaqiao Hospital; Guangzhou China
| | - Yan-Fei Wen
- Department of Cardiology; Guangzhou Liuhuaqiao Hospital; Guangzhou China
| | - Ding-Cheng Xiang
- Department of Cardiology; Guangzhou Liuhuaqiao Hospital; Guangzhou China
| | - Jian Qiu
- Department of Cardiology; Guangzhou Liuhuaqiao Hospital; Guangzhou China
| |
Collapse
|
163
|
Sage AP, Nus M, Bagchi Chakraborty J, Tsiantoulas D, Newland SA, Finigan AJ, Masters L, Binder CJ, Mallat Z. X-Box Binding Protein-1 Dependent Plasma Cell Responses Limit the Development of Atherosclerosis. Circ Res 2017; 121:270-281. [DOI: 10.1161/circresaha.117.310884] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 06/14/2017] [Accepted: 06/15/2017] [Indexed: 02/06/2023]
Abstract
Rationale:
Diverse B cell responses and functions may be involved in atherosclerosis. Protective antibody responses, such as those against oxidized lipid epitopes, are thought to mainly derive from T cell-independent innate B cell subsets. In contrast, both pathogenic and protective roles have been associated with T cell-dependent antibodies, and their importance in both humans and mouse models is still unclear.
Objective:
To specifically target antibody production by plasma cells and determine the impact on atherosclerotic plaque development in mice with and without CD4+ T cells.
Methods and Results:
We combined a model of specific antibody deficiency, B cell-specific CD79a-
Cre
x XBP1 (X-box binding protein-1) floxed mice (XBP1-conditional knockout), with antibody-mediated depletion of CD4+ T cells. Ldlr knockout mice transplanted with XBP1-conditional knockout (or wild-type control littermate) bone marrow were fed western diet for 8 weeks with or without anti-CD4 depletion. All groups had similar levels of serum cholesterol. In Ldlr/XBP1-conditional knockout mice, serum levels of IgG, IgE, and IgM were significantly attenuated, and local antibody deposition in atherosclerotic plaque was absent. Antibody deficiency significantly accelerated atherosclerosis at both the aortic root and aortic arch. T cell and monocyte responses were not modulated, but necrotic core size was greater, even when adjusting for plaque size, and collagen deposition significantly lower. Anti-CD4 depletion in Ldlr/wild-type mice led to a decrease of serum IgG1 and IgG2c but not IgG3, as well as decreased IgM, associated with increased atherosclerosis and necrotic cores, and a decrease in plaque collagen. The combination of antibody deficiency and anti-CD4 depletion has no additive effects on aortic root atherosclerosis.
Conclusions:
The endogenous T cell-dependent humoral response can be protective. This has important implications for novel vaccine strategies for atherosclerosis and in understanding the impacts of immunotherapies used in patients at high risk for cardiovascular disease.
Collapse
Affiliation(s)
- Andrew P. Sage
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Meritxell Nus
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Jayashree Bagchi Chakraborty
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Dimitrios Tsiantoulas
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Stephen A. Newland
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Alison J. Finigan
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Leanne Masters
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Christoph J. Binder
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Ziad Mallat
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.P.S., M.N., J.B.C., S.A.N., A.J.F., L.M., Z.M.); Department of Laboratory Medicine, (D.T., C.J.B.); Medical University of Vienna, Austria (D.T., C.J.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (D.T., C.J.B.); INSERM U970, Paris Cardiovascular Research Center, France (Z.M.); and Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| |
Collapse
|
164
|
Joffre J, Potteaux S, Zeboudj L, Loyer X, Boufenzer A, Laurans L, Esposito B, Vandestienne M, de Jager SCA, Hénique C, Zlatanova I, Taleb S, Bruneval P, Tedgui A, Mallat Z, Gibot S, Ait-Oufella H. Genetic and Pharmacological Inhibition of TREM-1 Limits the Development of Experimental Atherosclerosis. J Am Coll Cardiol 2017; 68:2776-2793. [PMID: 28007141 DOI: 10.1016/j.jacc.2016.10.015] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 09/12/2016] [Accepted: 10/04/2016] [Indexed: 01/19/2023]
Abstract
BACKGROUND Innate immune responses activated through myeloid cells contribute to the initiation, progression, and complications of atherosclerosis in experimental models. However, the critical upstream pathways that link innate immune activation to foam cell formation are still poorly identified. OBJECTIVES This study sought to investigate the hypothesis that activation of the triggering receptor expressed on myeloid cells (TREM-1) plays a determinant role in macrophage atherogenic responses. METHODS After genetically invalidating Trem-1 in chimeric Ldlr-/-Trem-1-/- mice and double knockout ApoE-/-Trem-1-/- mice, we pharmacologically inhibited Trem-1 using LR12 peptide. RESULTS Ldlr-/- mice reconstituted with bone marrow deficient for Trem-1 (Trem-1-/-) showed a strong reduction of atherosclerotic plaque size in both the aortic sinus and the thoracoabdominal aorta, and were less inflammatory compared to plaques of Trem-1+/+ chimeric mice. Genetic invalidation of Trem-1 led to alteration of monocyte recruitment into atherosclerotic lesions and inhibited toll-like receptor 4 (TLR 4)-initiated proinflammatory macrophage responses. We identified a critical role for Trem-1 in the upregulation of cluster of differentiation 36 (CD36), thereby promoting the formation of inflammatory foam cells. Genetic invalidation of Trem-1 in ApoE-/-/Trem-1-/- mice or pharmacological blockade of Trem-1 in ApoE-/- mice using LR-12 peptide also significantly reduced the development of atherosclerosis throughout the vascular tree, and lessened plaque inflammation. TREM-1 was expressed in human atherosclerotic lesions, mainly in lipid-rich areas with significantly higher levels of expression in atheromatous than in fibrous plaques. CONCLUSIONS We identified TREM-1 as a major upstream proatherogenic receptor. We propose that TREM-1 activation orchestrates monocyte/macrophage proinflammatory responses and foam cell formation through coordinated and combined activation of CD36 and TLR4. Blockade of TREM-1 signaling may constitute an attractive novel and double-hit approach for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Jeremie Joffre
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Stephane Potteaux
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Lynda Zeboudj
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Xavier Loyer
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | | | - Ludivine Laurans
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Bruno Esposito
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Marie Vandestienne
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Saskia C A de Jager
- Laboratory for Experimental Cardiology, University Medical Center, Utrecht, the Netherlands
| | - Carole Hénique
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Ivana Zlatanova
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Soraya Taleb
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Patrick Bruneval
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Department of Anatomopathology, Hôpital Européen Georges Pompidou, Assistance Publique-Hopitaux de Paris, Paris, France
| | - Alain Tedgui
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Ziad Mallat
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Sebastien Gibot
- INSERM Unité mixte de Recherche-S1116, Faculté de Médecine, Université de Lorraine, Medical Intensive Care Unit, Hôpital Central, Nancy, France
| | - Hafid Ait-Oufella
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Medical Intensive Care Unit, Hôpital Saint-Antoine, Assistance Publique-Hopitaux de Paris, Université Pierre-et-Marie Curie, Paris, France.
| |
Collapse
|
165
|
Abstract
Angiotensin II (AngII) promotes hypertension, atherogenesis, vascular aneurysm and impairs post-ischemic cardiac remodeling through concerted roles on vascular cells, monocytes and T lymphocytes. However, the role of AngII in B lymphocyte responses is largely unexplored. Here, we show that chronic B cell depletion (Baffr deficiency) significantly reduces atherosclerosis in Apoe−/− mice infused with AngII. While adoptive transfer of B cells in Apoe−/−/Baffr−/− mice reversed atheroprotection in the absence of AngII, infusion of AngII in B cell replenished Apoe−/−/Baffr−/− mice unexpectedly prevented the progression of atherosclerosis. Atheroprotection observed in these mice was associated with a significant increase in regulatory CD1dhiCD5+ B cells, which produced high levels of interleukin (IL)-10 (B10 cells). Replenishment of Apoe−/−/Baffr−/− mice with Il10−/− B cells reversed AngII-induced B cell-dependent atheroprotection, thus highlighting a protective role of IL-10+ regulatory B cells in this setting. Transfer of AngII type 1A receptor deficient (Agtr1a−/−) B cells into Apoe−/−/Baffr−/− mice substantially reduced the production of IL-10 by B cells and prevented the AngII-dependent atheroprotective B cell phenotype. Consistent with the in vivo data, AngII synergized with BAFF to induce IL-10 production by B cells in vitro via AngII type 1A receptor. Our data demonstrate a previously unknown synergy between AngII and BAFF in inducing IL-10 production by B cells, resulting in atheroprotection.
Collapse
|
166
|
Ponnuswamy P, Joffre J, Herbin O, Esposito B, Laurans L, Binder CJ, Tedder TF, Zeboudj L, Loyer X, Giraud A, Zhang Y, Tedgui A, Mallat Z, Ait-Oufella H. Angiotensin II synergizes with BAFF to promote atheroprotective regulatory B cells. Sci Rep 2017. [PMID: 28646220 DOI: 10.1038/s41598‐017‐04438‐6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Angiotensin II (AngII) promotes hypertension, atherogenesis, vascular aneurysm and impairs post-ischemic cardiac remodeling through concerted roles on vascular cells, monocytes and T lymphocytes. However, the role of AngII in B lymphocyte responses is largely unexplored. Here, we show that chronic B cell depletion (Baffr deficiency) significantly reduces atherosclerosis in Apoe -/- mice infused with AngII. While adoptive transfer of B cells in Apoe -/- /Baffr -/- mice reversed atheroprotection in the absence of AngII, infusion of AngII in B cell replenished Apoe -/- /Baffr -/- mice unexpectedly prevented the progression of atherosclerosis. Atheroprotection observed in these mice was associated with a significant increase in regulatory CD1dhiCD5+ B cells, which produced high levels of interleukin (IL)-10 (B10 cells). Replenishment of Apoe -/- /Baffr -/- mice with Il10 -/- B cells reversed AngII-induced B cell-dependent atheroprotection, thus highlighting a protective role of IL-10+ regulatory B cells in this setting. Transfer of AngII type 1A receptor deficient (Agtr1a -/-) B cells into Apoe -/- /Baffr -/- mice substantially reduced the production of IL-10 by B cells and prevented the AngII-dependent atheroprotective B cell phenotype. Consistent with the in vivo data, AngII synergized with BAFF to induce IL-10 production by B cells in vitro via AngII type 1A receptor. Our data demonstrate a previously unknown synergy between AngII and BAFF in inducing IL-10 production by B cells, resulting in atheroprotection.
Collapse
Affiliation(s)
- Padmapriya Ponnuswamy
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Jeremie Joffre
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Olivier Herbin
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Bruno Esposito
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Ludivine Laurans
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Christoph J Binder
- Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences and Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Thomas F Tedder
- Department of Immunology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Lynda Zeboudj
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Xavier Loyer
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Andreas Giraud
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Yujiao Zhang
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Alain Tedgui
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Ziad Mallat
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France.,Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 2QQ, England, UK
| | - Hafid Ait-Oufella
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France. .,Assistance Publique-Hôpitaux de Paris, Saint-Antoine Hospital, Université Pierre-et-Marie-Curie, Paris, France.
| |
Collapse
|
167
|
Temmerman L, Westra MM, Bot I, van Vlijmen BJM, van Bree N, Bot M, Habets KLL, Keulers TGH, van der Vlag J, Cotter TG, van Berkel TJC, Biessen EAL. Leukocyte Bim deficiency does not impact atherogenesis in ldlr -/- mice, despite a pronounced induction of autoimmune inflammation. Sci Rep 2017; 7:3086. [PMID: 28596542 PMCID: PMC5465223 DOI: 10.1038/s41598-017-02771-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 04/19/2017] [Indexed: 12/11/2022] Open
Abstract
Proapoptotic Bcl-2 family member Bim is particularly relevant for deletion of autoreactive and activated T and B cells, implicating Bim in autoimmunity. As atherosclerosis is a chronic inflammatory process with features of autoimmune disease, we investigated the impact of hematopoietic Bim deficiency on plaque formation and parameters of plaque stability. Bim -/- or wild type bone marrow transplanted ldlr -/- mice were fed a Western type diet (WTD) for 5 or 10 weeks, after which they were immunophenotyped and atherosclerotic lesions were analyzed. Bim -/- transplanted mice displayed splenomegaly and overt lymphocytosis. CD4+ and CD8+ T cells were more activated (increased CD69 and CD71 expression, increased interferon gamma production). B cells were elevated by 147%, with a shift towards the pro-atherogenic IgG-producing B2 cell phenotype, resulting in a doubling of anti-oxLDL IgG1 antibody titers in serum of bim -/- mice. Bim -/- mice displayed massive intraplaque accumulation of Ig complexes and of lesional T cells, although this did not translate in changes in plaque size or stability features (apoptotic cell and macrophage content). The surprising lack in plaque phenotype despite the profound pro-atherogenic immune effects may be attributable to the sharp reduction of serum cholesterol levels in WTD fed bim -/- mice.
Collapse
Affiliation(s)
- Lieve Temmerman
- Experimental Vascular Pathology, Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands.
| | - Marijke M Westra
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Ilze Bot
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Bart J M van Vlijmen
- Department of Thrombosis Hemostasis, Leiden University Medical Centre, Leiden, The Netherlands
| | - Niek van Bree
- Experimental Vascular Pathology, Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Martine Bot
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Kim L L Habets
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Tom G H Keulers
- Department of Radiotherapy (MAASTRO)/GROW, School for Developmental Biology and Oncology, Maastricht University, Maastricht, The Netherlands
| | - Johan van der Vlag
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Thomas G Cotter
- Cell Development and Disease Laboratory, Department of Biochemistry, Biosciences Research Institute, University College Cork, Cork, Ireland
| | - Theo J C van Berkel
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Erik A L Biessen
- Experimental Vascular Pathology, Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH, Aachen, Germany
| |
Collapse
|
168
|
Nus M, Sage AP, Lu Y, Masters L, Lam BYH, Newland S, Weller S, Tsiantoulas D, Raffort J, Marcus D, Finigan A, Kitt L, Figg N, Schirmbeck R, Kneilling M, Yeo GSH, Binder CJ, de la Pompa JL, Mallat Z. Marginal zone B cells control the response of follicular helper T cells to a high-cholesterol diet. Nat Med 2017; 23:601-610. [DOI: 10.1038/nm.4315] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 03/06/2017] [Indexed: 12/12/2022]
|
169
|
Abstract
Cardiovascular disease is the leading cause of death worldwide, both in the general population and among patients with chronic kidney disease (CKD). In most cases, the underlying cause of the cardiovascular event is atherosclerosis - a chronic inflammatory disease. CKD accelerates atherosclerosis via augmentation of inflammation, perturbation of lipid metabolism, and other mechanisms. In the artery wall, subendothelial retention of plasma lipoproteins triggers monocyte-derived macrophages and T helper type 1 (TH1) cells to form atherosclerotic plaques. Inflammation is initiated by innate immune reactions to modified lipoproteins and is perpetuated by TH1 cells that react to autoantigens from the apolipoprotein B100 protein of LDL. Other T cells are also active in atherosclerotic lesions; regulatory T cells inhibit pathological inflammation, whereas TH17 cells can promote plaque fibrosis. The slow build-up of atherosclerotic plaques is asymptomatic, but plaque rupture or endothelial erosion can induce thrombus formation, leading to myocardial infarction or ischaemic stroke. Targeting risk factors for atherosclerosis has reduced mortality, but a need exists for novel therapies to stabilize plaques and to treat arterial inflammation. Patients with CKD would likely benefit from such preventive measures.
Collapse
Affiliation(s)
- Anton Gisterå
- Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, SE-17176 Stockholm, Sweden
| | - Göran K Hansson
- Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, SE-17176 Stockholm, Sweden
| |
Collapse
|
170
|
Rademakers T, van der Vorst EPC, Daissormont ITMN, Otten JJT, Theodorou K, Theelen TL, Gijbels M, Anisimov A, Nurmi H, Lindeman JHN, Schober A, Heeneman S, Alitalo K, Biessen EAL. Adventitial lymphatic capillary expansion impacts on plaque T cell accumulation in atherosclerosis. Sci Rep 2017; 7:45263. [PMID: 28349940 PMCID: PMC5368662 DOI: 10.1038/srep45263] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 02/07/2017] [Indexed: 02/07/2023] Open
Abstract
During plaque progression, inflammatory cells progressively accumulate in the adventitia, paralleled by an increased presence of leaky vasa vasorum. We here show that next to vasa vasorum, also the adventitial lymphatic capillary bed is expanding during plaque development in humans and mouse models of atherosclerosis. Furthermore, we investigated the role of lymphatics in atherosclerosis progression. Dissection of plaque draining lymph node and lymphatic vessel in atherosclerotic ApoE-/- mice aggravated plaque formation, which was accompanied by increased intimal and adventitial CD3+ T cell numbers. Likewise, inhibition of VEGF-C/D dependent lymphangiogenesis by AAV aided gene transfer of hVEGFR3-Ig fusion protein resulted in CD3+ T cell enrichment in plaque intima and adventitia. hVEGFR3-Ig gene transfer did not compromise adventitial lymphatic density, pointing to VEGF-C/D independent lymphangiogenesis. We were able to identify the CXCL12/CXCR4 axis, which has previously been shown to indirectly activate VEGFR3, as a likely pathway, in that its focal silencing attenuated lymphangiogenesis and augmented T cell presence. Taken together, our study not only shows profound, partly CXCL12/CXCR4 mediated, expansion of lymph capillaries in the adventitia of atherosclerotic plaque in humans and mice, but also is the first to attribute an important role of lymphatics in plaque T cell accumulation and development.
Collapse
Affiliation(s)
- Timo Rademakers
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | - Emiel P C van der Vorst
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands.,Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Isabelle T M N Daissormont
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | - Jeroen J T Otten
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | - Kosta Theodorou
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | - Thomas L Theelen
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | - Marion Gijbels
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands.,Department of Molecular Genetics, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands.,Department of Medical Biochemistry, Academic Medical Center, Amsterdam, the Netherlands
| | - Andrey Anisimov
- Wihuri Research Institute, University of Helsinki, Helsinki, Finland
| | - Harri Nurmi
- Wihuri Research Institute, University of Helsinki, Helsinki, Finland
| | - Jan H N Lindeman
- Departments of Vascular Surgery and Transplantation Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Andreas Schober
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sylvia Heeneman
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | - Kari Alitalo
- Wihuri Research Institute, University of Helsinki, Helsinki, Finland
| | - Erik A L Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands.,Institute for Molecular Cardiovascular Research, RWTH Aachen, Germany
| |
Collapse
|
171
|
Srikakulapu P, McNamara CA. B cells and atherosclerosis. Am J Physiol Heart Circ Physiol 2017; 312:H1060-H1067. [PMID: 28314764 DOI: 10.1152/ajpheart.00859.2016] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 03/08/2017] [Accepted: 03/13/2017] [Indexed: 12/14/2022]
Abstract
B cells have emerged as important immune cells in cardiovascular disease. Initial studies have suggested that B cells protect against atherosclerosis development. However, subsequent studies demonstrating aggravation of atherosclerosis by B-2 cells have shed light on the subset-dependent effects of B cells. Here, we review the literature that has led to our current understanding of B cell regulation of atherosclerosis, touching on the importance of subsets, local regulation, human translation, and therapeutic potential.
Collapse
Affiliation(s)
| | - Coleen A McNamara
- Cardiovascular Research Center, Charlottesville, Virginia; and.,Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
172
|
Echeverri Tirado LC, Yassin LM. B cells interactions in lipid immune responses: implications in atherosclerotic disease. Lipids Health Dis 2017; 16:30. [PMID: 28166809 PMCID: PMC5295187 DOI: 10.1186/s12944-016-0390-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 12/14/2016] [Indexed: 12/26/2022] Open
Abstract
Atherosclerosis is considered as an inflammatory and chronic disorder with an important immunologic component, which underlies the majority of cardiovascular diseases; condition that belongs to a group of noncommunicable diseases that to date and despite of prevention and treatment approaches, they remain as the main cause of death worldwide, with 17.5 million of deaths every year. The impact of lipids in human health and disease is taking center stage in research, due to lipotoxicity explained by elevated concentration of circulating lipids, in addition to altered adipose tissue metabolism, and aberrant intracellular signaling. Immune response and metabolic regulation are highly integrated systems and the proper function of each one is dependent on the other. B lymphocytes express a variety of receptors that can recognize foreign, endogenous or modified self-antigens, among them oxidized low density lipoproteins, which are the main antigens in atherosclerosis. Mechanisms of B cells to recognize, remove and present lipids are not completely clear. However, it has been reported that B cell can recognize/remove lipids through a range of receptors, such as LDLR, CD1d, FcR and SR, which might have an atheroprotector or proatherogenic role during the course of atherosclerotic disease. Pertinent literature related to these receptors was examined to inform the present conclusions.
Collapse
Affiliation(s)
| | - Lina M Yassin
- Facultad de Medicina, Universidad CES, Calle 10 A Nro. 22-04, Medellín, Colombia.
| |
Collapse
|
173
|
Sage AP, Mallat Z. Readapting the adaptive immune response - therapeutic strategies for atherosclerosis. Br J Pharmacol 2017; 174:3926-3939. [PMID: 28052311 DOI: 10.1111/bph.13700] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/13/2016] [Accepted: 12/20/2016] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases remain a major global health issue, with the development of atherosclerosis as a major underlying cause. Our treatment of cardiovascular disease has improved greatly over the past three decades, but much remains to be done reduce disease burden. Current priorities include reducing atherosclerosis advancement to clinically significant stages and preventing plaque rupture or erosion. Inflammation and involvement of the adaptive immune system influences all these aspects and therefore is one focus for future therapeutic development. The atherosclerotic vascular wall is now recognized to be invaded from both sides (arterial lumen and adventitia), for better or worse, by the adaptive immune system. Atherosclerosis is also affected at several stages by adaptive immune responses, overall providing many opportunities to target these responses and to reduce disease progression. Protective influences that may be defective in diseased individuals include humoral responses to modified LDL and regulatory T cell responses. There are many strategies in development to boost these pathways in humans, including vaccine-based therapies. The effects of various existing adaptive immune targeting therapies, such as blocking critical co-stimulatory pathways or B cell depletion, on cardiovascular disease are beginning to emerge with important consequences for both autoimmune disease patients and the potential for wider use of such therapies. Entering the translation phase for adaptive immune targeting therapies is an exciting and promising prospect. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- Andrew P Sage
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK.,INSERM U970, Paris Cardiovascular Research Center, Paris, France, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
174
|
Taleb S. Inflammation in atherosclerosis. Arch Cardiovasc Dis 2016; 109:708-715. [DOI: 10.1016/j.acvd.2016.04.002] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/08/2016] [Accepted: 04/11/2016] [Indexed: 10/21/2022]
|
175
|
Deng J, Lü S, Liu H, Liu B, Jiang C, Xu Q, Feng J, Wang X. Homocysteine Activates B Cells via Regulating PKM2-Dependent Metabolic Reprogramming. THE JOURNAL OF IMMUNOLOGY 2016; 198:170-183. [PMID: 27903739 DOI: 10.4049/jimmunol.1600613] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 10/24/2016] [Indexed: 01/16/2023]
Abstract
The overactivation of immune cells plays an important role in the pathogenesis of hyperhomocysteinemia (HHcy)-accelerated atherosclerosis. Homocysteine (Hcy) activates B cell proliferation and Ab secretion; however, the underlying mechanisms for these effects remain largely unknown. Metabolic reprogramming is critical for lymphocyte activation and effector function. In this study, we showed that Hcy-activated B cells displayed an increase in both oxidative phosphorylation and glycolysis, with a tendency to shift toward the latter, as well as an accumulation of intermediates in the pentose phosphate pathway, to provide energy and biosynthetic substrates for cell growth and function. Mechanistically, Hcy increased both the protein expression and glycolytic enzyme activity of the pyruvate kinase muscle isozyme 2 (PKM2) in B cells, whereas the PKM2 inhibitor shikonin restored Hcy-induced metabolic changes, as well as B cell proliferation and Ab secretion both in vivo and in vitro, indicating that PKM2 plays a critical role in metabolic reprogramming in Hcy-activated B cells. Further investigation revealed that the Akt-mechanistic target of rapamycin signaling pathway was involved in this process, as the mechanistic target of rapamycin inhibitor rapamycin inhibited Hcy-induced changes in PKM2 enzyme activity and B cell activation. Notably, shikonin treatment effectively attenuated HHcy-accelerated atherosclerotic lesion formation in apolipoprotein E-deficient mice. In conclusion, our results demonstrate that PKM2 is required to support metabolic reprogramming for Hcy-induced B cell activation and function, and it might serve as a critical regulator in HHcy-accelerated initiation of atherosclerosis.
Collapse
Affiliation(s)
- Jiacheng Deng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, People's Republic of China; and
| | - Silin Lü
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, People's Republic of China; and
| | - Huiying Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, People's Republic of China; and
| | - Bo Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, People's Republic of China; and
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, People's Republic of China; and
| | - Qingbo Xu
- Cardiovascular Division, British Heart Foundation Centre for Vascular Regeneration, King's College London, London SE5 9NU, United Kingdom
| | - Juan Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, People's Republic of China; and
| | - Xian Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, People's Republic of China; and
| |
Collapse
|
176
|
Hosseini H, Li Y, Kanellakis P, Tay C, Cao A, Liu E, Peter K, Tipping P, Toh BH, Bobik A, Kyaw T. Toll-Like Receptor (TLR)4 and MyD88 are Essential for Atheroprotection by Peritoneal B1a B Cells. J Am Heart Assoc 2016; 5:e002947. [PMID: 27930350 PMCID: PMC5210362 DOI: 10.1161/jaha.115.002947] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 10/05/2016] [Indexed: 01/16/2023]
Abstract
BACKGROUND We previously identified peritoneal B1a cells that secrete natural IgM as a key atheroprotective B cell subset. However, the molecules that activate atheroprotective B1a cells are unknown. Here, we investigated whether Toll-like receptors (TLRs) TLR2, TLR4, and TLR9 expressed by B1a cells are required for IgM-mediated atheroprotection. METHODS AND RESULTS We adoptively transferred B1a cells from wild-type mice or from mice deficient in TLR2, TLR4, TLR9, or myeloid differentiation primary response 88 (MyD88) into ApoE-/- mice depleted of peritoneal B1a cells by splenectomy and fed a high-fat diet for 8 weeks. Elevations in plasma total, anti-oxLDL (oxidized low-density lipoprotein), anti-leukocyte, anti-CD3, anti-CD8, and anti-CD4 IgMs in atherosclerotic mice required B1a cells expressing TLR4 and MyD88, indicating a critical role for TLR4-MyD88 signaling for IgM secretion. Suppression of atherosclerosis was also critically dependent on B1a cells expressing TLR4-MyD88. Atherosclerosis suppression was associated not only with reductions in lesion apoptotic cells, necrotic cores, and oxLDL, but also with reduced lesion CD4+ and CD8+ T cells. Transforming growth factor beta 1 (TGF-β1) expression, including macrophages expressing TGF-β1, was increased, consistent with increased IgM-mediated phagocytosis of apoptotic cells by macrophages. Reductions in lesion inflammatory cytokines tumor necrosis factor alpha (TNF-α), interleukin (IL) 1β, and IL-18 were consistent with augmented TGF-β1 expression. CONCLUSIONS TLR4-MyD88 expression on B1a cells is critical for their IgM-dependent atheroprotection that not only reduced lesion apoptotic cells and necrotic cores, but also decreased CD4 and CD8 T-cell infiltrates and augmented TGF-β1 expression accompanied by reduced lesion inflammatory cytokines TNF-α, IL-1β, and IL-18.
Collapse
Affiliation(s)
- Hamid Hosseini
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
- Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School, Clayton, Australia
| | - Yi Li
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
- Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School, Clayton, Australia
| | | | - Christopher Tay
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
- Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School, Clayton, Australia
| | - Anh Cao
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
| | - Edgar Liu
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
| | - Karlheinz Peter
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
- Department of Immunology, Faculty of Medicine, Nursing and Health Sciences Monash University, Clayton, Australia
| | - Peter Tipping
- Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School, Clayton, Australia
| | - Ban-Hock Toh
- Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School, Clayton, Australia
| | - Alex Bobik
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
- Department of Immunology, Faculty of Medicine, Nursing and Health Sciences Monash University, Clayton, Australia
| | - Tin Kyaw
- BakerIDI heart and Diabetes Institute, Melbourne, Australia
- Department of Medicine, Centre for Inflammatory Diseases, Southern Clinical School, Clayton, Australia
| |
Collapse
|
177
|
Jackson SW, Scharping NE, Jacobs HM, Wang S, Chait A, Rawlings DJ. Cutting Edge: BAFF Overexpression Reduces Atherosclerosis via TACI-Dependent B Cell Activation. THE JOURNAL OF IMMUNOLOGY 2016; 197:4529-4534. [PMID: 27837104 DOI: 10.4049/jimmunol.1601198] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 10/13/2016] [Indexed: 12/27/2022]
Abstract
Patients with systemic lupus erythematosus exhibit accelerated atherosclerosis, a chronic inflammatory disease of the arterial wall. The impact of B cells in atherosclerosis is controversial, with both protective and pathogenic roles described. For example, natural IgM binding conserved oxidized lipid epitopes protect against atherosclerosis, whereas anti-oxidized low-density lipoprotein (oxLDL) IgG likely promotes disease. Because BAFF promotes B cell class-switch recombination and humoral autoimmunity, we hypothesized that excess BAFF would accelerate atherosclerosis. In contrast, BAFF overexpression markedly reduced hypercholesterolemia and atherosclerosis in hyperlipidemic mice. BAFF-mediated atheroprotection required B cells and was associated with increased protective anti-oxLDL IgM. Surprisingly, high-titer anti-oxLDL IgM production and reduced atherosclerosis was dependent on the BAFF family receptor transmembrane activator and CAML interactor. In summary, we identified a novel role for B cell-specific, BAFF-dependent transmembrane activator and CAML interactor signals in atherosclerosis pathogenesis, of particular relevance to the use of BAFF-targeted therapies in systemic lupus erythematosus.
Collapse
Affiliation(s)
- Shaun W Jackson
- Seattle Children's Research Institute, Seattle, WA 98101.,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195
| | | | - Holly M Jacobs
- Seattle Children's Research Institute, Seattle, WA 98101
| | - Shari Wang
- Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195; and
| | - Alan Chait
- Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195; and
| | - David J Rawlings
- Seattle Children's Research Institute, Seattle, WA 98101; .,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195.,Department of Immunology, University of Washington School of Medicine, Seattle, WA 98195
| |
Collapse
|
178
|
Bernelot Moens SJ, van Leuven SI, Zheng KH, Havik SR, Versloot MV, van Duivenvoorde LM, Hahne M, Stroes ESG, Baeten DL, Hamers AAJ. Impact of the B Cell Growth Factor APRIL on the Qualitative and Immunological Characteristics of Atherosclerotic Plaques. PLoS One 2016; 11:e0164690. [PMID: 27820817 PMCID: PMC5098816 DOI: 10.1371/journal.pone.0164690] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 09/29/2016] [Indexed: 11/18/2022] Open
Abstract
Studies on the role of B lymphocytes in atherosclerosis development, have yielded contradictory results. Whereas B lymphocyte-deficiency aggravates atherosclerosis in mice; depletion of mature B lymphocytes reduces atherosclerosis. These observations led to the notion that distinct B lymphocyte subsets have different roles. B1a lymphocytes exert an atheroprotective effect, which has been attributed to secretion of IgM, which can be deposited in atherosclerotic lesions thereby reducing necrotic core formation. Tumor necrosis factor (TNF)-family member 'A Proliferation-Inducing Ligand' (APRIL, also known as TNFSF13) was previously shown to increase serum IgM levels in a murine model. In this study, we investigated the effect of APRIL overexpression on advanced lesion formation and composition, IgM production and B cell phenotype. We crossed APRIL transgenic (APRIL-Tg) mice with ApoE knockout (ApoE-/-) mice. After a 12-week Western Type Diet, ApoE-/-APRIL-Tg mice and ApoE-/- littermates showed similar increases in body weight and lipid levels. Histologic evaluation showed no differences in lesion size, stage or necrotic area. However, smooth muscle cell (α-actin stain) content was increased in ApoE-/-APRIL-Tg mice, implying more stable lesions. In addition, increases in both plaque IgM deposition and plasma IgM levels were found in ApoE-/-APRIL-Tg mice compared with ApoE-/- mice. Flow cytometry revealed a concomitant increase in peritoneal B1a lymphocytes in ApoE-/-APRIL-Tg mice. This study shows that ApoE-/-APRIL-Tg mice have increased oxLDL-specific serum IgM levels, potentially mediated via an increase in B1a lymphocytes. Although no differences in lesion size were found, transgenic ApoE-/-APRIL-Tg mice do show potential plaque stabilizing features in advanced atherosclerotic lesions.
Collapse
Affiliation(s)
| | - Sander I. van Leuven
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Kang H. Zheng
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Stefan R. Havik
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Miranda V. Versloot
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Leonie M. van Duivenvoorde
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology, Academic Medical Center, Amsterdam, The Netherlands
- Department of Experimental Immunology, Academic Medical Center, Amsterdam, The Netherlands
| | - Michael Hahne
- Institut de Génétique Moléculaire de Montpellier, Centre National de la Recherche Scientifique, Université de Montpellier, Montpellier, France
| | - Erik S. G. Stroes
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Dominique L. Baeten
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology, Academic Medical Center, Amsterdam, The Netherlands
- Department of Experimental Immunology, Academic Medical Center, Amsterdam, The Netherlands
| | - Anouk A. J. Hamers
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
179
|
Chen L, Ishigami T, Nakashima-Sasaki R, Kino T, Doi H, Minegishi S, Umemura S. Commensal Microbe-specific Activation of B2 Cell Subsets Contributes to Atherosclerosis Development Independently of Lipid Metabolism. EBioMedicine 2016; 13:237-247. [PMID: 27810309 PMCID: PMC5264349 DOI: 10.1016/j.ebiom.2016.10.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 10/06/2016] [Accepted: 10/19/2016] [Indexed: 01/11/2023] Open
Abstract
The relation between B2 cells and commensal microbes during atherosclerosis remains largely unexplored. Here we show that under hyperlipidemic conditions intestinal microbiota resulted in recruitment and ectopic activation of B2 cells in perivascular adipose tissue, followed by an increase in circulating IgG, promoting disease development. In contrast, disruption of the intestinal microbiota by a broad-spectrum antibiotic cocktail (AVNM) led to the attenuation of atherosclerosis by suppressing B2 cells, despite the persistence of serum lipid abnormalities. Furthermore, pharmacological depletion of B2 cells with an anti-B2-cell surface CD23 antibody also attenuated commensal microbe-induced atherosclerosis. Moreover, expression analysis of TLR-signaling-related genes in the activated B2 cell subsets, assessed using the Toll-Like Receptor Signaling Pathway RT2 Profiler PCR Array, confirmed activation of the B2-cell autoantibody-production axis, which was associated with an increased capacity of B2 cells to bind to intestinal microbiota. Together, our findings reveal the critical role of commensal microbe-specific activation of B2 cells in the development of atherogenesis through lipid metabolism-independent mechanisms.
Collapse
Affiliation(s)
- Lin Chen
- Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan
| | - Tomoaki Ishigami
- Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan.
| | - Rie Nakashima-Sasaki
- Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan
| | - Tabito Kino
- Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan
| | - Hiroshi Doi
- Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan
| | - Shintaro Minegishi
- Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan
| | - Satoshi Umemura
- Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan
| |
Collapse
|
180
|
Su KY, Watanabe A, Yeh CH, Kelsoe G, Kuraoka M. Efficient Culture of Human Naive and Memory B Cells for Use as APCs. THE JOURNAL OF IMMUNOLOGY 2016; 197:4163-4176. [PMID: 27815447 DOI: 10.4049/jimmunol.1502193] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 08/30/2016] [Indexed: 12/27/2022]
Abstract
The ability to culture and expand B cells in vitro has become a useful tool for studying human immunity. A limitation of current methods for human B cell culture is the capacity to support mature B cell proliferation. We developed a culture method to support the efficient activation and proliferation of naive and memory human B cells. This culture supports extensive B cell proliferation, with ∼103-fold increases following 8 d in culture and 106-fold increases when cultures are split and cultured for 8 more days. In culture, a significant fraction of naive B cells undergo isotype switching and differentiate into plasmacytes. Culture-derived (CD) B cells are readily cryopreserved and, when recovered, retain their ability to proliferate and differentiate. Significantly, proliferating CD B cells express high levels of MHC class II, CD80, and CD86. CD B cells act as APCs and present alloantigens and microbial Ags to T cells. We are able to activate and expand Ag-specific memory B cells; these cultured cells are highly effective in presenting Ag to T cells. We characterized the TCR repertoire of rare Ag-specific CD4+ T cells that proliferated in response to tetanus toxoid (TT) presented by autologous CD B cells. TCR Vβ usage by TT-activated CD4+ T cells differs from resting and unspecifically activated CD4+ T cells. Moreover, we found that TT-specific TCR Vβ usage by CD4+ T cells was substantially different between donors. This culture method provides a platform for studying the BCR and TCR repertoires within a single individual.
Collapse
Affiliation(s)
- Kuei-Ying Su
- Department of Immunology, Duke University, Durham, NC 27710.,Tzu Chi Medical Center, Hualien 970, Taiwan; and
| | - Akiko Watanabe
- Department of Immunology, Duke University, Durham, NC 27710
| | - Chen-Hao Yeh
- Department of Immunology, Duke University, Durham, NC 27710
| | - Garnett Kelsoe
- Department of Immunology, Duke University, Durham, NC 27710; .,Human Vaccine Institute, Duke University, Durham, NC 27710
| | | |
Collapse
|
181
|
Abstract
In the current era, one of the major factors limiting graft survival is chronic antibody-mediated rejection (ABMR), whilst patient survival is impacted by the effects of immunosuppression on susceptibility to infection, malignancy and atherosclerosis. IgG antibodies play a role in all of these processes, and many of their cellular effects are mediated by Fc gamma receptors (FcγRs). These surface receptors are expressed by most immune cells, including B cells, natural killer cells, dendritic cells and macrophages. Genetic variation in FCGR genes is likely to affect susceptibility to ABMR and to modulate the physiological functions of IgG. In this review, we discuss the potential role played by FcγRs in determining outcomes in solid organ transplantation, and how genetic polymorphisms in these receptors may contribute to variations in transplant outcome.
Collapse
Affiliation(s)
- Tomas Castro-Dopico
- Molecular Immunity Unit, Department of Medicine, MRC Laboratory of Molecular Biology, University of Cambridge, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, CB2 0QH UK
| | - Menna R. Clatworthy
- Molecular Immunity Unit, Department of Medicine, MRC Laboratory of Molecular Biology, University of Cambridge, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, CB2 0QH UK
| |
Collapse
|
182
|
Abstract
PURPOSE OF REVIEW The number of deaths associated with cardiovascular disease remains high, despite great advances in treating the associated high levels of cholesterol. The main underlying pathology of cardiovascular disease is atherosclerosis, which is recognized as a chronic autoimmune-like inflammatory disease. Hence, there is a pressing need to shed light on the immune pathways associated with atherosclerosis. B cells have long been thought to have a general protective effect in atherosclerosis. However, findings in the last decade have challenged this paradigm, showing that it is crucial to differentiate between the various B-cell subsets when assessing their role/effect on atherosclerosis. RECENT FINDINGS It has become increasingly recognized lately that B cells can have significant effects on the immune system independent of antibody production. The understanding that B cells form a major source of cytokines and can directly influence T-cell responses via surface markers, have led to the identification of novel B-cell subsets. These subsets are important modulators of autoimmune disorders but have not yet been fully investigated in atherosclerosis. SUMMARY Here we review the current known roles of B-cell subsets and the putative effects of recently identified B cells on atherosclerosis.
Collapse
Affiliation(s)
- Hidde Douna
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | | |
Collapse
|
183
|
Selvaraj UM, Poinsatte K, Torres V, Ortega SB, Stowe AM. Heterogeneity of B Cell Functions in Stroke-Related Risk, Prevention, Injury, and Repair. Neurotherapeutics 2016; 13:729-747. [PMID: 27492770 PMCID: PMC5081124 DOI: 10.1007/s13311-016-0460-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
It is well established that post-stroke inflammation contributes to neurovascular injury, blood-brain barrier disruption, and poor functional recovery in both animal and clinical studies. However, recent studies also suggest that several leukocyte subsets, activated during the post-stroke immune response, can exhibit both pro-injury and pro-recovery phenotypes. In accordance with these findings, B lymphocytes, or B cells, play a heterogeneous role in the adaptive immune response to stroke. This review highlights what is currently understood about the various roles of B cells, with an emphasis on stroke risk factors, as well as post-stroke injury and repair. This includes an overview of B cell functions, such as antibody production, cytokine secretion, and contribution to the immune response as antigen presenting cells. Next, evidence for B cell-mediated mechanisms in stroke-related risk factors, including hypertension, diabetes, and atherosclerosis, is outlined, followed by studies that focus on B cells during endogenous protection from stroke. Subsequently, animal studies that investigate the role of B cells in post-stroke injury and repair are summarized, and the final section describes current B cell-related clinical trials for stroke, as well as other central nervous system diseases. This review reveals the complex role of B cells in stroke, with a focus on areas for potential clinical intervention for a disease that affects millions of people globally each year.
Collapse
Affiliation(s)
- Uma Maheswari Selvaraj
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, MC8813, Dallas, TX, 75390, USA
| | - Katherine Poinsatte
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, MC8813, Dallas, TX, 75390, USA
| | - Vanessa Torres
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, MC8813, Dallas, TX, 75390, USA
| | - Sterling B Ortega
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, MC8813, Dallas, TX, 75390, USA
| | - Ann M Stowe
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, MC8813, Dallas, TX, 75390, USA.
| |
Collapse
|
184
|
McGaha TL, Karlsson MCI. Apoptotic cell responses in the splenic marginal zone: a paradigm for immunologic reactions to apoptotic antigens with implications for autoimmunity. Immunol Rev 2016; 269:26-43. [PMID: 26683143 DOI: 10.1111/imr.12382] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Apoptotic cells drive innate regulatory responses that result in tolerogenic immunity. This is a critical aspect of cell physiology as apoptotic cells expose potentially dangerous nuclear antigens on the surface in apoptotic blebs, and failure in their recognition, phagocytosis, or destruction can cause dramatic autoimmunity in experimental models and is linked to development and progression of systemic pathology in human. The marginal zone is a specialized splenic environment that serves as a transitional site from circulation to peripheral lymphoid structures. The marginal zone serves a key role in trapping of particulates and initiation of innate responses against systemic microbial pathogens. However in recent years, it has become clear the marginal zone is also important for initiation of immune tolerance to apoptotic cells, driving a coordinated response involving multiple phagocyte and lymphocyte subsets. Recent reports linking defects in splenic macrophage function to systemic lupus erythematosus in a manner analogous to marginal zone macrophages in lupus-prone mice provide an impetus to better understand the mechanistic basis of the apoptotic cell response in the marginal zone and its general applicability to apoptotic cell-driven tolerance at other tissue sites. In this review, we discuss immune responses to apoptotic cells in the spleen in general and the marginal zone in particular, the relationship of these responses to autoimmune disease, and comparisons to apoptotic cell immunity in humans.
Collapse
Affiliation(s)
- Tracy L McGaha
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
185
|
Mason JC. Cytoprotective pathways in the vascular endothelium. Do they represent a viable therapeutic target? Vascul Pharmacol 2016; 86:41-52. [PMID: 27520362 DOI: 10.1016/j.vph.2016.08.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/08/2016] [Indexed: 12/28/2022]
Abstract
The vascular endothelium is a critical interface, which separates the organs from the blood and its contents. The endothelium has a wide variety of functions and maintenance of endothelial homeostasis is a multi-dimensional active process, disruption of which has potentially deleterious consequences if not reversed. Vascular injury predisposes to endothelial apoptosis, dysfunction and development of atherosclerosis. Endothelial dysfunction is an end-point, a central feature of which is increased ROS generation, a reduction in endothelial nitric oxide synthase and increased nitric oxide consumption. A dysfunctional endothelium is a common feature of diseases including rheumatoid arthritis, systemic lupus erythematosus, diabetes mellitus and chronic renal impairment. The endothelium is endowed with a variety of constitutive and inducible mechanisms that act to minimise injury and facilitate repair. Endothelial cytoprotection can be enhanced by exogenous factors such as vascular endothelial growth factor, prostacyclin and laminar shear stress. Target genes include endothelial nitric oxide synthase, heme oxygenase-1, A20 and anti-apoptotic members of the B cell lymphoma protein-2 family. In light of the importance of endothelial function, and the link between its disruption and the risk of atherothrombosis, interest has focused on therapeutic conditioning and reversal of endothelial dysfunction. A detailed understanding of cytoprotective signalling pathways, their regulation and target genes is now required to identify novel therapeutic targets. The ultimate aim is to add vasculoprotection to current therapeutic strategies for systemic inflammatory diseases, in an attempt to reduce vascular injury and prevent or retard atherogenesis.
Collapse
Affiliation(s)
- Justin C Mason
- Vascular Science, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Hospital, London, UK.
| |
Collapse
|
186
|
Thayaparan D, Shen P, Stämpfli MR, Morissette MC. Induction of pulmonary antibodies against oxidized lipids in mice exposed to cigarette smoke. Respir Res 2016; 17:97. [PMID: 27488019 PMCID: PMC4973059 DOI: 10.1186/s12931-016-0416-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 07/27/2016] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Chronic cigarette smoke exposure is known to activate the adaptive immune system; however, the functional role of these processes is currently unknown. Given the role of oxidized lipids in driving innate inflammatory responses to cigarette smoke, we investigated whether an adaptive immune response against damaged lipids was induced following chronic cigarette smoke exposure. METHODS AND RESULTS Using a well-established mouse model, we showed that cigarette smoke exposure led to a progressive increase in pulmonary antibodies against oxidized low-density lipoprotein (OxLDL). Functionally, we found that intranasal delivery of an antibody against oxidized phosphatidylcholine (anti-OxPC; clone E06) increased lipid and particle uptake by pulmonary macrophages without exacerbating cigarette smoke-induced neutrophilia. We also found that anti-OxPC treatment increased particle uptake following smoking cessation. Finally, the frequency of pulmonary macrophages with internalized particles was increased after prolonged smoke exposure, at which time lung anti-OxPC responses were highest. CONCLUSIONS Altogether, this is the first report to demonstrate a non-pathogenic, and possibly protective, function of a newly identified autoantibody induced by chronic cigarette smoke exposure.
Collapse
Affiliation(s)
- Danya Thayaparan
- Medical Sciences Graduate Program, McMaster University, Hamilton, ON, Canada
| | - Pamela Shen
- Medical Sciences Graduate Program, McMaster University, Hamilton, ON, Canada
| | - Martin R Stämpfli
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Department of Medicine, Firestone Institute of Respiratory Health at St. Joseph's Healthcare, McMaster University, Hamilton, ON, Canada
| | - Mathieu C Morissette
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725, Chemin Ste-Foy, G1V 4G5, Quebec City, PQ, Canada.
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, PQ, Canada.
| |
Collapse
|
187
|
Tay C, Liu YH, Hosseini H, Kanellakis P, Cao A, Peter K, Tipping P, Bobik A, Toh BH, Kyaw T. B-cell-specific depletion of tumour necrosis factor alpha inhibits atherosclerosis development and plaque vulnerability to rupture by reducing cell death and inflammation. Cardiovasc Res 2016; 111:385-97. [PMID: 27492217 DOI: 10.1093/cvr/cvw186] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 07/08/2016] [Indexed: 12/31/2022] Open
Abstract
AIMS B2 lymphocytes promote atherosclerosis development but their mechanisms of action are unknown. Here, we investigated the role of tumour necrosis factor alpha (TNF-α) produced by B2 cells in atherogenesis. METHODS AND RESULTS We found that 50% of TNF-α-producing spleen lymphocytes were B2 cells and ∼20% of spleen and aortic B cells produced TNF-α in hyperlipidemic ApoE(-/-) mice. We generated mixed bone marrow (80% μMT/20% TNF-α(-/-)) chimeric LDLR(-/-) mice where only B cells did not express TNF-α. Atherosclerosis was reduced in chimeric LDLR(-/-) mice with TNF-α-deficient B cells. TNF-α expression in atherosclerotic lesions and in macrophages were also reduced accompanied by fewer apoptotic cells, reduced necrotic cores, and reduced lesion Fas, interleukin-1β and MCP-1 in mice with TNF-α-deficient B cells compared to mice with TNF-α-sufficient B cells. To confirm that the reduced atherosclerosis is attributable to B2 cells, we transferred wild-type and TNF-α-deficient B2 cells into ApoE(-/-) mice deficient in B cells or in lymphocytes. After 8 weeks of high fat diet, we found that atherosclerosis was increased by wild-type but not TNF-α-deficient B2 cells. Lesions of mice with wild-type B2 cells but not TNF-α-deficient B2 cells also had increased apoptotic cells and necrotic cores. Transferred B2 cells were found in lesions of recipient mice, suggesting that TNF-α-producing B2 cells promote atherosclerosis within lesions. CONCLUSION We conclude that TNF-α produced by B2 cells is a key mechanism by which B2 cells promote atherogenesis through augmenting macrophage TNF-α production to induce cell death and inflammation that promote plaque vulnerability.
Collapse
Affiliation(s)
- Christopher Tay
- Vascular Biology and Atherosclerosis Laboratory, Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, Victoria 3004, Australia Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Faculty of Medicine, Nursing and Health Sciences. Monash University, 246 Clayton Road, Clayton, Victoria 3168, Australia
| | - Yu-Han Liu
- Vascular Biology and Atherosclerosis Laboratory, Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, Victoria 3004, Australia
| | - Hamid Hosseini
- Vascular Biology and Atherosclerosis Laboratory, Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, Victoria 3004, Australia Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Faculty of Medicine, Nursing and Health Sciences. Monash University, 246 Clayton Road, Clayton, Victoria 3168, Australia
| | - Peter Kanellakis
- Vascular Biology and Atherosclerosis Laboratory, Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, Victoria 3004, Australia
| | - Anh Cao
- Vascular Biology and Atherosclerosis Laboratory, Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, Victoria 3004, Australia Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Faculty of Medicine, Nursing and Health Sciences. Monash University, 246 Clayton Road, Clayton, Victoria 3168, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Laboratory, Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, Victoria 3004, Australia
| | - Peter Tipping
- Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Faculty of Medicine, Nursing and Health Sciences. Monash University, 246 Clayton Road, Clayton, Victoria 3168, Australia
| | - Alex Bobik
- Vascular Biology and Atherosclerosis Laboratory, Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, Victoria 3004, Australia
| | - Ban-Hock Toh
- Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Faculty of Medicine, Nursing and Health Sciences. Monash University, 246 Clayton Road, Clayton, Victoria 3168, Australia
| | - Tin Kyaw
- Vascular Biology and Atherosclerosis Laboratory, Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, Victoria 3004, Australia Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Faculty of Medicine, Nursing and Health Sciences. Monash University, 246 Clayton Road, Clayton, Victoria 3168, Australia
| |
Collapse
|
188
|
Adaptive immunity against gut microbiota enhances apoE-mediated immune regulation and reduces atherosclerosis and western-diet-related inflammation. Sci Rep 2016; 6:29353. [PMID: 27383250 PMCID: PMC4935993 DOI: 10.1038/srep29353] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 06/16/2016] [Indexed: 01/11/2023] Open
Abstract
Common features of immune-metabolic and inflammatory diseases such as metabolic syndrome, diabetes, obesity and cardiovascular diseases are an altered gut microbiota composition and a systemic pro-inflammatory state. We demonstrate that active immunization against the outer membrane protein of bacteria present in the gut enhances local and systemic immune control via apoE-mediated immune-modulation. Reduction of western-diet-associated inflammation was obtained for more than eighteen weeks after immunization. Immunized mice had reduced serum cytokine levels, reduced insulin and fasting glucose concentrations; and gene expression in both liver and visceral adipose tissue confirmed a reduced inflammatory steady-state after immunization. Moreover, both gut and atherosclerotic plaques of immunized mice showed reduced inflammatory cells and an increased M2 macrophage fraction. These results suggest that adaptive responses directed against microbes present in our microbiota have systemic beneficial consequences and demonstrate the key role of apoE in this mechanism that could be exploited to treat immune-metabolic diseases.
Collapse
|
189
|
Abstract
Atherosclerosis is a chronic inflammatory disease that is initiated by the retention and accumulation of cholesterol-containing lipoproteins, particularly low-density lipoprotein, in the artery wall. In the arterial intima, lipoprotein components that are generated through oxidative, lipolytic, and proteolytic activities lead to the formation of several danger-associated molecular patterns, which can activate innate immune cells as well as vascular cells. Moreover, self- and non-self-antigens, such as apolipoprotein B-100 and heat shock proteins, can contribute to vascular inflammation by triggering the response of T and B cells locally. This process can influence the initiation, progression, and stability of plaques. Substantial clinical and experimental data support that the modulation of adaptive immune system may be used for treating and preventing atherosclerosis. This may lead to the development of more selective and less harmful interventions, while keeping host defense mechanisms against infections and tumors intact. Approaches such as vaccination might become a realistic option for cardiovascular disease, especially if they can elicit regulatory T and B cells and the secretion of atheroprotective antibodies. Nevertheless, difficulties in translating certain experimental data into new clinical therapies remain a challenge. In this review, we discuss important studies on the function of T- and B-cell immunity in atherosclerosis and their manipulation to develop novel therapeutic strategies against cardiovascular disease.
Collapse
Affiliation(s)
- Daniel F J Ketelhuth
- From the Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | - Göran K Hansson
- From the Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
190
|
Khamis RY, Hughes AD, Caga-Anan M, Chang CL, Boyle JJ, Kojima C, Welsh P, Sattar N, Johns M, Sever P, Mayet J, Haskard DO. High Serum Immunoglobulin G and M Levels Predict Freedom From Adverse Cardiovascular Events in Hypertension: A Nested Case-Control Substudy of the Anglo-Scandinavian Cardiac Outcomes Trial. EBioMedicine 2016; 9:372-380. [PMID: 27333022 PMCID: PMC4972545 DOI: 10.1016/j.ebiom.2016.06.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 05/20/2016] [Accepted: 06/06/2016] [Indexed: 10/25/2022] Open
Abstract
AIMS We aimed to determine whether the levels of total serum IgM and IgG, together with specific antibodies against malondialdehyde-conjugated low-density lipoprotein (MDA-LDL), can improve cardiovascular risk discrimination. METHODS AND RESULTS The Anglo-Scandinavian Cardiac Outcomes Trial (ASCOT) randomized 9098 patients in the UK and Ireland into the Blood Pressure-Lowering Arm. 485 patients that had cardiovascular (CV) events over 5.5years were age and sex matched with 1367 controls. Higher baseline total serum IgG, and to a lesser extent IgM, were associated with decreased risk of CV events (IgG odds ratio (OR) per one standard deviation (SD) 0.80 [95% confidence interval, CI 0.72,0.89], p<0.0001; IgM 0.83[0.75,0.93], p=0.001), and particularly events due to coronary heart disease (CHD) (IgG OR 0.66 (0.57,0.76); p<0.0001, IgM OR 0.81 (0.71,0.93); p=0.002). The association persisted after adjustment for a basic model with variables in the Framingham Risk Score (FRS) as well as following inclusion of C-reactive protein (CRP) and N-terminal pro-B-type natriuretic peptide (NtProBNP). IgG and IgM antibodies against MDA-LDL were also associated with CV events but their significance was lost following adjustment for total serum IgG and IgM respectively. The area under the receiver operator curve for CV events was improved from the basic risk model when adding in total serum IgG, and there was improvement in continuous and categorical net reclassification (17.6% and 7.5% respectively) as well as in the integrated discrimination index. CONCLUSION High total serum IgG levels are an independent predictor of freedom from adverse cardiovascular events, particularly those attributed to CHD, in patients with hypertension.
Collapse
Affiliation(s)
- Ramzi Y Khamis
- Vascular Sciences Section, NHLI, Imperial College London, United Kingdom
| | - Alun D Hughes
- International Centre for Circulatory Health, NHLI, Imperial College London, United Kingdom; Institute of Cardiovascular Science, University College London, United Kingdom
| | - Mikhail Caga-Anan
- Vascular Sciences Section, NHLI, Imperial College London, United Kingdom
| | - Choon L Chang
- International Centre for Circulatory Health, NHLI, Imperial College London, United Kingdom
| | - Joseph J Boyle
- Vascular Sciences Section, NHLI, Imperial College London, United Kingdom
| | - Chiari Kojima
- Vascular Sciences Section, NHLI, Imperial College London, United Kingdom
| | - Paul Welsh
- Division of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| | - Naveed Sattar
- Division of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| | - Michael Johns
- Vascular Sciences Section, NHLI, Imperial College London, United Kingdom
| | - Peter Sever
- International Centre for Circulatory Health, NHLI, Imperial College London, United Kingdom
| | - Jamil Mayet
- International Centre for Circulatory Health, NHLI, Imperial College London, United Kingdom
| | - Dorian O Haskard
- Vascular Sciences Section, NHLI, Imperial College London, United Kingdom.
| |
Collapse
|
191
|
Nus M, Mallat Z. Immune-mediated mechanisms of atherosclerosis and implications for the clinic. Expert Rev Clin Immunol 2016; 12:1217-1237. [PMID: 27253721 DOI: 10.1080/1744666x.2016.1195686] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION A large body of evidence supports the inflammatory hypothesis of atherosclerosis, and both innate and adaptive immune responses play important roles in all disease stages. Areas covered: Here, we review our understanding of the role of the immune response in atherosclerosis, focusing on the pathways currently amenable to therapeutic modulation. We also discuss the advantages or undesirable effects that may be foreseen from targeting the immune response in patients at high cardiovascular risk, suggesting new avenues for research. Expert commentary: There is an extraordinary opportunity to directly test the inflammatory hypothesis of atherosclerosis in the clinic using currently available therapeutics. However, a more balanced interpretation of the experimental and translational data is needed, which may help address and identify in more detail the appropriate settings where an immune pathway can be targeted with minimal risk.
Collapse
Affiliation(s)
- Meritxell Nus
- a Division of Cardiovascular Medicine, Department of Medicine , University of Cambridge , Cambridge , UK
| | - Ziad Mallat
- a Division of Cardiovascular Medicine, Department of Medicine , University of Cambridge , Cambridge , UK
| |
Collapse
|
192
|
Abstract
PURPOSE OF REVIEW Cardiovascular disease is the leading cause of mortality worldwide. The underlying cause of the majority of cardiovascular disease is atherosclerosis. In the past, atherosclerosis was considered to be the result of passive lipid accumulation in the vessel wall. However, today's picture of the pathogenesis of atherosclerosis is much more complex, with a key role for immune cells and inflammation in conjunction with hyperlipidemia, especially elevated (modified) LDL levels. Knowledge on immune cells and immune responses in atherosclerosis has progressed tremendously over the past decades, and the same is true for the role of lipid metabolism and the different lipid components. However, it is largely unknown how lipids and the immune system interact. In this review, we will describe the effect of lipids on immune cell development and function, and the effects of immune cells on lipid metabolism. RECENT FINDINGS Recently, novel data have emerged that show that immune cells are affected, and behave differently in a hyperlipidemic environment. Moreover, immune cells have reported to be able to affect lipid metabolism. SUMMARY In this review, we will summarize the latest findings on the interactions between lipids and the immune system, and we will discuss the potential consequences of these novel insights for future therapies for atherosclerosis.
Collapse
Affiliation(s)
- Frank Schaftenaar
- aDivision of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden bDepartment of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands cInstitute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Munich, Germany
| | | | | | | |
Collapse
|
193
|
Foks AC, Van Puijvelde GHM, Wolbert J, Kröner MJ, Frodermann V, Van Der Heijden T, Van Santbrink PJ, Boon L, Bot I, Kuiper J. CD11b+Gr-1+ myeloid-derived suppressor cells reduce atherosclerotic lesion development in LDLr deficient mice. Cardiovasc Res 2016; 111:252-61. [PMID: 27234908 DOI: 10.1093/cvr/cvw114] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 05/16/2016] [Indexed: 01/25/2023] Open
Abstract
AIMS Myeloid-derived suppressor cells (MDSCs) form a heterogeneous population of cells composed of early myeloid progenitor cells and immature myeloid cells, which strongly suppress pro-inflammatory immune cells in inflammatory diseases. Currently, it is unknown whether MDSCs contribute to atherosclerosis, a chronic inflammatory disease in which accumulation of lipoproteins in the arterial wall activates the immune system causing abnormal vascular remodelling and vessel occlusion. Here, we investigated whether and how MDSCs contribute to the development of atherosclerosis. METHODS AND RESULTS We show that MDSCs arise in the bone marrow of LDLr(-/-) mice during atherosclerosis and strongly suppress proliferation of T cells. Adoptive transfer of MDSCs into both female and male LDLr(-/-) mice fed a Western-type diet (WTD) ameliorates atherosclerosis with 35%. We observed a 54% reduction in adventitial T cells, and more specifically, MDSCs suppress Th1 and Th17 cells. In addition, treatment with MDSCs reduces circulating pro-atherogenic B2 cells. We found two subsets of MDSCs in the bone marrow of hypercholesterolemic mice, monocytic and granulocytic MDSCs (mo- and gr-MDSCs, respectively), of which the percentage of mo-MDSCs significantly increased during WTD feeding. Moreover, mo-MDSCs completely abolished splenocyte proliferation, whereas gr-MDSCs were unable to suppress proliferation. Mechanistically, we show that MDSCs from atherosclerotic mice suppress T cells in an IFN-γ- and nitric oxide-dependent manner, which is associated with the action of mo-MDSCs. CONCLUSION This study demonstrates that MDSCs develop during atherosclerosis and reduce atherosclerosis via suppression of pro-inflammatory immune responses.
Collapse
Affiliation(s)
- Amanda C Foks
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Gijs H M Van Puijvelde
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Jolien Wolbert
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Mara J Kröner
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Vanessa Frodermann
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Thomas Van Der Heijden
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Peter J Van Santbrink
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | | | - Ilze Bot
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Johan Kuiper
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| |
Collapse
|
194
|
Immune-inflammatory responses in atherosclerosis: Role of an adaptive immunity mainly driven by T and B cells. Immunobiology 2016; 221:1014-33. [PMID: 27262513 DOI: 10.1016/j.imbio.2016.05.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 05/06/2016] [Accepted: 05/23/2016] [Indexed: 01/22/2023]
Abstract
Adaptive immune response plays an important role in atherogenesis. In atherosclerosis, the proinflammatory immune response driven by Th1 is predominant but the anti-inflammatory response mediated mainly by regulatory T cells is also present. The role of Th2 and Th17 cells in atherogenesis is still debated. In the plaque, other T helper cells can be observed such as Th9 and Th22 but is little is known about their impact in atherosclerosis. Heterogeneity of CD4(+) T cell subsets presented in the plaque may suggest for plasticity of T cell that can switch the phenotype dependening on the local microenvironment and activating/blocking stimuli. Effector T cells are able to recognize self-antigens released by necrotic and apoptotic vascular cells and induce a humoral immune reaction. Tth cells resided in the germinal centers help B cells to switch the antibody class to the production of high-affinity antibodies. Humoral immunity is mediated by B cells that release antigen-specific antibodies. A variety of B cell subsets were found in human and murine atherosclerotic plaques. In mice, B1 cells could spontaneously produce atheroprotective natural IgM antibodies. Conventional B2 lymphocytes secrete either proatherogenic IgG, IgA, and IgE or atheroprotective IgG and IgM antibodies reactive with oxidation-specific epitopes on atherosclerosis-associated antigens. A small population of innate response activator (IRA) B cells, which is phenotypically intermediate between B1 and B2 cells, produces IgM but possesses proatherosclerotic properties. Finally, there is a minor subset of splenic regulatory B cells (Bregs) that protect against atherosclerotic inflammation through support of generation of Tregs and production of anti-inflammatory cytokines IL-10 and TGF-β and proapoptotic molecules.
Collapse
|
195
|
Barsalou J, Bradley TJ, Tyrrell PN, Slorach C, Ng LWK, Levy DM, Silverman ED. Impact of Disease Duration on Vascular Surrogates of Early Atherosclerosis in Childhood-Onset Systemic Lupus Erythematosus. Arthritis Rheumatol 2016; 68:237-46. [PMID: 26361097 DOI: 10.1002/art.39423] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 09/01/2015] [Indexed: 12/27/2022]
Abstract
OBJECTIVE To determine whether longer disease duration negatively impacts carotid intima-media thickness (CIMT), flow-mediated dilation (FMD), and pulse wave velocity (PWV) in a cohort of patients with childhood-onset systemic lupus erythematosus (SLE), and to compare CIMT, FMD, and PWV in patients with childhood-onset SLE with those in healthy children and explore determinants of vascular test results in childhood-onset SLE. METHODS Cross-sectional analysis was performed in a prospective longitudinal cohort of patients with childhood-onset SLE at the latest followup visit. Clinical and laboratory data were collected for patients with childhood-onset SLE. CIMT, FMD, and PWV were measured using standardized protocols in patients with childhood-onset SLE and healthy children. Correlations between disease duration and results of the 3 vascular tests were performed. Vascular data in patients with childhood-onset SLE were compared with those in healthy children. Multivariable linear regression was used to identify determinants of CIMT, FMD, and PWV in childhood-onset SLE. RESULTS Patients with childhood-onset SLE (n = 149) and healthy controls (n = 178) were enrolled. The median age of the patients was 17.2 years (interquartile range [IQR] 15.7-17.9 years), and their median disease duration was 3.2 years (IQR 1.8-4.9 years). The median age of the healthy children was 14.7 years (IQR 13.1-15.9 years). Longer disease duration correlated with worse FMD (r = -0.2, P = 0.031) in patients with childhood-onset SLE. Patients with childhood-onset SLE had smaller (better) CIMT, higher (better) FMD, and similar PWV compared with healthy controls. Linear regression analysis explained <24% of the variation in vascular test results in patients with childhood-onset SLE, suggesting that other variables should be explored as important determinants of CIMT, FMD, and PWV. CONCLUSION In this cohort of 149 patients with childhood-onset SLE, patients did not have worse CIMT, FMD, or PWV than did healthy controls. Longer disease duration was associated with worse FMD, suggesting progressive endothelial dysfunction over time.
Collapse
Affiliation(s)
- Julie Barsalou
- Centre Hospitalier Universitaire St. Justine, University of Montreal, Montreal, Quebec, Canada), Timothy J. Bradley, MBChB, FRACP, Cameron Slorach, RDCS, Lawrence W. K. Ng, BSc, Deborah M. Levy, MD, MS, FRCPC: The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Timothy J Bradley
- Centre Hospitalier Universitaire St. Justine, University of Montreal, Montreal, Quebec, Canada), Timothy J. Bradley, MBChB, FRACP, Cameron Slorach, RDCS, Lawrence W. K. Ng, BSc, Deborah M. Levy, MD, MS, FRCPC: The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Cameron Slorach
- Centre Hospitalier Universitaire St. Justine, University of Montreal, Montreal, Quebec, Canada), Timothy J. Bradley, MBChB, FRACP, Cameron Slorach, RDCS, Lawrence W. K. Ng, BSc, Deborah M. Levy, MD, MS, FRCPC: The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lawrence W K Ng
- Centre Hospitalier Universitaire St. Justine, University of Montreal, Montreal, Quebec, Canada), Timothy J. Bradley, MBChB, FRACP, Cameron Slorach, RDCS, Lawrence W. K. Ng, BSc, Deborah M. Levy, MD, MS, FRCPC: The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Deborah M Levy
- Centre Hospitalier Universitaire St. Justine, University of Montreal, Montreal, Quebec, Canada), Timothy J. Bradley, MBChB, FRACP, Cameron Slorach, RDCS, Lawrence W. K. Ng, BSc, Deborah M. Levy, MD, MS, FRCPC: The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Earl D Silverman
- The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
196
|
Srikakulapu P, Hu D, Yin C, Mohanta SK, Bontha SV, Peng L, Beer M, Weber C, McNamara CA, Grassia G, Maffia P, Manz RA, Habenicht AJR. Artery Tertiary Lymphoid Organs Control Multilayered Territorialized Atherosclerosis B-Cell Responses in Aged ApoE-/- Mice. Arterioscler Thromb Vasc Biol 2016; 36:1174-85. [PMID: 27102965 PMCID: PMC4894775 DOI: 10.1161/atvbaha.115.306983] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 04/11/2016] [Indexed: 12/20/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Explore aorta B-cell immunity in aged apolipoprotein E-deficient (ApoE−/−) mice. Approach and Results— Transcript maps, fluorescence-activated cell sorting, immunofluorescence analyses, cell transfers, and Ig-ELISPOT (enzyme-linked immunospot) assays showed multilayered atherosclerosis B-cell responses in artery tertiary lymphoid organs (ATLOs). Aging-associated aorta B-cell–related transcriptomes were identified, and transcript atlases revealed highly territorialized B-cell responses in ATLOs versus atherosclerotic lesions: ATLOs showed upregulation of bona fide B-cell genes, including Cd19, Ms4a1 (Cd20), Cd79a/b, and Ighm although intima plaques preferentially expressed molecules involved in non–B effector responses toward B-cell–derived mediators, that is, Fcgr3 (Cd16), Fcer1g (Cd23), and the C1q family. ATLOs promoted B-cell recruitment. ATLO B-2 B cells included naive, transitional, follicular, germinal center, switched IgG1+, IgA+, and IgE+ memory cells, plasmablasts, and long-lived plasma cells. ATLOs recruited large numbers of B-1 cells whose subtypes were skewed toward interleukin-10+ B-1b cells versus interleukin-10− B-1a cells. ATLO B-1 cells and plasma cells constitutively produced IgM and IgG and a fraction of plasma cells expressed interleukin-10. Moreover, ApoE−/− mice showed increased germinal center B cells in renal lymph nodes, IgM-producing plasma cells in the bone marrow, and higher IgM and anti–MDA-LDL (malondialdehyde-modified low-density lipoprotein) IgG serum titers. Conclusions— ATLOs orchestrate dichotomic, territorialized, and multilayered B-cell responses in the diseased aorta; germinal center reactions indicate generation of autoimmune B cells within the diseased arterial wall during aging.
Collapse
Affiliation(s)
- Prasad Srikakulapu
- From the Cardiovascular Research Center, Department of Medicine (P.S., C.A.M.), Department of Surgery (S.V.B.), University of Virginia, Charlottesville; Institute for Immunology (D.H.) and Institute for Cardiovascular Prevention (C.Y., S.K.M., C.W., A.J.R.H.), Ludwig-Maximilians-University, Munich, Germany; Institute of Molecular Immunology, Helmholtz-Zentrum München, Oberschleißheim, Germany (D.H.); Department for Information Technology, University of Jena, Jena University Hospital, Jena, Germany (M.B.); Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (G.G., P.M.); BHF Centre for Excellence in Vascular Science and Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M.); Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M.); Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany (R.A.M.); and Department of Traditional Chinese Medicine, Medical College of Xiamen University, Xiamen University, Xiamen, China (L.P.)
| | - Desheng Hu
- From the Cardiovascular Research Center, Department of Medicine (P.S., C.A.M.), Department of Surgery (S.V.B.), University of Virginia, Charlottesville; Institute for Immunology (D.H.) and Institute for Cardiovascular Prevention (C.Y., S.K.M., C.W., A.J.R.H.), Ludwig-Maximilians-University, Munich, Germany; Institute of Molecular Immunology, Helmholtz-Zentrum München, Oberschleißheim, Germany (D.H.); Department for Information Technology, University of Jena, Jena University Hospital, Jena, Germany (M.B.); Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (G.G., P.M.); BHF Centre for Excellence in Vascular Science and Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M.); Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M.); Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany (R.A.M.); and Department of Traditional Chinese Medicine, Medical College of Xiamen University, Xiamen University, Xiamen, China (L.P.)
| | - Changjun Yin
- From the Cardiovascular Research Center, Department of Medicine (P.S., C.A.M.), Department of Surgery (S.V.B.), University of Virginia, Charlottesville; Institute for Immunology (D.H.) and Institute for Cardiovascular Prevention (C.Y., S.K.M., C.W., A.J.R.H.), Ludwig-Maximilians-University, Munich, Germany; Institute of Molecular Immunology, Helmholtz-Zentrum München, Oberschleißheim, Germany (D.H.); Department for Information Technology, University of Jena, Jena University Hospital, Jena, Germany (M.B.); Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (G.G., P.M.); BHF Centre for Excellence in Vascular Science and Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M.); Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M.); Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany (R.A.M.); and Department of Traditional Chinese Medicine, Medical College of Xiamen University, Xiamen University, Xiamen, China (L.P.)
| | - Sarajo K Mohanta
- From the Cardiovascular Research Center, Department of Medicine (P.S., C.A.M.), Department of Surgery (S.V.B.), University of Virginia, Charlottesville; Institute for Immunology (D.H.) and Institute for Cardiovascular Prevention (C.Y., S.K.M., C.W., A.J.R.H.), Ludwig-Maximilians-University, Munich, Germany; Institute of Molecular Immunology, Helmholtz-Zentrum München, Oberschleißheim, Germany (D.H.); Department for Information Technology, University of Jena, Jena University Hospital, Jena, Germany (M.B.); Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (G.G., P.M.); BHF Centre for Excellence in Vascular Science and Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M.); Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M.); Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany (R.A.M.); and Department of Traditional Chinese Medicine, Medical College of Xiamen University, Xiamen University, Xiamen, China (L.P.)
| | - Sai Vineela Bontha
- From the Cardiovascular Research Center, Department of Medicine (P.S., C.A.M.), Department of Surgery (S.V.B.), University of Virginia, Charlottesville; Institute for Immunology (D.H.) and Institute for Cardiovascular Prevention (C.Y., S.K.M., C.W., A.J.R.H.), Ludwig-Maximilians-University, Munich, Germany; Institute of Molecular Immunology, Helmholtz-Zentrum München, Oberschleißheim, Germany (D.H.); Department for Information Technology, University of Jena, Jena University Hospital, Jena, Germany (M.B.); Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (G.G., P.M.); BHF Centre for Excellence in Vascular Science and Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M.); Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M.); Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany (R.A.M.); and Department of Traditional Chinese Medicine, Medical College of Xiamen University, Xiamen University, Xiamen, China (L.P.)
| | - Li Peng
- From the Cardiovascular Research Center, Department of Medicine (P.S., C.A.M.), Department of Surgery (S.V.B.), University of Virginia, Charlottesville; Institute for Immunology (D.H.) and Institute for Cardiovascular Prevention (C.Y., S.K.M., C.W., A.J.R.H.), Ludwig-Maximilians-University, Munich, Germany; Institute of Molecular Immunology, Helmholtz-Zentrum München, Oberschleißheim, Germany (D.H.); Department for Information Technology, University of Jena, Jena University Hospital, Jena, Germany (M.B.); Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (G.G., P.M.); BHF Centre for Excellence in Vascular Science and Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M.); Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M.); Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany (R.A.M.); and Department of Traditional Chinese Medicine, Medical College of Xiamen University, Xiamen University, Xiamen, China (L.P.)
| | - Michael Beer
- From the Cardiovascular Research Center, Department of Medicine (P.S., C.A.M.), Department of Surgery (S.V.B.), University of Virginia, Charlottesville; Institute for Immunology (D.H.) and Institute for Cardiovascular Prevention (C.Y., S.K.M., C.W., A.J.R.H.), Ludwig-Maximilians-University, Munich, Germany; Institute of Molecular Immunology, Helmholtz-Zentrum München, Oberschleißheim, Germany (D.H.); Department for Information Technology, University of Jena, Jena University Hospital, Jena, Germany (M.B.); Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (G.G., P.M.); BHF Centre for Excellence in Vascular Science and Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M.); Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M.); Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany (R.A.M.); and Department of Traditional Chinese Medicine, Medical College of Xiamen University, Xiamen University, Xiamen, China (L.P.)
| | - Christian Weber
- From the Cardiovascular Research Center, Department of Medicine (P.S., C.A.M.), Department of Surgery (S.V.B.), University of Virginia, Charlottesville; Institute for Immunology (D.H.) and Institute for Cardiovascular Prevention (C.Y., S.K.M., C.W., A.J.R.H.), Ludwig-Maximilians-University, Munich, Germany; Institute of Molecular Immunology, Helmholtz-Zentrum München, Oberschleißheim, Germany (D.H.); Department for Information Technology, University of Jena, Jena University Hospital, Jena, Germany (M.B.); Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (G.G., P.M.); BHF Centre for Excellence in Vascular Science and Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M.); Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M.); Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany (R.A.M.); and Department of Traditional Chinese Medicine, Medical College of Xiamen University, Xiamen University, Xiamen, China (L.P.)
| | - Coleen A McNamara
- From the Cardiovascular Research Center, Department of Medicine (P.S., C.A.M.), Department of Surgery (S.V.B.), University of Virginia, Charlottesville; Institute for Immunology (D.H.) and Institute for Cardiovascular Prevention (C.Y., S.K.M., C.W., A.J.R.H.), Ludwig-Maximilians-University, Munich, Germany; Institute of Molecular Immunology, Helmholtz-Zentrum München, Oberschleißheim, Germany (D.H.); Department for Information Technology, University of Jena, Jena University Hospital, Jena, Germany (M.B.); Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (G.G., P.M.); BHF Centre for Excellence in Vascular Science and Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M.); Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M.); Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany (R.A.M.); and Department of Traditional Chinese Medicine, Medical College of Xiamen University, Xiamen University, Xiamen, China (L.P.)
| | - Gianluca Grassia
- From the Cardiovascular Research Center, Department of Medicine (P.S., C.A.M.), Department of Surgery (S.V.B.), University of Virginia, Charlottesville; Institute for Immunology (D.H.) and Institute for Cardiovascular Prevention (C.Y., S.K.M., C.W., A.J.R.H.), Ludwig-Maximilians-University, Munich, Germany; Institute of Molecular Immunology, Helmholtz-Zentrum München, Oberschleißheim, Germany (D.H.); Department for Information Technology, University of Jena, Jena University Hospital, Jena, Germany (M.B.); Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (G.G., P.M.); BHF Centre for Excellence in Vascular Science and Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M.); Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M.); Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany (R.A.M.); and Department of Traditional Chinese Medicine, Medical College of Xiamen University, Xiamen University, Xiamen, China (L.P.)
| | - Pasquale Maffia
- From the Cardiovascular Research Center, Department of Medicine (P.S., C.A.M.), Department of Surgery (S.V.B.), University of Virginia, Charlottesville; Institute for Immunology (D.H.) and Institute for Cardiovascular Prevention (C.Y., S.K.M., C.W., A.J.R.H.), Ludwig-Maximilians-University, Munich, Germany; Institute of Molecular Immunology, Helmholtz-Zentrum München, Oberschleißheim, Germany (D.H.); Department for Information Technology, University of Jena, Jena University Hospital, Jena, Germany (M.B.); Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (G.G., P.M.); BHF Centre for Excellence in Vascular Science and Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M.); Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M.); Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany (R.A.M.); and Department of Traditional Chinese Medicine, Medical College of Xiamen University, Xiamen University, Xiamen, China (L.P.)
| | - Rudolf A Manz
- From the Cardiovascular Research Center, Department of Medicine (P.S., C.A.M.), Department of Surgery (S.V.B.), University of Virginia, Charlottesville; Institute for Immunology (D.H.) and Institute for Cardiovascular Prevention (C.Y., S.K.M., C.W., A.J.R.H.), Ludwig-Maximilians-University, Munich, Germany; Institute of Molecular Immunology, Helmholtz-Zentrum München, Oberschleißheim, Germany (D.H.); Department for Information Technology, University of Jena, Jena University Hospital, Jena, Germany (M.B.); Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (G.G., P.M.); BHF Centre for Excellence in Vascular Science and Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M.); Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M.); Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany (R.A.M.); and Department of Traditional Chinese Medicine, Medical College of Xiamen University, Xiamen University, Xiamen, China (L.P.)
| | - Andreas J R Habenicht
- From the Cardiovascular Research Center, Department of Medicine (P.S., C.A.M.), Department of Surgery (S.V.B.), University of Virginia, Charlottesville; Institute for Immunology (D.H.) and Institute for Cardiovascular Prevention (C.Y., S.K.M., C.W., A.J.R.H.), Ludwig-Maximilians-University, Munich, Germany; Institute of Molecular Immunology, Helmholtz-Zentrum München, Oberschleißheim, Germany (D.H.); Department for Information Technology, University of Jena, Jena University Hospital, Jena, Germany (M.B.); Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (G.G., P.M.); BHF Centre for Excellence in Vascular Science and Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M.); Department of Pharmacy, University of Naples Federico II, Naples, Italy (P.M.); Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany (R.A.M.); and Department of Traditional Chinese Medicine, Medical College of Xiamen University, Xiamen University, Xiamen, China (L.P.).
| |
Collapse
|
197
|
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. [PMID: 26848161 PMCID: PMC4808396 DOI: 10.1161/atvbaha.115.306931,10.1161/atvbaha.115.306931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
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
|
198
|
Rheumatoid Arthritis Pharmacotherapies: Do They Have Anti-Atherosclerotic Activity? Curr Rheumatol Rep 2016; 18:27. [DOI: 10.1007/s11926-016-0578-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
199
|
Yan W, Song H, Jiang J, Xu W, Gong Z, Duan Q, Li C, Xie Y, Wang L. Characteristics of B cell‑associated gene expression in patients with coronary artery disease. Mol Med Rep 2016; 13:4113-21. [PMID: 27035867 DOI: 10.3892/mmr.2016.5029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 02/12/2016] [Indexed: 11/05/2022] Open
Abstract
The current study aimed to identify differentially expressed B cell‑associated genes in peripheral blood mononuclear cells and observe the changes in B cell activation at different stages of coronary artery disease. Groups of patients with acute myocardial infarction (AMI) and stable angina (SA), as well as healthy volunteers, were recruited into the study (n=20 per group). Whole human genome microarray analysis was performed to examine the expression of B cell‑associated genes among these three groups. The mRNA expression levels of 60 genes associated with B cell activity and regulation were measured using reverse transcription‑quantitative polymerase chain reaction. The mRNA expression of the B cell antigen receptor (BCR)‑associated genes, CD45, NFAM, SYK and LYN, were significantly upregulated in patients with AMI; however, FCRL3, CD79B, CD19, CD81, FYN, BLK, CD22 and CD5 mRNA expression levels were significantly downregulated, compared with patients in the SA and control group. The mRNA levels of the T‑independent B cell‑associated genes, CD16, CD32, LILRA1 and TLR9, were significantly increased in AMI patients compared with SA and control patients. The mRNA expression of genes associated with T‑dependent B cells were also measured: EMR2 and CD97 were statistically upregulated, whereas SLAMF1, LY9, CD28, CD43, CD72, ICOSL, PD1, CD40 and CD20 mRNAs were significantly downregulated in AMI group patients compared with the two other groups. Additionally the gene expression levels of B cell regulatory genes were measured. In patients with AMI, CR1, LILRB2, LILRB3 and VAV1 mRNA expression levels were statistically increased, whereas, CS1 and IL4I1 mRNAs were significantly reduced compared with the SA and control groups. There was no statistically significant difference in B cell‑associated gene expression levels between patients with SA and the control group. The present study identified the downregulation of genes associated with BCRs, B2 cells and B cell regulators in patients with AMI, indicating a weakened T cell‑B cell interaction and reduced B2 cell activation during AMI. Thus, improving B2 cell‑mediated humoral immunity may be a potential target for medical intervention in patients with AMI.
Collapse
Affiliation(s)
- Wenwen Yan
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Haoming Song
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Jinfa Jiang
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Wenjun Xu
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Zhu Gong
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Qianglin Duan
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Chuangrong Li
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Yuan Xie
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Lemin Wang
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| |
Collapse
|
200
|
Gruber S, Hendrikx T, Tsiantoulas D, Ozsvar-Kozma M, Göderle L, Mallat Z, Witztum JL, Shiri-Sverdlov R, Nitschke L, Binder CJ. Sialic Acid-Binding Immunoglobulin-like Lectin G Promotes Atherosclerosis and Liver Inflammation by Suppressing the Protective Functions of B-1 Cells. Cell Rep 2016; 14:2348-61. [PMID: 26947073 PMCID: PMC4802221 DOI: 10.1016/j.celrep.2016.02.027] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 12/21/2015] [Accepted: 02/01/2016] [Indexed: 12/21/2022] Open
Abstract
Atherosclerosis is initiated and sustained by hypercholesterolemia, which results in the generation of oxidized LDL (OxLDL) and other metabolic byproducts that trigger inflammation. Specific immune responses have been shown to modulate the inflammatory response during atherogenesis. The sialic acid-binding immunoglobulin-like lectin G (Siglec-G) is a negative regulator of the functions of several immune cells, including myeloid cells and B-1 cells. Here, we show that deficiency of Siglec-G in atherosclerosis-prone mice inhibits plaque formation and diet-induced hepatic inflammation. We further demonstrate that selective deficiency of Siglec-G in B cells alone is sufficient to mediate these effects. Levels of B-1 cell-derived natural IgM with specificity for OxLDL were significantly increased in the plasma and peritoneal cavity of Siglec-G-deficient mice. Consistent with the neutralizing functions of OxLDL-specific IgM, Siglec-G-deficient mice were protected from OxLDL-induced sterile inflammation. Thus, Siglec-G promotes atherosclerosis and hepatic inflammation by suppressing protective anti-inflammatory effector functions of B cells.
Collapse
MESH Headings
- Animals
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- B-Lymphocyte Subsets/cytology
- B-Lymphocyte Subsets/immunology
- B-Lymphocyte Subsets/metabolism
- Chemokines/analysis
- Chemokines/blood
- Cytokines/analysis
- Cytokines/blood
- Diet, High-Fat
- Immunoassay
- Immunoglobulin M/blood
- Inflammation/pathology
- Lectins/deficiency
- Lectins/genetics
- Lectins/metabolism
- Leukocyte Common Antigens/metabolism
- Lipoproteins, LDL/blood
- Lipoproteins, LDL/immunology
- Lipoproteins, LDL/metabolism
- Liver/metabolism
- Liver/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Peritonitis/prevention & control
- Peritonitis/veterinary
- RNA, Messenger/metabolism
- Receptors, Antigen, B-Cell/deficiency
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/metabolism
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
- Serum Amyloid A Protein/analysis
- Sialic Acid Binding Immunoglobulin-like Lectins
Collapse
Affiliation(s)
- Sabrina Gruber
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Tim Hendrikx
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria; Department of Molecular Genetics, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6229 ER Maastricht, the Netherlands
| | - Dimitrios Tsiantoulas
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Maria Ozsvar-Kozma
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Laura Göderle
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, CB2 0SZ Cambridge, UK
| | - Joseph L Witztum
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA 92110, USA
| | - Ronit Shiri-Sverdlov
- Department of Molecular Genetics, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6229 ER Maastricht, the Netherlands
| | - Lars Nitschke
- Division of Genetics, Department of Biology, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Christoph J Binder
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria.
| |
Collapse
|