Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Arterioscler Thromb Vasc Biol ; 40(11): 2598-2604, 2020 11.
Article in English | MEDLINE | ID: mdl-32907369

ABSTRACT

OBJECTIVE: NR4A orphan receptors have been well studied in vascular and myeloid cells where they play important roles in the regulation of inflammation in atherosclerosis. NR4A1 (nerve growth factor IB) is among the most highly induced transcription factors in B cells following BCR (B-cell receptor) stimulation. Given that B cells substantially contribute to the development of atherosclerosis, we examined whether NR4A1 regulates B-cell function during atherogenesis. Approach and Results: We found that feeding Ldlr-/- mice a Western diet substantially increased Nr4a1 expression in marginal zone B (MZB) cells compared with follicular B cells. We then generated Ldlr-/- mice with complete B- or specific MZB-cell deletion of Nr4a1. Complete B-cell deletion of Nr4a1 led to increased atherosclerosis, which was accompanied by increased T follicular helper cell-germinal center axis response, as well as increased serum total cholesterol and triglycerides levels. Interestingly, specific MZB-cell deletion of Nr4a1 increased atherosclerosis in association with an increased T follicular helper-germinal center response but without any impact on serum cholesterol or triglyceride levels. Nr4a1-/- MZB cells showed decreased PDL1 (programmed death ligand-1) expression, which may have contributed to the enhanced T follicular helper response. CONCLUSIONS: Our findings reveal a previously unsuspected role for NR4A1 in the atheroprotective role of MZB cells.


Subject(s)
Aorta/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , B-Lymphocytes/metabolism , Gene Deletion , Lymphoid Tissue/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/deficiency , Animals , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , B-Lymphocytes/pathology , Disease Models, Animal , Disease Progression , Lymphoid Tissue/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Plaque, Atherosclerotic , Receptors, LDL/deficiency , Receptors, LDL/genetics , Signal Transduction
2.
Arterioscler Thromb Vasc Biol ; 39(7): 1379-1389, 2019 07.
Article in English | MEDLINE | ID: mdl-31092015

ABSTRACT

Objective- Investigate the impact of modulating B cell FcγRIIb (Fcγ receptor IIb) expression on atherosclerosis. Approach and Results- Western diet-induced atherosclerosis was assessed in Ldlr-/- or Apoe-/- mice with B cell-specific overexpression of FcγRIIb or with an FcγRIIb promoter mutation that alters FcγRIIb expression in germinal center (GC) B cells. In males, overexpression of FcγRIIb on B cells severely reduced activated, class switched B cell responses, as indicated by reductions in GC B cells, plasma cells, and serum IgG but not IgM antibodies. Male mice overexpressing FcγRIIb developed less atherosclerosis, suggesting a pathogenic role for GC B cell IgG responses. In support of this hypothesis, male mice with a promoter polymorphism-driven reduction in FcγRIIb on GC B cells but not plasma cells have a converse phenotype of enhanced GC responses and IgG2c antibodies and enhanced atherosclerosis. IgG2c significantly enhanced TNF (tumor necrosis factor) secretion by CD11b+ CD11c+ cells expressing the high-affinity receptor FcγRIV. In females, overexpression of FcγRIIb on B cells not only reduced GC B cell responses but also substantially reduced B-1 cells and IgM antibodies, which translated into acceleration of atherosclerosis. Promoter-driven reduction in FcγRIIb did not alter GC B cell responses in females and, therefore, had no impact on atherosclerosis. Conclusions- B cell FcγRIIb differentially alters proatherogenic adaptive GC B cell and atheroprotective innate B-1 responses in male and female mice fed a western diet. Our results highlight the importance of a better understanding and ability to selectively target B cell responses in future immunotherapeutic approaches against human cardiovascular disease. Visual Overview- An online visual overview is available for this article.


Subject(s)
Atherosclerosis/immunology , B-Lymphocytes/immunology , Germinal Center/immunology , Receptors, IgG/physiology , Animals , Apolipoproteins E/physiology , Female , Immunity, Innate , Immunoglobulin M/biosynthesis , Male , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/biosynthesis
3.
Circulation ; 138(20): 2263-2273, 2018 11 13.
Article in English | MEDLINE | ID: mdl-29858401

ABSTRACT

BACKGROUND: Atherosclerotic cardiovascular disease (heart attacks and strokes) is the major cause of death globally and is caused by the buildup of a plaque in the arterial wall. Genomic data showed that the B cell-activating factor (BAFF) receptor pathway, which is specifically essential for the survival of conventional B lymphocytes (B-2 cells), is a key driver of coronary heart disease. Deletion or antibody-mediated blockade of BAFF receptor ablates B-2 cells and decreases experimental atherosclerosis. Anti-BAFF immunotherapy is approved for treatment of autoimmune systemic lupus erythematosus, and can therefore be expected to limit their associated cardiovascular risk. However, direct effects of anti-BAFF immunotherapy on atherosclerosis remain unknown. METHODS: To investigate the effect of BAFF neutralization in atherosclerosis, the authors treated Apoe-/- and Ldlr-/- mice with a well-characterized blocking anti-BAFF antibody. Moreover, to investigate the mechanism by which BAFF impacts atherosclerosis, the authors studied atherosclerosis-prone mice that lack the alternative receptor for BAFF: transmembrane activator and calcium modulator and cyclophilin ligand interactor. RESULTS: The authors demonstrate here that anti-BAFF antibody treatment increased atherosclerosis in mice, despite efficient depletion of mature B-2 cells, suggesting a unique mechanism of action. Indeed, myeloid cell-specific deletion of transmembrane activator and calcium modulator and cyclophilin ligand interactor also results in increased atherosclerosis, while B cell-specific transmembrane activator and calcium modulator and cyclophilin ligand interactor deletion had no effect. Mechanistically, BAFF-transmembrane activator and calcium modulator and cyclophilin ligand interactor signaling represses macrophage IRF7-dependent (but not NF-κB-dependent) Toll-like receptor 9 responses including proatherogenic CXCL10 production. CONCLUSIONS: These data identify a novel B cell-independent anti-inflammatory role for BAFF in atherosclerosis and may have important clinical implications.


Subject(s)
Antibodies/therapeutic use , Atherosclerosis/therapy , B-Cell Activating Factor/immunology , Animals , Antibodies/immunology , Aorta/pathology , Bone Marrow Cells/cytology , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Cholesterol/blood , Immunotherapy , Interferon Regulatory Factor-7/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Toll-Like Receptor 9/metabolism , Transmembrane Activator and CAML Interactor Protein/genetics , Transmembrane Activator and CAML Interactor Protein/metabolism
4.
Circ Res ; 121(3): 270-281, 2017 Jul 21.
Article in English | MEDLINE | ID: mdl-28620068

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.


Subject(s)
Atherosclerosis/metabolism , B-Lymphocytes/metabolism , T-Lymphocytes/metabolism , X-Box Binding Protein 1/deficiency , Animals , Atherosclerosis/immunology , Atherosclerosis/pathology , B-Lymphocytes/immunology , Immunity, Humoral/physiology , Male , Mice , Mice, Knockout , Plasma Cells/immunology , Plasma Cells/metabolism , T-Lymphocytes/immunology , X-Box Binding Protein 1/immunology
5.
Circ Res ; 114(10): 1640-60, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24812352

ABSTRACT

Chronic inflammation in response to lipoprotein accumulation in the arterial wall is central in the development of atherosclerosis. Both innate and adaptive immunity are involved in this process. Adaptive immune responses develop against an array of potential antigens presented to effector T lymphocytes by antigen-presenting cells, especially dendritic cells. Functional analysis of the role of different T-cell subsets identified the Th1 responses as proatherogenic, whereas regulatory T-cell responses exert antiatherogenic activities. The effect of Th2 and Th17 responses is still debated. Atherosclerosis is also associated with B-cell activation. Recent evidence established that conventional B-2 cells promote atherosclerosis. In contrast, innate B-1 B cells offer protection through secretion of natural IgM antibodies. This review discusses the recent development in our understanding of the role of T- and B-cell subsets in atherosclerosis and addresses the role of dendritic cell subpopulations in the control of adaptive immunity.


Subject(s)
Adaptive Immunity/immunology , Atherosclerosis/immunology , B-Lymphocyte Subsets/immunology , Dendritic Cells/immunology , T-Lymphocyte Subsets/immunology , Animals , Atherosclerosis/pathology , B-Lymphocyte Subsets/pathology , Dendritic Cells/pathology , Humans , T-Lymphocyte Subsets/pathology
6.
Arterioscler Thromb Vasc Biol ; 35(2): 296-302, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25359862

ABSTRACT

Atherosclerotic plaque formation is strongly influenced by different arms of the immune system, including B lymphocytes. B cells are divided into 2 main families: the B1 and the B2 cells. B1 cells are atheroprotective mainly via the production of natural IgM antibodies that bind oxidized low-density lipoprotein and apoptotic cells. B2 cells, which include follicular and marginal zone B cells, are suggested to be proatherogenic. Antibody-mediated depletion of B cells has become a valuable treatment option for certain autoimmune diseases, such as systemic lupus erythematosus and rheumatoid arthritis that are also characterized by the development of premature atherosclerosis. Thus, B cells represent a novel interesting target for therapeutic modulation of the atherosclerotic disease process. Here, we discuss the effect of different of B-cell subsets in experimental atherosclerosis, their mechanism of action as well as potential ways to exploit these findings for the treatment of human disease.


Subject(s)
Atherosclerosis/drug therapy , B-Lymphocyte Subsets/drug effects , Immunologic Factors/therapeutic use , Animals , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Disease Models, Animal , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Molecular Targeted Therapy , Myocardial Infarction/drug therapy , Myocardial Infarction/immunology , Myocardial Infarction/metabolism , Phenotype , Signal Transduction/drug effects , Translational Research, Biomedical
7.
Arterioscler Thromb Vasc Biol ; 35(8): 1770-3, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26088575

ABSTRACT

OBJECTIVE: To determine the role of regulatory B cell-derived interleukin (IL)-10 in atherosclerosis. APPROACH AND RESULTS: We created chimeric Ldlr(-/-) mice with a B cell-specific deficiency in IL-10, and confirmed that purified B cells stimulated with lipopolysaccharide failed to produce IL-10 compared with control Ldlr(-/-) chimeras. Mice lacking B-cell IL-10 demonstrated enhanced splenic B-cell numbers but no major differences in B-cell subsets, T cell or monocyte distribution, and unchanged body weights or serum cholesterol levels compared with control mice. After 8 weeks on high-fat diet, there were no differences in aortic root or aortic arch atherosclerosis. In addition to plaque size, plaque composition (macrophages, T cells, smooth muscle cells, and collagen) was similar between groups. CONCLUSIONS: In contrast to its prominent regulatory role in many immune-mediated diseases and its proposed modulatory role in atherosclerosis, B cell-derived IL-10 does not alter atherosclerosis in mice.


Subject(s)
Aorta/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , B-Lymphocytes, Regulatory/metabolism , Interleukin-10/metabolism , Animals , Aorta/immunology , Aorta/pathology , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/pathology , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , B-Lymphocytes, Regulatory/immunology , Biomarkers/blood , Cells, Cultured , Diet, High-Fat , Disease Models, Animal , Interleukin-10/deficiency , Interleukin-10/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic , Receptors, LDL/deficiency , Receptors, LDL/genetics , Time Factors
8.
Arterioscler Thromb Vasc Biol ; 35(4): 817-28, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25614278

ABSTRACT

OBJECTIVE: Atherosclerosis, the cause of 50% of deaths in westernized societies, is widely regarded as a chronic vascular inflammatory disease. Vascular smooth muscle cell (VSMC) inflammatory activation in response to local proinflammatory stimuli contributes to disease progression and is a pervasive feature in developing atherosclerotic plaques. Therefore, it is of considerable therapeutic importance to identify mechanisms that regulate the VSMC inflammatory response. APPROACH AND RESULTS: We report that myocardin, a powerful myogenic transcriptional coactivator, negatively regulates VSMC inflammatory activation and vascular disease. Myocardin levels are reduced during atherosclerosis, in association with phenotypic switching of smooth muscle cells. Myocardin deficiency accelerates atherogenesis in hypercholesterolemic apolipoprotein E(-/-) mice. Conversely, increased myocardin expression potently abrogates the induction of an array of inflammatory cytokines, chemokines, and adhesion molecules in VSMCs. Expression of myocardin in VSMCs reduces lipid uptake, macrophage interaction, chemotaxis, and macrophage-endothelial tethering in vitro, and attenuates monocyte accumulation within developing lesions in vivo. These results demonstrate that endogenous levels of myocardin are a critical regulator of vessel inflammation. CONCLUSIONS: We propose myocardin as a guardian of the contractile, noninflammatory VSMC phenotype, with loss of myocardin representing a critical permissive step in the process of phenotypic transition and inflammatory activation, at the onset of vascular disease.


Subject(s)
Atherosclerosis/metabolism , Carotid Artery Injuries/metabolism , Inflammation/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Carotid Artery Injuries/genetics , Carotid Artery Injuries/immunology , Carotid Artery Injuries/pathology , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cells, Cultured , Chemokines/metabolism , Chemotaxis , Cytokines/metabolism , Disease Models, Animal , Genotype , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Lipid Metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/pathology , Neointima , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Phenotype , RNA Interference , Rats, Wistar , Time Factors , Trans-Activators/deficiency , Trans-Activators/genetics , Transfection
9.
Circulation ; 130(16): 1363-73, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25223984

ABSTRACT

BACKGROUND: Plasmacytoid dendritic cells (pDCs) bridge innate and adaptive immune responses and are important regulators of immuno-inflammatory diseases. However, their role in atherosclerosis remains elusive. METHODS AND RESULTS: Here, we used genetic approaches to investigate the role of pDCs in atherosclerosis. Selective pDC deficiency in vivo was achieved using CD11c-Cre × Tcf4(-/flox) bone marrow transplanted into Ldlr(-/-) mice. Compared with control Ldlr(-/-) chimeric mice, CD11c-Cre × Tcf4(-/flox) mice had reduced atherosclerosis levels. To begin to understand the mechanisms by which pDCs regulate atherosclerosis, we studied chimeric Ldlr(-/-) mice with selective MHCII deficiency on pDCs. Significantly, these mice also developed reduced atherosclerosis compared with controls without reductions in pDC numbers or changes in conventional DCs. MHCII-deficient pDCs showed defective stimulation of apolipoprotein B100-specific CD4(+) T cells in response to native low-density lipoprotein, whereas production of interferon-α was not affected. Finally, the atheroprotective effect of selective MHCII deficiency in pDCs was associated with significant reductions of proatherogenic T cell-derived interferon-γ and lesional T cell infiltration, and was abrogated in CD4(+) T cell-depleted animals. CONCLUSIONS: This study supports a proatherogenic role for pDCs in murine atherosclerosis and identifies a critical role for MHCII-restricted antigen presentation by pDCs in driving proatherogenic T cell immunity.


Subject(s)
Antigen-Presenting Cells/immunology , Atherosclerosis/immunology , Atherosclerosis/pathology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class II/immunology , Adaptive Immunity/immunology , Animals , Aorta/cytology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/immunology , CD4-Positive T-Lymphocytes/cytology , Cell Communication/immunology , Cells, Cultured , Dendritic Cells/cytology , Flow Cytometry , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL/genetics , Receptors, LDL/immunology , Transcription Factor 4
10.
Cardiovasc Res ; 120(3): 318-328, 2024 03 14.
Article in English | MEDLINE | ID: mdl-38381113

ABSTRACT

AIMS: The adaptive immune response plays an important role in atherosclerosis. In response to a high-fat/high-cholesterol (HF/HC) diet, marginal zone B (MZB) cells activate an atheroprotective programme by regulating the differentiation and accumulation of 'poorly differentiated' T follicular helper (Tfh) cells. On the other hand, Tfh cells activate the germinal centre response, which promotes atherosclerosis through the production of class-switched high-affinity antibodies. We therefore investigated the direct role of Tfh cells and the role of IL18 in Tfh differentiation in atherosclerosis. METHODS AND RESULTS: We generated atherosclerotic mouse models with selective genetic deletion of Tfh cells, MZB cells, or IL18 signalling in Tfh cells. Surprisingly, mice lacking Tfh cells had increased atherosclerosis. Lack of Tfh not only reduced class-switched IgG antibodies against oxidation-specific epitopes (OSEs) but also reduced atheroprotective natural IgM-type anti-phosphorylcholine (PC) antibodies, despite no alteration of natural B1 cells. Moreover, the absence of Tfh cells was associated with an accumulation of MZB cells with substantially reduced ability to secrete antibodies. In the same manner, MZB cell deficiency in Ldlr-/- mice was associated with a significant decrease in atheroprotective IgM antibodies, including natural anti-PC IgM antibodies. In humans, we found a positive correlation between circulating MZB-like cells and anti-OSE IgM antibodies. Finally, we identified an important role for IL18 signalling in HF/HC diet-induced Tfh. CONCLUSION: Our findings reveal a previously unsuspected role of MZB cells in regulating atheroprotective 'natural' IgM antibody production in a Tfh-dependent manner, which could have important pathophysiological and therapeutic implications.


Subject(s)
Atherosclerosis , Interleukin-18 , Humans , Mice , Animals , Immunoglobulin M , B-Lymphocytes , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Cholesterol , T-Lymphocytes, Helper-Inducer
12.
Arterioscler Thromb Vasc Biol ; 32(7): 1573-6, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22426131

ABSTRACT

OBJECTIVE: The goal of this study was to assess the role of B-cell activating factor (BAFF) receptor in B-cell regulation of atherosclerosis. METHODS AND RESULTS: Male LDL receptor-deficient mice (Ldlr(-/-)) were lethally irradiated and reconstituted with either wild type or BAFF receptor (BAFF-R)-deficient bone marrow. After 4 weeks of recovery, mice were put on a high-fat diet for 6 or 8 weeks. BAFF-R deficiency in bone marrow cells led to a marked reduction of conventional mature B2 cells but did not affect the B1a cell subtype. This was associated with a significant reduction of dendritic cell activation and T-cell proliferation along with a reduction of IgG antibodies against malondialdehyde-modified low-density lipoprotein. In contrast, serum IgM type antibodies were preserved. Interestingly, BAFF-R deficiency was associated with a significant reduction in atherosclerotic lesion development and reduced numbers of plaque T cells. Selective BAFF-R deficiency on B cells led to a similar reduction in lesion size and T-cell infiltration but in contrast did not affect dendritic cell activation. CONCLUSIONS: BAFF-R deficiency in mice selectively alters mature B2 cell-dependent cellular and humoral immune responses and limits the development of atherosclerosis.


Subject(s)
Atherosclerosis/etiology , Animals , B-Cell Activation Factor Receptor/physiology , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology
13.
Cardiovasc Res ; 118(3): 872-882, 2022 02 21.
Article in English | MEDLINE | ID: mdl-33783498

ABSTRACT

AIMS: In pre-clinical models of acute myocardial infarction (MI), mature B cells mobilize inflammatory monocytes into the heart, leading to increased infarct size and deterioration of cardiac function, whilst anti-CD20 antibody-mediated depletion of B cells limits myocardial injury and improves cardiac function. Rituximab is a monoclonal anti-CD20 antibody targeted against human B cells. However, its use in cardiovascular disease is untested and is currently contraindicated. Therefore, we assessed the safety, feasibility, and pharmacodynamic effect of rituximab given to patients with acute ST-elevation MI (STEMI). METHODS AND RESULTS: Rituximab in patients with acute ST-elevation myocardial infarction (RITA-MI) was a prospective, open-label, dose-escalation, single-arm, phase 1/2a clinical trial, which tested rituximab administered as a single intravenous dose in patients with STEMI within 48 h of symptom onset. Four escalating doses (200, 500, 700, and 1000 mg) were used. The primary endpoint was safety, whilst secondary endpoints were changes in circulating immune cell subsets including B cells, and cardiac and inflammatory biomarkers. A total of 24 patients were dosed. Rituximab appeared well tolerated. Seven serious adverse events were reported, none of which were assessed as being related to the rituximab infusion. Rituximab caused a mean 96.3% (95% confidence interval 93.8-98.8%) depletion of circulating B cells within 30 min of starting the infusion. Maximal B-cell depletion was seen at Day 6, which was significantly lower than baseline for all doses (P < 0.001). B-cell repopulation at 6 months was dose-dependent, with modulation of returning B-cell subsets. Immunoglobulin (IgG, IgM, and IgA) levels were not affected during the 6 months of follow-up. CONCLUSIONS: A single infusion of rituximab appears safe when given in the acute STEMI setting and substantially alters circulating B-cell subsets. We provide important new insight into the feasibility and pharmacodynamics of rituximab in acute STEMI, which will inform further clinical translation of this potential therapy. CLINICAL TRIAL REGISTRATION: NCT03072199 at https://www.clinicaltrials.gov/.


Subject(s)
Biomedical Research , Myocardial Infarction , ST Elevation Myocardial Infarction , Humans , Prospective Studies , Rituximab/adverse effects , ST Elevation Myocardial Infarction/diagnosis , ST Elevation Myocardial Infarction/drug therapy , Treatment Outcome
14.
NEJM Evid ; 1(1): EVIDoa2100009, 2022 01.
Article in English | MEDLINE | ID: mdl-38319239

ABSTRACT

BACKGROUND: Atherosclerosis is a chronic inflammatory disease of the artery wall. Regulatory T cells (Tregs) limit inflammation and promote tissue healing. Low doses of interleukin (IL)-2 have the potential to increase Tregs, but its use is contraindicated for patients with ischemic heart disease. METHODS: In this randomized, double-blind, placebo-controlled, dose-escalation trial, we tested low-dose subcutaneous aldesleukin (recombinant IL-2), given once daily for 5 consecutive days. In study part A, the primary end point was safety, and patients with stable ischemic heart disease were randomly assigned to receive placebo or to one of five dose groups (range, 0.3 to 3.0 × 106 IU daily). In study part B, patients with acute non-ST elevation myocardial infarction or unstable angina were randomly assigned to receive placebo or to one of two dose groups (1.5 and 2.5 × 106 IU daily). The coprimary end points were safety and the dose required to increase circulating Tregs by 75%. Single-cell RNA-sequencing of circulating immune cells was used to provide a mechanistic assessment of the effects of aldesleukin. RESULTS: Forty-four patients were randomly assigned to either study part A (n=26) or part B (n=18). In total, 3 patients withdrew before dosing, 27 received active treatment, and 14 received placebo. The majority of adverse events were mild. Two serious adverse events occurred, with one occurring after drug administration. In parts A and B, there was a dose-dependent increase in Tregs. In part B, the estimated dose to achieve a 75% increase in Tregs was 1.46 × 106 IU (95% confidence interval, 1.06 to 1.87). Single-cell RNA-sequencing demonstrated the engagement of distinct pathways and cell­cell interactions. CONCLUSIONS: In this phase 1b/2a study, low-dose IL-2 expanded Tregs without adverse events of major concern. Larger trials are needed to confirm the safety and to further evaluate the efficacy of low-dose IL-2 as an anti-inflammatory therapy for patients with ischemic heart disease. (Funded by the Medical Research Council, the British Heart Foundation, and others; ClinicalTrials.gov number, NCT03113773)


Subject(s)
Interleukin-2 , Interleukin-2/analogs & derivatives , Myocardial Ischemia , T-Lymphocytes, Regulatory , Humans , Interleukin-2/administration & dosage , Interleukin-2/therapeutic use , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Myocardial Ischemia/immunology , Myocardial Ischemia/drug therapy , Double-Blind Method , Male , Middle Aged , Female , Recombinant Proteins
15.
Kidney Int ; 79(4): 414-22, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20944546

ABSTRACT

Vascular calcification, which contributes to cardiovascular disease in patients with uremic hyperphosphatemia, is associated with vascular cell expression of osteogenic genes, including bone morphogenetic protein (BMP)-2 and osteopontin (OPN). High inorganic phosphate levels in vitro stimulate the osteogenic conversion of smooth muscle cells; however, the mechanism governing this is not clear. We found that high-phosphate medium increased the expression of BMP-2 and OPN in mouse smooth muscle cells in culture. However, this effect was lost in the presence of the mineralization inhibitor, pyrophosphate, suggesting a contribution of calcium phosphate crystals. Addition of 1-2 mmol/l phosphate alone to growth medium was sufficient to induce nanosized crystals after 1 day at 37 °C. Isolated crystals were about 160 nm in diameter and had a calcium to phosphate ratio of 1.35, consistent with the hydroxyapatite precursor octacalcium phosphate. Nanocrystal formation increased fourfold in the absence of serum, was blocked by fetuin-A, and was dependent on time and on the concentrations of phosphate and calcium. Purified synthetic hydroxyapatite nanocrystals and isolated high-phosphate-induced nanocrystals, but not nanocrystal-free high-phosphate medium, also induced BMP-2 and OPN. Thus, our results suggest that BMP-2 and OPN are induced by calcium phosphate nanocrystals, rather than soluble phosphate. This mechanism may contribute, in part, to hyperphosphatemia-related vascular cell differentiation and calcification.


Subject(s)
Bone Morphogenetic Protein 2/genetics , Hyperphosphatemia/genetics , Hyperphosphatemia/metabolism , Myocytes, Smooth Muscle/metabolism , Nanoparticles/chemistry , Osteopontin/genetics , Animals , Base Sequence , Calcinosis/etiology , Calcinosis/metabolism , Calcinosis/pathology , Calcium Phosphates/metabolism , Calcium Phosphates/pharmacology , Cell Differentiation , Cell Line , DNA Primers/genetics , Hyperphosphatemia/complications , In Vitro Techniques , Mice , Microscopy, Electron, Scanning , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Nanoparticles/ultrastructure , Osteogenesis/genetics , Up-Regulation/drug effects
17.
J Lipid Res ; 51(5): 1010-6, 2010 May.
Article in English | MEDLINE | ID: mdl-19965598

ABSTRACT

Epidemiological evidence suggests that cardiovascular disease is associated with osteoporosis, independent of age. Bone resorptive surface is increased in mice on a high-fat diet, and osteoclastic differentiation of bone marrow preosteoclasts is promoted by oxidized phospholipids. Because osteoclastic differentiation requires cytokines produced by osteoblasts, we hypothesized that the stimulatory mechanism of oxidized phospholipids is via induction of osteoclast-regulating cytokines in osteoblasts. To investigate the effects of oxidized phospholipids on expression of such cytokines, murine calvarial preosteoblasts, MC3T3-E1, were treated with oxidized 1-palmitoyl-2-arachidonyl-sn-glycero-3-phosphocholine (ox-PAPC), an active component of oxidized lipoproteins. Results showed that ox-PAPC increased expression of interleukin-6 (IL-6) and tumor necrosis factor-alpha. IL-6 expression was also elevated in calvarial tissues from hyperlipidemic but not in wild-type mice. Ox-PAPC also induced IL-6 protein levels in both MC3T3-E1 and primary calvarial cells. Promoter-reporter assay analysis showed that ox-PAPC, but not PAPC, induced murine IL-6 promoter activity. Effects of ox-PAPC on IL-6 expression and the promoter activity were attenuated by H89, a PKA inhibitor. Analysis of deletion and mutant IL-6 promoter constructs suggested that CAAT/enhancer binding protein (C/EBP) partly mediates the ox-PAPC effects. Taken together, the data suggest that oxidized phospholipids induce IL-6 expression in osteoblasts in part via C/EBP.


Subject(s)
Gene Expression Regulation/drug effects , Interleukin-6/genetics , Osteoblasts/drug effects , Osteoblasts/metabolism , Phospholipids/metabolism , Phospholipids/pharmacology , Animals , CCAAT-Enhancer-Binding Proteins/genetics , Cell Line , Intracellular Space/drug effects , Intracellular Space/metabolism , Mice , Osteoblasts/cytology , Oxidation-Reduction , Phosphatidylcholines/metabolism , Phosphatidylcholines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Response Elements/genetics , Transcription, Genetic
18.
Circ Res ; 100(4): 502-9, 2007 Mar 02.
Article in English | MEDLINE | ID: mdl-17255529

ABSTRACT

The calcification of blood vessels correlates with increased morbidity and mortality in patients with atherosclerosis, diabetes, and end-stage kidney disease. The receptor tyrosine kinase Axl is emerging as an important regulator of adult mammalian physiology and pathology. This study tests the hypothesis that Axl prevents the deposition of a calcified matrix by vascular smooth muscle cells (VSMCs) and that this occurs via the phosphatidylinositol 3-kinase (PI3K) signaling pathway. First, we demonstrate that Axl is expressed and phosphorylated in confluent VSMCs and that its expression is markedly downregulated as these cells calcify their matrix. Second, we demonstrate that overexpression of wild-type Axl, using recombinant adenoviruses, enhances Axl phosphorylation and downstream signaling via PI3K and Akt. Furthermore, overexpression of Axl significantly inhibits mineral deposition by VSMCs, as assessed by alizarin red staining and (45)Ca accumulation. Third, the addition of a PI3K inhibitor, wortmannin, negates the inhibition of mineralization by overexpression of wild-type Axl, suggesting that activation of downstream signaling via PI3K is crucial for its inhibitory activity. In contrast, Axl-mediated signaling is not enhanced by overexpression of kinase-dead Axl and mineralization is accelerated, although beta-glycerophosphate is still required for this effect. Finally, the caspase inhibitor zVAD.fmk attenuates the increased mineralization induced by kinase-dead Axl, suggesting that kinase-dead Axl stimulates mineralization by inhibiting the antiapoptotic effect of endogenous Axl. Together, these results demonstrate that signaling through Axl inhibits vascular calcification in vitro and suggest that therapeutics targeting this receptor may open up new avenues for the prevention of vascular calcification in vivo.


Subject(s)
Calcinosis/enzymology , Calcinosis/prevention & control , Calcium/metabolism , Muscle, Smooth, Vascular/enzymology , Oncogene Proteins/biosynthesis , Phosphatidylinositol 3-Kinases/physiology , Receptor Protein-Tyrosine Kinases/biosynthesis , Signal Transduction/physiology , Animals , Calcinosis/genetics , Calcium/antagonists & inhibitors , Cattle , Cells, Cultured , Humans , Mice , Muscle, Smooth, Vascular/pathology , Oncogene Proteins/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction/genetics , Axl Receptor Tyrosine Kinase
19.
Nat Rev Cardiol ; 16(3): 180-196, 2019 03.
Article in English | MEDLINE | ID: mdl-30410107

ABSTRACT

The cardiovascular system is subject to hyperlipidaemic, inflammatory, and pro-oxidant stressors. Over time, these factors drive prevalent chronic diseases, of which atherosclerosis is most prominent and accounts for the majority of deaths globally. Antibody-producing B cells perform a unique role in responses to stress, injury, and infection. The power, inducibility, and adaptability of the antibody repertoire require an equally complex range of control measures. Defects and chronic perturbations in these checkpoints lead to inappropriate antibody responses, which might have important roles in shaping the development and outcome of atherosclerotic disease. A unique aspect related to atherosclerosis is the prominent role of natural antibodies, specifically those binding to the oxidized epitopes that are abundant on modified lipoproteins and cellular debris. B cells control cellular immune responses through cell-cell contact, antigen presentation, and cytokine production, and thereby participate in systemic and local immune responses in atherosclerotic arteries. To date, both proatherogenic and antiatherogenic properties have been assigned to B cells, depending on subsets and how they are functionally targeted. For these reasons, a deeper understanding of how B cells influence atherosclerotic plaque development is being pursued with the hope of providing novel B cell-targeted interventions to prevent inflammation-driven cardiovascular events.


Subject(s)
Atherosclerosis/metabolism , B-Lymphocyte Subsets/metabolism , Dyslipidemias/metabolism , Plaque, Atherosclerotic , Animals , Atherosclerosis/immunology , Atherosclerosis/pathology , Atherosclerosis/therapy , Autoimmunity , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , Dyslipidemias/immunology , Dyslipidemias/therapy , Humans , Immunotherapy , Phenotype , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, B-Cell/metabolism , Signal Transduction
20.
Biochem Biophys Res Commun ; 374(3): 553-8, 2008 Sep 26.
Article in English | MEDLINE | ID: mdl-18655772

ABSTRACT

Vascular calcification is associated with increased cardiovascular risk and occurs by osteochondrogenic differentiation of vascular cells. Many of the same regulatory factors that control skeletal mineralization, including the complex metabolic pathway controlling levels of the activator, inorganic phosphate, and the potent inhibitor, pyrophosphate, also govern vascular calcification. We previously found that the cAMP/PKA signaling pathway mediates in vitro vascular cell calcification induced by inflammatory factors including tumor necrosis factor-alpha 1 and oxidized phospholipids. In this report, we tested whether this signaling pathway modulates phosphate and pyrophosphate metabolism. Treatment of primary murine aortic cells with the PKA activator, forskolin, significantly induced osteoblastic differentiation markers, including alkaline phosphatase (ALP), osteopontin, and osteocalcin as well as the pyrophosphate generator, ectonucleotide-pyrophosphatase/phosphodiesterase-1 (Enpp1) and the pyrophosphate transporter, ankylosis protein, but not the sodium/phosphate cotransporter, Pit-1. In the presence of a substrate for ALP, beta-glycerophosphate, which generates inorganic phosphate, forskolin also enhanced matrix mineralization. Inhibitors of ALP or Pit-1 abrogated forskolin-induced osteopontin expression and mineralization but not forskolin-induced osteocalcin or ALP. These results suggest that phosphate is necessary for PKA-induced calcification of vascular cells and that the extent of PKA-induced calcification is controlled by feedback induction of the inhibitor, pyrophosphate.


Subject(s)
Aorta/metabolism , Calcinosis/etiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Diphosphates/metabolism , Hyperparathyroidism/complications , Phosphates/metabolism , Alkaline Phosphatase/metabolism , Animals , Aorta/drug effects , Aorta/pathology , Biological Transport , Calcinosis/metabolism , Calcinosis/pathology , Cell Differentiation , Cells, Cultured , Colforsin/pharmacology , Hyperparathyroidism/metabolism , Mice , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/pathology , Osteocalcin/metabolism , Osteopontin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL