Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
FASEB J ; 37(4): e22846, 2023 04.
Article in English | MEDLINE | ID: mdl-36856983

ABSTRACT

Colchicine is a broad-acting anti-inflammatory agent that has attracted interest for repurposing in atherosclerotic cardiovascular disease. Here, we studied its ability at a human equivalent dose of 0.5 mg/day to modify plaque formation and composition in murine atherosclerosis and investigated its actions on macrophage responses to atherogenic stimuli in vitro. In atherosclerosis induced by high-cholesterol diet, Apoe-/- mice treated with colchicine had 50% reduction in aortic oil Red O+ plaque area compared to saline control (p = .001) and lower oil Red O+ staining of aortic sinus lesions (p = .03). In vitro, addition of 10 nM colchicine inhibited foam cell formation from murine and human macrophages after treatment with oxidized LDL (ox-LDL). Mechanistically, colchicine downregulated glycosylation and surface expression of the ox-LDL uptake receptor, CD36, and reduced CD36+ staining in aortic sinus plaques. It also decreased macrophage uptake of cholesterol crystals, resulting in lower intracellular lysosomal activity, inhibition of the NLRP3 inflammasome, and reduced secretion of IL-1ß and IL-18. Colchicine's anti-atherosclerotic actions were accentuated in a mouse model of unstable plaque induced by carotid artery tandem stenosis surgery, where it decreased lesion size by 48% (p = .01), reduced lipid (p = .006) and necrotic core area (p = .007), increased collagen content and cap-to-necrotic core ratio (p = .05), and attenuated plaque neutrophil extracellular traps (p < .001). At low dose, colchicine's effects were not accompanied by the evidence of microtubule depolymerization. Together, these results show that colchicine exerts anti-atherosclerotic and plaque-stabilizing effects at low dose by inhibiting foam cell formation and cholesterol crystal-induced inflammation. This provides a new framework to support its repurposing for atherosclerotic cardiovascular disease.


Subject(s)
Atherosclerosis , Cardiovascular Diseases , Carotid Stenosis , Humans , Animals , Mice , Foam Cells , Colchicine , Cholesterol
2.
Commun Biol ; 6(1): 265, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36914713

ABSTRACT

Atherosclerotic plaque rupture leading to myocardial infarction is a major global health burden. Applying the tandem stenosis (TS) mouse model, which distinctively exhibits the characteristics of human plaque instability/rupture, we use quantitative proteomics to understand and directly compare unstable and stable atherosclerosis. Our data highlight the disparate natures and define unique protein signatures of unstable and stable atherosclerosis. Key proteins and pathway networks are identified such as the innate immune system, and neutrophil degranulation. The latter includes calprotectin S100A8/A9, which we validate in mouse and human unstable plaques, and we demonstrate the plaque-stabilizing effects of its inhibition. Overall, we provide critical insights into the unique proteomic landscape of unstable atherosclerosis (as distinct from stable atherosclerosis and vascular tissue). We further establish the TS model as a reliable preclinical tool for the discovery and testing of plaque-stabilizing drugs. Finally, we provide a knowledge resource defining unstable atherosclerosis that will facilitate the identification and validation of long-sought-after therapeutic targets and drugs for plaque stabilization.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Humans , Animals , Mice , Plaque, Atherosclerotic/drug therapy , Proteomics , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Atherosclerosis/metabolism , Disease Models, Animal
3.
Front Immunol ; 13: 1040233, 2022.
Article in English | MEDLINE | ID: mdl-36483558

ABSTRACT

Aims: Cardiac fibrosis is central to heart failure (HF), especially HF with preserved ejection fraction (HFpEF), often caused by hypertension. Despite fibrosis causing diastolic dysfunction and impaired electrical conduction, responsible for arrhythmia-induced sudden cardiac death, the mechanisms are poorly defined and effective therapies are lacking. Here we show that crosstalk between cardiac cytotoxic memory CD8+ T cells and overly stressed cardiomyocytes is essential for development of non-ischemic hypertensive cardiac fibrosis. Methods and results: CD8 T cell depletion in hypertensive mice, strongly attenuated CF, reduced cardiac apoptosis and improved ventricular relaxation. Interaction between cytotoxic memory CD8+ T cells and overly stressed cardiomyocytes is highly dependent on the CD8+ T cells expressing the innate stress-sensing receptor NKG2D and stressed cardiomyocytes expressing the NKG2D activating ligand RAE-1. The interaction between NKG2D and RAE-1 results in CD8+ T cell activation, release of perforin, cardiomyocyte apoptosis, increased numbers of TGF-ß1 expressing macrophages and fibrosis. Deleting NKG2D or perforin from CD8+ T cells greatly attenuates these effects. Activation of the cytoplasmic DNA-STING-TBK1-IRF3 signaling pathway in overly stressed cardiomyocytes is responsible for elevating RAE-1 and MCP-1, a macrophage attracting chemokine. Inhibiting STING activation greatly attenuates cardiomyocyte RAE-1 expression, the cardiomyocyte apoptosis, TGF-ß1 and fibrosis. Conclusion: Our data highlight a novel pathway by which CD8 T cells contribute to an early triggering mechanism in CF development; preventing CD8+ T cell activation by inhibiting the cardiomyocyte RAE-1-CD8+ T cell-NKG2D axis holds promise for novel therapeutic strategies to limit hypertensive cardiac fibrosis.


Subject(s)
Heart Failure , Hypertension , Animals , Mice , Transforming Growth Factor beta1 , Heart Failure/etiology , Stroke Volume , Fibrosis , T-Lymphocytes
4.
Thromb Haemost ; 122(2): 196-207, 2022 02.
Article in English | MEDLINE | ID: mdl-34619795

ABSTRACT

BACKGROUND: 3F7 is a monoclonal antibody targeting the enzymatic pocket of activated factor XII (FXIIa), thereby inhibiting its catalytic activity. Given the emerging role of FXIIa in promoting thromboinflammation, along with its apparent redundancy for hemostasis, the selective inhibition of FXIIa represents a novel and highly attractive approach targeting pathogenic processes that cause thromboinflammation-driven cardiovascular diseases. METHODS: The effects of FXIIa inhibition were investigated using three distinct mouse models of cardiovascular disease-angiotensin II-induced abdominal aortic aneurysm (AAA), an ApoE-/- model of atherosclerosis, and a tandem stenosis model of atherosclerotic plaque instability. 3F7 or its isotype control, BM4, was administered to mice (10 mg/kg) on alternate days for 4 to 8 weeks, depending on the experimental model. Mice were examined for the development and size of AAAs, or the burden and instability of atherosclerosis and associated markers of inflammation. RESULTS: Inhibition of FXIIa resulted in a reduced incidence of larger AAAs, with less acute aortic ruptures and an associated fibro-protective phenotype. FXIIa inhibition also decreased stable atherosclerotic plaque burden and achieved plaque stabilization associated with increased deposition of fibrous structures, a >2-fold thicker fibrous cap, increased cap-to-core ratio, and reduction in localized and systemic inflammatory markers. CONCLUSION: Inhibition of FXIIa attenuates disease severity across three mouse models of thromboinflammation-driven cardiovascular diseases. Specifically, the FXIIa-inhibiting monoclonal antibody 3F7 reduces AAA severity, inhibits the development of atherosclerosis, and stabilizes vulnerable plaques. Ultimately, clinical trials in patients with cardiovascular diseases such as AAA and atherosclerosis are warranted to demonstrate the therapeutic potential of FXIIa inhibition.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Aortic Aneurysm, Abdominal/prevention & control , Atherosclerosis/prevention & control , Factor XIIa/antagonists & inhibitors , Plaque, Atherosclerotic/metabolism , Animals , Aortic Aneurysm, Abdominal/epidemiology , Apolipoproteins E , Disease Models, Animal , Inflammation , Male , Mice
5.
Nat Commun ; 12(1): 6546, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34764270

ABSTRACT

Acute myeloid leukemia (AML) is a malignancy of immature progenitor cells. AML differentiation therapies trigger leukemia maturation and can induce remission, but relapse is prevalent and its cellular origin is unclear. Here we describe high resolution analysis of differentiation therapy response and relapse in a mouse AML model. Triggering leukemia differentiation in this model invariably produces two phenotypically distinct mature myeloid lineages in vivo. Leukemia-derived neutrophils dominate the initial wave of leukemia differentiation but clear rapidly and do not contribute to residual disease. In contrast, a therapy-induced population of mature AML-derived eosinophil-like cells persists during remission, often in extramedullary organs. Using genetic approaches we show that restricting therapy-induced leukemia maturation to the short-lived neutrophil lineage markedly reduces relapse rates and can yield cure. These results indicate that relapse can originate from therapy-resistant mature AML cells, and suggest differentiation therapy combined with targeted eradication of mature leukemia-derived lineages may improve disease outcome.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Neoplasm, Residual/metabolism , Cell Differentiation , Humans , Leukemia, Myeloid, Acute/genetics , Neoplasm, Residual/genetics
6.
Eur Heart J ; 42(9): 938-947, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33338208

ABSTRACT

AIMS: Myocardial infarction (MI) accelerates atherosclerosis and greatly increases the risk of recurrent cardiovascular events for many years, in particular, strokes and MIs. Because B cell-derived autoantibodies produced in response to MI also persist for years, we investigated the role of B cells in adaptive immune responses to MI. METHODS AND RESULTS: We used an apolipoprotein-E-deficient (ApoE-/-) mouse model of MI-accelerated atherosclerosis to assess the importance of B cells. One week after inducing MI in atherosclerotic mice, we depleted B cells using an anti-CD20 antibody. This treatment prevented subsequent immunoglobulin G accumulation in plaques and MI-induced accelerated atherosclerosis. In gain of function experiments, we purified spleen B cells from mice 1 week after inducing MI and transferred these cells into atherosclerotic ApoE-/- mice, which greatly increased immunoglobulin G (IgG) accumulation in plaque and accelerated atherosclerosis. These B cells expressed many cytokines that promote humoural immunity and in addition, they formed germinal centres within the spleen where they differentiated into antibody-producing plasma cells. Specifically deleting Blimp-1 in B cells, the transcriptional regulator that drives their terminal differentiation into antibody-producing plasma cells prevented MI-accelerated atherosclerosis. Alarmins released from infarcted hearts were responsible for activating B cells via toll-like receptors and deleting MyD88, the canonical adaptor protein for inflammatory signalling downstream of toll-like receptors, prevented B-cell activation and MI-accelerated atherosclerosis. CONCLUSION: Our data implicate early B-cell activation and autoantibodies as a central cause for accelerated atherosclerosis post-MI and identifies novel therapeutic strategies towards preventing recurrent cardiovascular events such as MI and stroke.


Subject(s)
Alarmins , Atherosclerosis , B-Lymphocytes , Myocardial Infarction , Plaque, Atherosclerotic , Animals , Atherosclerosis/etiology , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Plasma Cells
7.
Front Immunol ; 10: 3046, 2019.
Article in English | MEDLINE | ID: mdl-31998318

ABSTRACT

Interaction between B and CD4 T cells is crucial for their optimal responses in adaptive immunity. Immune responses augmented by their partnership promote chronic inflammation. Here we report that interaction between B and CD4 T cells augments their atherogenicity to promote lipid-induced atherosclerosis. Genetic deletion of the gene encoding immunoglobulin mu (µ) heavy chain (µMT) in ApoE-/- mice resulted in global loss of B cells including those in atherosclerotic plaques, undetectable immunoglobulins and impaired germinal center formation. Despite unaffected numbers in the circulation and peripheral lymph nodes, CD4 T cells were also reduced in spleens as were activated and memory CD4 T cells. In hyperlipidemic µMT-/- ApoE-/- mice, B cell deficiency decreased atherosclerotic lesions, accompanied by absence of immunoglobulins and reduced CD4 T cell accumulation in lesions. Adoptive transfer of B cells deficient in either MHCII or co-stimulatory molecule CD40, molecules required for B and CD4 T cell interaction, into B cell-deficient µMT-/- ApoE-/- mice failed to increase atherosclerosis. In contrast, wildtype B cells transferred into µMT-/- ApoE-/- mice increased atherosclerosis and increased CD4 T cells in lesions including activated and memory CD4 T cells. Transferred B cells also increased their expression of atherogenic cytokines IL-1ß, TGF-ß, MCP-1, M-CSF, and MIF, with partial restoration of germinal centers and plasma immunoglobulins. Our study demonstrates that interaction between B and CD4 T cells utilizing MHCII and CD40 is essential to augment their function to increase atherosclerosis in hyperlipidemic mice. These findings suggest that targeting B cell and CD4 T cell interaction may be a therapeutic strategy to limit atherosclerosis progression.


Subject(s)
Atherosclerosis/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Animals , Apolipoproteins E/immunology , Cytokines/immunology , Immunoglobulins/immunology , Male , Mice , Mice, Inbred C57BL , Plaque, Atherosclerotic/immunology
8.
J Am Heart Assoc ; 7(13)2018 06 23.
Article in English | MEDLINE | ID: mdl-29936416

ABSTRACT

BACKGROUND: Peritoneal B1a cells attenuate atherosclerosis by secreting natural polyclonal immunoglobulin M (IgM). Regulatory B cells expressing T-cell immunoglobulin mucin domain-1 (TIM-1) expanded through TIM-1 ligation by anti-TIM-1 monoclonal antibody (RMT1-10) induces immune tolerance. METHODS AND RESULTS: We examined the capacity of RMT1-10 to expand peritoneal B1a cells to prevent atherosclerosis development and retard progression of established atherosclerosis. RMT1-10 treatment selectively doubled peritoneal B1a cells, tripled TIM-1+ B1a cells and increased TIM-1+IgM+interleukin (IL)-10+ by 3-fold and TIM-1+IgM+IL-10- B1a cells by 2.5-fold. Similar expansion of B1a B cells was observed in spleens. These effects reduced atherosclerotic lesion size, increased plasma IgM and lesion IgM deposits, and decreased oxidatively modified low-density lipoproteins in lesions. Lesion CD4+ and CD8+ T cells, macrophages and monocyte chemoattractant protein-1, vascular cell adhesion molecule-1, expression of proinflammatory cytokines monocyte chemoattractant protein-1, vascular cell adhesion molecule-1, IL1ß, apoptotic cell numbers and necrotic cores were also reduced. RMT1-10 treatment failed to expand peritoneal B1a cells and reduce atherosclerosis after splenectomy that reduces B1a cells, indicating that these effects are B1a cell-dependent. Apolipoprotein E-KO mice fed a high-fat diet for 6 weeks before treatment with RMT1-10 also increased TIM-1+IgM+IL-10+ and TIM-1+IgM+IL-10- B1a cells and IgM levels and attenuated progression of established atherosclerosis. CONCLUSIONS: RMT1-10 treatment attenuates atherosclerosis development and progression by selectively expanding IgM producing atheroprotective B1a cells. Antibody-based in vivo expansion of B1a cells could be an attractive approach for treating atherosclerosis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , B-Lymphocytes, Regulatory/drug effects , Cell Proliferation/drug effects , Hepatitis A Virus Cellular Receptor 1/antagonists & inhibitors , Immunoglobulin M/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Aorta/immunology , Aorta/metabolism , Aorta/pathology , Aortic Diseases/immunology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apoptosis/drug effects , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/metabolism , Disease Models, Animal , Disease Progression , Hepatitis A Virus Cellular Receptor 1/immunology , Hepatitis A Virus Cellular Receptor 1/metabolism , Immunoglobulin M/immunology , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Necrosis , Plaque, Atherosclerotic , Signal Transduction/drug effects
9.
Arterioscler Thromb Vasc Biol ; 38(5): e71-e84, 2018 05.
Article in English | MEDLINE | ID: mdl-29599140

ABSTRACT

OBJECTIVE: B cells promote or protect development of atherosclerosis. In this study, we examined the role of MHCII (major histocompatibility II), CD40 (cluster of differentiation 40), and Blimp-1 (B-lymphocyte-induced maturation protein) expression by follicular B (FO B) cells in development of atherosclerosis together with the effects of IgG purified from atherosclerotic mice. APPROACH AND RESULTS: Using mixed chimeric Ldlr-/- mice whose B cells are deficient in MHCII or CD40, we demonstrate that these molecules are critical for the proatherogenic actions of FO B cells. During development of atherosclerosis, these deficiencies affected T-B cell interactions, germinal center B cells, plasma cells, and IgG. As FO B cells differentiating into plasma cells require Blimp-1, we also assessed its role in the development of atherosclerosis. Blimp-1-deficient B cells greatly attenuated atherosclerosis and immunoglobulin-including IgG production, preventing IgG accumulation in atherosclerotic lesions; Blimp-1 deletion also attenuated lesion proinflammatory cytokines, apoptotic cell numbers, and necrotic core. To determine the importance of IgG for atherosclerosis, we purified IgG from atherosclerotic mice. Their transfer but not IgG from nonatherosclerotic mice into Ldlr-/- mice whose B cells are Blimp-1-deficient increased atherosclerosis; transfer was associated with IgG accumulating in atherosclerotic lesions, increased lesion inflammatory cytokines, apoptotic cell numbers, and necrotic core size. CONCLUSIONS: The mechanism by which FO B cells promote atherosclerosis is highly dependent on their expression of MHCII, CD40, and Blimp-1. FO B cell differentiation into IgG-producing plasma cells also is critical for their proatherogenic actions. Targeting B-T cell interactions and pathogenic IgG may provide novel therapeutic strategies to prevent atherosclerosis and its adverse cardiovascular complications.


Subject(s)
Atherosclerosis/immunology , B-Lymphocytes/immunology , Cell Differentiation , Germinal Center/immunology , Immunoglobulin G/immunology , Plasma Cells/immunology , T-Lymphocytes/immunology , Animals , Apoptosis , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , B-Lymphocytes/metabolism , CD40 Antigens/genetics , CD40 Antigens/immunology , CD40 Antigens/metabolism , Disease Models, Animal , Disease Progression , Germinal Center/metabolism , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Immunoglobulin G/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Necrosis , Phenotype , Plaque, Atherosclerotic , Plasma Cells/metabolism , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/immunology , Positive Regulatory Domain I-Binding Factor 1/metabolism , Receptors, LDL/deficiency , Receptors, LDL/genetics , Secretory Pathway , Signal Transduction , T-Lymphocytes/metabolism
10.
J Am Heart Assoc ; 5(11)2016 11 14.
Article in English | MEDLINE | ID: mdl-27930350

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-ß1) expression, including macrophages expressing TGF-ß1, 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) 1ß, and IL-18 were consistent with augmented TGF-ß1 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-ß1 expression accompanied by reduced lesion inflammatory cytokines TNF-α, IL-1ß, and IL-18.


Subject(s)
Atherosclerosis/immunology , B-Lymphocyte Subsets/immunology , Immunoglobulin M/immunology , Myeloid Differentiation Factor 88/immunology , Toll-Like Receptor 4/immunology , Animals , Atherosclerosis/genetics , B-Lymphocytes/immunology , Diet, High-Fat , Interleukin-18/immunology , Interleukin-1beta/immunology , Male , Mice , Mice, Knockout , Mice, Knockout, ApoE , Myeloid Differentiation Factor 88/genetics , Peritoneum/cytology , Peritoneum/immunology , Phagocytosis/immunology , Real-Time Polymerase Chain Reaction , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Transforming Growth Factor beta1/immunology , Tumor Necrosis Factor-alpha/immunology
11.
Sci Rep ; 6: 38161, 2016 12 02.
Article in English | MEDLINE | ID: mdl-27909290

ABSTRACT

A reliable method for the diagnosis of minimal cardiac ischemia would meet a strong demand for the sensitive diagnosis of coronary artery disease in cardiac stress testing and risk stratification in patients with chest pain but unremarkable ECGs and biomarkers. We hypothesized that platelets accumulate early on in ischemic myocardium and a newly developed technology of non-invasive molecular PET imaging of activated platelets can thus detect minimal degrees of myocardial ischemia. To induce different degrees of minimal cardiac ischemia, the left anterior descending artery (LAD) was ligated for 10, 20 or 60 min. Mice were injected with a newly generated scFvanti-GPIIb/IIIa-64CuMeCOSar radiotracer, composed of a single-chain antibody that only binds to activated integrin GPIIb/IIIa (αIIbßIII) and thus to activated platelets, and a sarcophagine cage MeCOSar complexing the long half-life PET tracer copper-64. A single PET/CT scan was performed. Evans Blue/TTC staining to detect necrosis as well as classical serological biomarkers like Troponin I and heart-type fatty acid-binding protein (H-FABP) were negative, whereas PET imaging of activated platelets was able to detect small degrees of ischemia. Taken together, molecular PET imaging of activated platelets represents a unique and highly sensitive method to detect minimal cardiac ischemia.


Subject(s)
Contrast Media/pharmacology , Coronary Angiography/methods , Drug Delivery Systems , Myocardial Ischemia/diagnostic imaging , Platelet Activation , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Positron-Emission Tomography/methods , Single-Chain Antibodies/pharmacology , Animals , Blood Platelets , Humans , Mice
12.
Cardiovasc Res ; 111(4): 385-97, 2016 09.
Article in English | MEDLINE | ID: mdl-27492217

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-1ß 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.


Subject(s)
B-Lymphocytes/metabolism , Inflammation/metabolism , Macrophages/metabolism , Plaque, Atherosclerotic/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Aorta/metabolism , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Cell Death , Diet, High-Fat , Mice, Knockout , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics
13.
Cardiovasc Res ; 109(2): 305-17, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26604037

ABSTRACT

AIMS: Atherosclerosis-related deaths from heart attacks and strokes remain leading causes of global mortality, despite the use of lipid-lowering statins. Thus, there is an urgent need to develop additional therapies. METHODS AND RESULTS: Reports that NKT cells promote atherosclerosis and an NKT cell CD1d-dependent lipid antagonist (DPPE-PEG350, 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine-N[methoxy(polyethyleneglycol)-350]) reduces allergen-induced inflammation led us to investigate its therapeutic potential in preventing the development and progression of experimental atherosclerosis. DPPE-PEG350 was administered to hyperlipidaemic ApoE(-/-) mice with/without established atherosclerosis. Atherosclerosis and immune cells were assessed in the aortic sinus lesions. Lesion expression of monocyte chemoattractant protein-1 (MCP-1) and vascular cell adhesion protein-1 (VCAM-1) responsible for inflammatory immune cell recruitment as well as mRNA expression of IFNγ and its plasma levels were investigated. Necrotic cores and lesion smooth muscle and collagen contents important in plaque stability were determined as were plasma lipid levels. DPPE-PEG350 reduced atherosclerosis development and delayed progression of established atherosclerosis without affecting plasma lipids. CD4 and CD8 T cells and B cells in atherosclerotic lesions were decreased in DPPE-PEG350-treated mice. Lesion MCP-1 and VCAM-1 protein expression and necrotic core size were reduced without affecting lesion smooth muscle and collagen content. IFNγ and lymphocytes were unaffected by the treatment. CONCLUSION: The attenuation of progression of established atherosclerosis together with reduced development of atherosclerosis in hyperlipidaemic mice by the NKT antagonist, without affecting NKT cell or other lymphocyte numbers, suggests that targeting lesion inflammation via CD1d-dependent activation of NKT cells using DPPE-PEG350 has a therapeutic potential in treating atherosclerosis.


Subject(s)
Antigens, CD1d/metabolism , Atherosclerosis/metabolism , Natural Killer T-Cells/cytology , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Atherosclerosis/immunology , Inflammation/immunology , Lymphocyte Activation/immunology , Mice, Knockout , Necrosis/genetics , Necrosis/immunology , Plaque, Atherosclerotic/pathology , Vascular Cell Adhesion Molecule-1/metabolism
14.
Cardiovasc Res ; 106(3): 443-52, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25681396

ABSTRACT

AIMS: To investigate whether activation of atheroprotective peritoneal B1a cells by apoptotic cells or phosphatidylserine liposomes (PSLs) can enhance their protective actions during atherosclerosis development. METHODS AND RESULTS: Male apolipoprotein E-knockout (ApoE-/-) mice were treated with apoptotic cells or PSLs at the beginning of 8-week high-fat diet. Intraperitoneally administered apoptotic cells attenuated atherosclerosis in hypercholesterolemic ApoE-/- mice by 53% and macrophage accumulation by 52%, effects mimicked by administering PSLs and abolished by B1a cell depletion by splenectomy. These effects were associated with reduced lesion CD4+ and CD8+ T cells, mRNAs of MCP-1, VCAM-1, TNF-α, IL-1ß, IL-12, and IL-18 while anti-inflammatory TGF-ß mRNA levels doubled. Apoptotic cells or PSLs increased B1a lymphocytes including TIM-1+ B1a cells in vivo and in vitro while other lymphocyte populations were unaffected. Total plasma IgM, anti-leucocyte, anti-CD3, anti-CD4, and anti-oxLDL IgM were elevated. IgM in atherosclerotic lesions was also elevated and this was associated with reduced lesion MDA-LDL (oxLDL), apoptotic cells and necrotic core size. These effects of activating B1a cells could be attributed to B1a-derived polyreactive IgM deposited in lesions that reduce inflammatory cytokines by lowering lesion ox-LDL via anti-oxLDL IgM, T-cells via anti-leucocyte, anti-CD3, and anti-CD4 IgM, apoptotic cells and necrotic core size via IgM binding to apoptotic cells and enhancing phagocytosis, which also elevates anti-inflammatory cytokines. CONCLUSION: Targeting B1a cell activation by PSLs may be a potentially potent therapeutic strategy to attenuate atherosclerosis and reduce the incidence of atherosclerosis-dependent myocardial infarction and stroke.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Apoptosis , Atherosclerosis/prevention & control , B-Lymphocyte Subsets/drug effects , Immunoglobulin M/biosynthesis , Lymphocyte Activation/drug effects , Phosphatidylserines/administration & dosage , Thymocytes/transplantation , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Cells, Cultured , Diet, High-Fat , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation , Hypercholesterolemia/complications , Hypercholesterolemia/genetics , Hypercholesterolemia/metabolism , Immunoglobulin M/immunology , Inflammation Mediators/metabolism , Lipoproteins, LDL/metabolism , Liposomes , Male , Mice, Knockout , Necrosis , Phagocytosis , Phenotype , Splenectomy , Thymocytes/immunology , Thymocytes/metabolism , Thymocytes/pathology , Time Factors
15.
Circ Res ; 116(2): 245-54, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25398236

ABSTRACT

RATIONALE: CD4(+) natural killer T (NKT) cells augment atherosclerosis in apolipoprotein E-deficient (ApoE)(-/-) mice but their mechanisms of action are unknown. OBJECTIVES: We investigated the roles of bystander T, B, and NK cells; NKT cell-derived interferon-γ, interleukin (IL)-4, and IL-21 cytokines; and NKT cell-derived perforin and granzyme B cytotoxins in promoting CD4(+) NKT cell atherogenicity. METHODS AND RESULTS: Transfer of CD4(+) NKT cells into T- and B-cell-deficient ApoE(-/-)Rag2(-/-) mice augmented aortic root atherosclerosis by ≈75% that was ≈30% of lesions in ApoE(-/-) mice; macrophage accumulation similarly increased. Transferred NKT cells were identified in the liver and atherosclerotic lesions of recipient mice. Transfer of CD4(+) NKT cells into T-, B-cell-deficient, and NK cell-deficient ApoE(-/-)Rag2(-/-)γC(-/-) mice also augmented atherosclerosis. These data indicate that CD4(+) NKT cells can exert proatherogenic effects independent of other lymphocytes. To investigate the role of NKT cell-derived interferon-γ, IL-4, and IL-21 cytokines and perforin and granzyme B cytotoxins, CD4(+) NKT cells from mice deficient in these molecules were transferred into NKT cell-deficient ApoE(-/-)Jα18(-/-) mice. CD4(+) NKT cells deficient in IL-4, interferon-γ, or IL-21 augmented atherosclerosis in ApoE(-/-)Jα18(-/-) mice by ≈95%, ≈80%, and ≈70%, respectively. Transfer of CD4(+) NKT cells deficient in perforin or granzyme B failed to augment atherosclerosis. Apoptotic cells, necrotic cores, and proinflammatory VCAM-1 (vascular cell adhesion molecule) and MCP-1 (monocyte chemotactic protein) were reduced in mice receiving perforin-deficient NKT cells. CD4(+) NKT cells are twice as potent as CD4(+) T cells in promoting atherosclerosis. CONCLUSIONS: CD4(+) NKT cells potently promote atherosclerosis by perforin and granzyme B-dependent apoptosis that increases postapoptotic necrosis and inflammation.


Subject(s)
Atherosclerosis/metabolism , CD4-Positive T-Lymphocytes/metabolism , Granzymes/deficiency , Natural Killer T-Cells/metabolism , Pore Forming Cytotoxic Proteins/deficiency , Sinus of Valsalva/metabolism , Adoptive Transfer/methods , Animals , Atherosclerosis/immunology , Atherosclerosis/pathology , CD4-Positive T-Lymphocytes/immunology , Male , Mice , Mice, Knockout , Natural Killer T-Cells/immunology , Sinus of Valsalva/immunology , Sinus of Valsalva/pathology
16.
PLoS One ; 9(5): e97422, 2014.
Article in English | MEDLINE | ID: mdl-24846287

ABSTRACT

Soluble P-selectin (sP-selectin), a biomarker of inflammatory related pathologies including cardiovascular and peripheral vascular diseases, also has pro-atherosclerotic effects including the ability to increase leukocyte recruitment and modulate thrombotic responses in vivo. The current study explores its role in progressing atherosclerotic plaque disease. Apoe-/- mice placed on a high fat diet (HFD) were given daily injections of recombinant dimeric murine P-selectin (22.5 µg/kg/day) for 8 or 16 weeks. Saline or sE-selectin injections were used as negative controls. In order to assess the role of sP-selectin on atherothrombosis an experimental plaque remodelling murine model, with sm22α-hDTR Apoe-/- mice on a HFD in conjunction with delivery of diphtheria toxin to induce targeted vascular smooth muscle apoptosis, was used. These mice were similarly given daily injections of sP-selectin for 8 or 16 weeks. While plaque mass and aortic lipid content did not change with sP-selectin treatment in Apoe-/- or SM22α-hDTR Apoe-/- mice on HFD, increased plasma MCP-1 and a higher plaque CD45 content in Apoe-/- HFD mice was observed. As well, a significant shift towards a more unstable plaque phenotype in the SM22α-hDTR Apoe-/- HFD mice, with increased macrophage accumulation and lower collagen content, leading to a lower plaque stability index, was observed. These results demonstrate that chronically raised sP-selectin favours progression of an unstable atherosclerotic plaque phenotype.


Subject(s)
Dietary Fats/adverse effects , Macrophages/metabolism , P-Selectin/blood , Plaque, Atherosclerotic/metabolism , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Dietary Fats/pharmacology , Disease Models, Animal , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Macrophages/pathology , Mice , Mice, Knockout , P-Selectin/genetics , Plaque, Atherosclerotic/chemically induced , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/pathology
17.
Clin Sci (Lond) ; 127(7): 485-97, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24724734

ABSTRACT

RAGE (receptor for advanced glycation end-products) is expressed on multiple cell types implicated in the progression of atherosclerosis and plays a role in DAA (diabetes-associated atherosclerosis). The aim of the present study was to determine the relative role of either BM (bone marrow)- or non-BM-derived RAGE in the pathogenesis of STZ (streptozotocin)-induced DAA. Male ApoE (apolipoprotein E)-null (ApoE-/-:RAGE+/+) and ApoE:RAGE-null (ApoE-/-:RAGE-/-) mice at 7 weeks of age were rendered diabetic with STZ. At 8 weeks of age, ApoE-/- and ApoE-/-:RAGE-/- control and diabetic mice received BM from either RAGE-null or RAGE-bearing mice, generating various chimaeras. After 10 and 20 weeks of diabetes, mice were killed and gene expression and atherosclerotic lesion formation were evaluated respectively. Deletion of RAGE in either the BM cells or non-BM cells both resulted in a significant attenuation in DAA, which was associated with reduced VCAM-1 (vascular cell adhesion molecule-1) expression and translated into reduced adhesion in vitro. In conclusion, the results of the present study highlight the importance of both BM- and non-BM-derived RAGE in attenuating the development of DAA.


Subject(s)
Atherosclerosis/genetics , Diabetes Mellitus, Experimental/genetics , Receptors, Immunologic/physiology , Animals , Atherosclerosis/complications , Atherosclerosis/pathology , Bone Marrow/immunology , Bone Marrow/metabolism , Cell Adhesion/genetics , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Gene Deletion , Gene Expression Regulation , Male , Matrix Metalloproteinases/metabolism , Mice , Mice, Transgenic , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Vascular Cell Adhesion Molecule-1/metabolism
18.
J Mol Cell Cardiol ; 69: 32-42, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24508700

ABSTRACT

Myocardial infarction (MI) provokes regional inflammation which facilitates the healing, whereas excessive inflammation leads to adverse cardiac remodelling. Our aim was to determine the role of macrophage migration inhibitory factor (MIF) in inflammation and cardiac remodelling following MI. Wild type (WT) or global MIF deficient (MIFKO) mice were subjected to coronary artery occlusion. Compared to WT mice, MIFKO mice had a significantly lower incidence of post-MI cardiac rupture (27% vs. 53%) and amelioration of cardiac remodelling. These were associated with suppressed myocardial leukocyte infiltration, inflammatory mediators' expression, and reduced activity of MMP-2, MMP-9, p38 and JNK MAPK. Infarct myocardium-derived or exogenous MIF mediated macrophage chemotaxis in vitro that was suppressed by inhibition of p38 MAPK or NF-κB. To further dissect the role of MIF derived from different cellular sources in post-MI cardiac remodelling, we generated chimeric mice with MIF deficiency either in bone marrow derived-cells (WT(KO)) or in somatic-cells (KO(WT)). Compared to WT and KO(WT) mice, WT(KO) mice had reduced rupture risk and ameliorated cardiac remodelling, associated with attenuated regional leukocyte infiltration and expression of inflammatory mediators. In contrast, KO(WT) mice had delayed healing and enhanced expression of M1 macrophage markers, but diminished expression of M2 markers during the early healing phase. In conclusion, global MIF deletion protects the heart from post-infarct cardiac rupture and remodelling through suppression of leukocyte infiltration and inflammation. Leukocyte-derived MIF promotes inflammatory responses after MI, whereas cardiac-derived MIF affects early but not ultimate healing process.


Subject(s)
Heart Rupture, Post-Infarction/immunology , Inflammation/immunology , Intramolecular Oxidoreductases/physiology , Leukocytes/immunology , Macrophage Migration-Inhibitory Factors/physiology , Myocardial Infarction/immunology , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Heart Rupture, Post-Infarction/metabolism , Heart Rupture, Post-Infarction/pathology , Immunoenzyme Techniques , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Leukocytes/metabolism , Leukocytes/pathology , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Phosphorylation , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
19.
Cardiovasc Res ; 102(1): 128-37, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24469537

ABSTRACT

AIM: Although natural killer (NK) cells, a key component of the innate immune system, have been identified in human and mouse atherosclerotic lesions, their role in atherosclerosis development remains unclear. To determine their role in atherosclerosis, we used both loss- and gain-of-function experiments in ApoE(-/-) mice fed a high-fat diet. METHODS AND RESULTS: Treatment of ApoE(-/-) mice with anti-Asialo-GM1 antibodies depleted NK cells without affecting other lymphocytes, including natural killer T cells, and greatly attenuated atherosclerosis. These effects were independent of plasma lipids. To confirm the atherogenicity of NK cells, these cells were isolated from mouse spleens for adoptive transfer into lymphocyte-deficient ApoE(-/-)Rag2(-/-)IL2rg(-/-) mice. Transfer of NK cells from wild-type mice into ApoE(-/-)Rag2(-/-)IL2rg(-/-) mice doubled lesion size, confirming a pro-atherogenic role for NK cells. To determine whether their atherogenicity was dependent on production of interferon-γ (IFN-γ) or cytotoxins, we compared the transfer of NK cells deficient in IFN-γ, perforin, and granzyme B with the transfer of wild-type NK cells. Transfer of IFN-γ-deficient NK cells increased lesion size in the lymphocyte-deficient ApoE(-/-) mice as wild-type NK cells. However, granzyme B- and perforin-deficient NK cells did not affect lesion size. Only wild-type NK cells increased necrotic core size, whereas perforin- and granzyme B-deficient NK cells did not. Plasma lipid levels were largely unaffected by the cell transfer. CONCLUSION: Our loss- and gain-of-function findings provide definitive evidence that NK cells are atherogenic and their production of perforin and granzyme B contributes to atherosclerosis and the expansion of necrotic cores.


Subject(s)
Atherosclerosis/metabolism , Cytotoxicity, Immunologic , Killer Cells, Natural/metabolism , Adoptive Transfer , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Atherosclerosis/pathology , Disease Models, Animal , Granzymes/metabolism , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Male , Mice , Perforin/metabolism
20.
PLoS One ; 8(4): e60430, 2013.
Article in English | MEDLINE | ID: mdl-23560095

ABSTRACT

AIMS: Option to attenuate atherosclerosis by depleting B2 cells is currently limited to anti-CD20 antibodies which deplete all B-cell subtypes. In the present study we evaluated the capacity of a monoclonal antibody to B cell activating factor-receptor (BAFFR) to selectively deplete atherogenic B2 cells to prevent both development and progression of atherosclerosis in the ApoE(-/-) mouse. METHODS AND RESULTS: To determine whether the BAFFR antibody prevents atherosclerosis development, we treated ApoE(-/-) mice with the antibody while feeding them a high fat diet (HFD) for 8 weeks. Mature CD93(-) CD19(+) B2 cells were reduced by treatment, spleen B-cell zones disrupted and spleen CD20 mRNA expression decreased while B1a cells and non-B cells were spared. Atherosclerosis was ameliorated in the hyperlipidemic mice and CD19(+) B cells, CD4(+) and CD8(+) T cells were reduced in atherosclerotic lesions. Expressions of proinflammatory cytokines, IL1ß, TNFα, and IFNγ in the lesions were also reduced, while MCP1, MIF and VCAM-1 expressions were unaffected. Plasma immunoglobulins were reduced, but MDA-oxLDL specific antibodies were unaffected. To determine whether anti-BAFFR antibody ameliorates progression of atherosclerosis, we first fed ApoE(-/-) mice a HFD for 6 weeks, and then instigated anti-BAFFR antibody treatment for a further 6 week-HFD. CD93(-) CD19(+) B2 cells were selectively decreased and atherosclerotic lesions were reduced by this treatment. CONCLUSION: Anti-BAFFR monoclonal antibody selectively depletes mature B2 cells while sparing B1a cells, disrupts spleen B-cell zones and ameliorates atherosclerosis development and progression in hyperlipidemic ApoE(-/-) mice. Our findings have potential for clinical translation to manage atherosclerosis-based cardiovascular diseases.


Subject(s)
Antibodies, Monoclonal/pharmacology , Apolipoproteins E/deficiency , Atherosclerosis/drug therapy , B-Lymphocyte Subsets/drug effects , Hyperlipidemias/drug therapy , Spleen/drug effects , Animals , Antibodies, Monoclonal/immunology , Antigens, CD/genetics , Antigens, CD/immunology , Apolipoproteins E/genetics , Apolipoproteins E/immunology , Atherosclerosis/complications , Atherosclerosis/immunology , Atherosclerosis/pathology , B-Cell Activation Factor Receptor/antagonists & inhibitors , B-Cell Activation Factor Receptor/genetics , B-Cell Activation Factor Receptor/immunology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , Cytokines/biosynthesis , Cytokines/immunology , Diet, High-Fat , Disease Progression , Hyperlipidemias/complications , Hyperlipidemias/immunology , Hyperlipidemias/pathology , Lymphocyte Depletion , Male , Mice , Mice, Knockout , Spleen/immunology , Spleen/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...