Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Arterioscler Thromb Vasc Biol ; 44(6): 1346-1364, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38660806

ABSTRACT

BACKGROUND: Atherosclerosis is the major underlying pathology of cardiovascular disease and is driven by dyslipidemia and inflammation. Inhibition of the immunoproteasome, a proteasome variant that is predominantly expressed by immune cells and plays an important role in antigen presentation, has been shown to have immunosuppressive effects. METHODS: We assessed the effect of ONX-0914, an inhibitor of the immunoproteasomal catalytic subunits LMP7 (proteasome subunit ß5i/large multifunctional peptidase 7) and LMP2 (proteasome subunit ß1i/large multifunctional peptidase 2), on atherosclerosis and metabolism in LDLr-/- and APOE*3-Leiden.CETP mice. RESULTS: ONX-0914 treatment significantly reduced atherosclerosis, reduced dendritic cell and macrophage levels and their activation, as well as the levels of antigen-experienced T cells during early plaque formation, and Th1 cells in advanced atherosclerosis in young and aged mice in various immune compartments. Additionally, ONX-0914 treatment led to a strong reduction in white adipose tissue mass and adipocyte progenitors, which coincided with neutrophil and macrophage accumulation in white adipose tissue. ONX-0914 reduced intestinal triglyceride uptake and gastric emptying, likely contributing to the reduction in white adipose tissue mass, as ONX-0914 did not increase energy expenditure or reduce total food intake. Concomitant with the reduction in white adipose tissue mass upon ONX-0914 treatment, we observed improvements in markers of metabolic syndrome, including lowered plasma triglyceride levels, insulin levels, and fasting blood glucose. CONCLUSIONS: We propose that immunoproteasomal inhibition reduces 3 major causes underlying cardiovascular disease, dyslipidemia, metabolic syndrome, and inflammation and is a new target in drug development for atherosclerosis treatment.


Subject(s)
Adipose Tissue, White , Atherosclerosis , Disease Models, Animal , Metabolic Syndrome , Mice, Inbred C57BL , Proteasome Endopeptidase Complex , Receptors, LDL , Animals , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Atherosclerosis/drug therapy , Atherosclerosis/immunology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Metabolic Syndrome/drug therapy , Metabolic Syndrome/immunology , Adipose Tissue, White/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/pathology , Receptors, LDL/genetics , Receptors, LDL/deficiency , Proteasome Endopeptidase Complex/metabolism , Male , Proteasome Inhibitors/pharmacology , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Aortic Diseases/prevention & control , Aortic Diseases/pathology , Aortic Diseases/genetics , Aortic Diseases/enzymology , Aortic Diseases/immunology , Aortic Diseases/metabolism , Macrophages/drug effects , Macrophages/metabolism , Macrophages/immunology , Plaque, Atherosclerotic , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Mice, Knockout, ApoE , Mice , Energy Metabolism/drug effects , Oligopeptides
2.
Atherosclerosis ; 352: 1-9, 2022 07.
Article in English | MEDLINE | ID: mdl-35636144

ABSTRACT

BACKGROUND AND AIMS: Increasing evidence has shown that immune checkpoint molecules of the T-cell immunoglobulin and mucin domain (Tim) family are associated with diverse physiologic and pathologic processes. Previous studies of the role of Tim-1 in atherosclerosis using anti-Tim-1 antibodies have yielded contradictory results. We thus aimed to investigate atherosclerosis development in Tim-1 deficient mice. METHODS: Mice with a specific loss of the Tim-1 mucin-domain (Tim-1Δmucin) and C57BL/6 (WT) mice received a single injection of a recombinant adeno-associated virus encoding murine Pcsk9 (rAAV2/8-D377Y-mPcsk9) and were fed a Western type diet for 13 weeks to introduce atherosclerosis. RESULTS: Tim-1Δmucin mice developed significantly larger lesions in the aortic root compared to WT mice, with significantly more macrophages and a trend towards a larger necrotic core. Furthermore, Tim-1Δmucin mice showed a significant loss of IL-10+ B cells and regulatory B cell subsets and increased pro-atherogenic splenic follicular B cells compared to WT mice. Moreover, Tim-1Δmucin mice displayed a dramatic reduction in Th2-associated immune response compared to controls but we did not observe any changes in humoral immunity. CONCLUSIONS: In summary, Tim-1Δmucin mice displayed a profound impairment in IL-10+ B cells and an imbalance in the Th1/Th2 ratio, which associated with exacerbated atherosclerosis.


Subject(s)
Atherosclerosis , Hepatitis A Virus Cellular Receptor 1/metabolism , Proprotein Convertase 9 , Animals , Atherosclerosis/pathology , Interleukin-10/genetics , Mice , Mice, Inbred C57BL , Mucins
3.
Front Cardiovasc Med ; 9: 781436, 2022.
Article in English | MEDLINE | ID: mdl-35187121

ABSTRACT

B and T cells are interconnected in the T follicular helper-germinal center B cell (TFH-GC B cell) axis, which is hyperactive during atherosclerosis development and loss of control along this axis results in exacerbated atherosclerosis. Inhibition of the TFH-GC B cell axis can be achieved by providing negative co-stimulation to TFH cells through the PD-1/PD-L1 pathway. Therefore, we investigated a novel therapeutic strategy using PD-L1-expressing B cells to inhibit atherosclerosis. We found that IFNγ-stimulated B cells significantly enhanced PD-L1 expression and limited TFH cell development. To determine whether IFNγ-B cells can reduce collar-induced atherosclerosis, apoE -/- mice fed a Western-type diet were treated with PBS, B cells or IFNγ-B cells for a total of 5 weeks following collar placement. IFNγ-B cells significantly increased PD-L1hi GC B cells and reduced plasmablasts. Interestingly, IFNγ-B cells-treated mice show increased atheroprotective Tregs and T cell-derived IL-10. In line with these findings, we observed a significant reduction in total lesion volume in carotid arteries of IFNγ-B cells-treated mice compared to PBS-treated mice and a similar trend was observed compared to B cell-treated mice. In conclusion, our data show that IFNγ-stimulated B cells strongly upregulate PD-L1, inhibit TFH cell responses and protect against atherosclerosis.

4.
Cardiovasc Res ; 117(5): 1309-1324, 2021 04 23.
Article in English | MEDLINE | ID: mdl-32653923

ABSTRACT

AIMS: A hallmark of advanced atherosclerosis is inadequate immunosuppression by regulatory T (Treg) cells inside atherosclerotic lesions. Dyslipidemia has been suggested to alter Treg cell migration by affecting the expression of specific membrane proteins, thereby decreasing Treg cell migration towards atherosclerotic lesions. Besides membrane proteins, cellular metabolism has been shown to be a crucial factor in Treg cell migration. We aimed to determine whether dyslipidemia contributes to altered migration of Treg cells, in part, by affecting cellular metabolism. METHODS AND RESULTS: Dyslipidemia was induced by feeding Ldlr-/- mice a western-type diet for 16-20 weeks and intrinsic changes in Treg cells affecting their migration and metabolism were examined. Dyslipidemia was associated with altered mTORC2 signalling in Treg cells, decreased expression of membrane proteins involved in migration, including CD62L, CCR7, and S1Pr1, and decreased Treg cell migration towards lymph nodes. Furthermore, we discovered that diet-induced dyslipidemia inhibited mTORC1 signalling, induced PPARδ activation and increased fatty acid (FA) oxidation in Treg cells. Moreover, mass-spectrometry analysis of serum from Ldlr-/- mice with normolipidemia or dyslipidemia showed increases in multiple PPARδ ligands during dyslipidemia. Treatment with a synthetic PPARδ agonist increased the migratory capacity of Treg cells in vitro and in vivo in an FA oxidation-dependent manner. Furthermore, diet-induced dyslipidemia actually enhanced Treg cell migration into the inflamed peritoneum and into atherosclerotic lesions in vitro. CONCLUSION: Altogether, our findings implicate that dyslipidemia does not contribute to atherosclerosis by impairing Treg cell migration as dyslipidemia associated with an effector-like migratory phenotype in Treg cells.


Subject(s)
Atherosclerosis/metabolism , Cell Movement , Diet, High-Fat , Dyslipidemias/metabolism , Energy Metabolism , Inflammation/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Cell Movement/drug effects , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Dyslipidemias/genetics , Dyslipidemias/immunology , Energy Metabolism/drug effects , Fatty Acids/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Inflammation Mediators/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice, Knockout, ApoE , Oxidation-Reduction , PPAR gamma/agonists , PPAR gamma/metabolism , Phenotype , Plaque, Atherosclerotic , Receptors, LDL/genetics , Receptors, LDL/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Thiazoles/pharmacology
5.
RSC Chem Biol ; 1(4): 263-272, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-34458765

ABSTRACT

Fluorescent cell surface receptor agonists allow visualization of processes that are set in motion by receptor activation. This study describes the synthesis of two fluorescent, low molecular weight ligands for the follicle-stimulating hormone receptor (FSHR), based on a dihydropyridine (DHP) agonist. We show that both BODIPY- and Cy5-conjugated DHP (m-DHP-BDP and m-DHP-Cy5) are potent FSHR agonists, able to activate receptor signalling with nanomolar potencies and to effect receptor internalisation at higher concentrations. FSHR-dependent uptake of m-DHP-Cy5 is in stark contrast to the cellular uptake of m-DHP-BDP which was efficiently internalised also in the absence of FSHR. Our results comprise a first-in-class fluorescent low molecular weight ligand for in situ FSHR imaging and pertain the potential means for targeted delivery of drugs into the endolysosomal pathway of FSHR-expressing cells.

6.
Cardiovasc Res ; 116(2): 295-305, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31150053

ABSTRACT

AIMS: The immune system is strongly involved in atherosclerosis and immune regulation generally leads to attenuated atherosclerosis. B- and T-lymphocyte attenuator (BTLA) is a novel co-receptor that negatively regulates the activation of B and T cells; however, there have been no reports of BTLA and its function in atherosclerosis or cardiovascular disease (CVD). We aimed to assess the dominant BTLA expressing leucocyte in CVD patients and to investigate whether BTLA has a functional role in experimental atherosclerosis. METHODS AND RESULTS: We show that BTLA is primarily expressed on B cells in CVD patients and follicular B2 cells in low-density lipoprotein receptor-deficient (Ldlr-/-) mice. We treated Ldlr-/- mice that were fed a western-type diet (WTD) with phosphate-buffered saline, an isotype antibody, or an agonistic BTLA antibody (3C10) for 6 weeks. We report here that the agonistic BTLA antibody significantly attenuated atherosclerosis. This was associated with a strong reduction in follicular B2 cells, while regulatory B and T cells were increased. The BTLA antibody showed similar immunomodulating effects in a progression study in which Ldlr-/- mice were fed a WTD for 10 weeks before receiving antibody treatment. Most importantly, BTLA stimulation enhanced collagen content, a feature of stable lesions, in pre-existing lesions. CONCLUSION: Stimulation of the BTLA pathway in Ldlr-/- mice reduces initial lesion development and increases collagen content of established lesions, presumably by shifting the balance between atherogenic follicular B cells and atheroprotective B cells and directing CD4+ T cells towards regulatory T cells. We provide the first evidence that BTLA is a very promising target for the treatment of atherosclerosis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , B-Lymphocytes/drug effects , Lymphocyte Activation/drug effects , Plaque, Atherosclerotic , Receptors, Immunologic/agonists , Animals , Aorta/immunology , Aorta/metabolism , Aorta/pathology , Aortic Diseases/immunology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes, Regulatory/drug effects , B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/metabolism , Cells, Cultured , Collagen/metabolism , Diet, High-Fat , Disease Models, Animal , Disease Progression , Humans , Male , Mice, Knockout , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Receptors, LDL/deficiency , Receptors, LDL/genetics , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
7.
Sci Rep ; 9(1): 17391, 2019 11 22.
Article in English | MEDLINE | ID: mdl-31757993

ABSTRACT

Cardiovascular diseases form the most common cause of death worldwide, with atherosclerosis as main etiology. Atherosclerosis is marked by cholesterol rich lipoprotein deposition in the artery wall, evoking a pathogenic immune response. Characteristic for the disease is the pathogenic accumulation of macrophages in the atherosclerotic lesion, which become foam cells after ingestion of large quantities of lipoproteins. We hypothesized that, by inducing a CD8 T cell response towards lipoprotein derived apolipoprotein-B100 (ApoB100), lesional macrophages, that are likely to cross-present lipoprotein constituents, can specifically be eliminated. Based on in silico models for protein processing and MHC-I binding, 6 putative CD8 T cell epitopes derived from ApoB100 were synthesized. HLA-A2 binding was confirmed for all peptides by T2 cell binding assays and recall responses after vaccination with the peptides proved that 5 of 6 peptides could induce CD8 T cell responses. Induction of ApoB100 specific CD8 T cells did not impact plaque size and cellular composition in HLA-A2 and human ApoB100 transgenic LDLr-/- mice. No recall response could be detected in cultures of cells isolated from the aortic arch, which were observed in cell cultures of splenocytes and mesenteric lymph nodes, suggesting that the atherosclerotic environment impairs CD8 T cell activation.


Subject(s)
Apolipoprotein B-100/immunology , Atherosclerosis/immunology , CD8-Positive T-Lymphocytes/immunology , HLA-A2 Antigen/immunology , Animals , Antibodies, Monoclonal, Humanized/metabolism , Apolipoprotein B-100/chemistry , Atherosclerosis/genetics , Atherosclerosis/pathology , Cells, Cultured , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/genetics , HLA-A2 Antigen/metabolism , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptides/immunology , Receptors, LDL/genetics , Receptors, LDL/metabolism
8.
Atherosclerosis ; 280: 118-125, 2019 01.
Article in English | MEDLINE | ID: mdl-30500604

ABSTRACT

BACKGROUND AND AIMS: Limiting the pro-inflammatory immune response is critical for the treatment of atherosclerosis. Regulatory B cells (Bregs) can modulate the immune response through interleukin-10 (IL-10). Current data regarding Bregs and atherosclerosis is scarce and conflicting. METHODS: In this study, we investigated the frequency of IL-10+ B cells during the development of atherosclerosis in low-density lipoprotein receptor-deficient (Ldlr-/-) mice and studied the effect of adoptive transfer of IL-10+ B cells on atherosclerosis. RESULTS: We found a very strong inverse correlation between atherosclerosis severity and the frequency of IL-10+ B cells. This effect was cholesterol-independent and observed in spleen, draining lymph nodes and peritoneal cavity. To directly assess the effects of IL-10+ B cells on atherosclerosis, we expanded IL-10+ B cells ex vivo with anti-CD40 and selected pure and viable IL-10-secreting B cells and IL-10- B cells and adoptively transferred them to Ldlr-/- mice, respectively. While IL-10- B cells were strongly atherogenic compared to control-treated mice, IL-10+ B cells did not affect lesion size. Adoptive transfer of IL-10+ B cells strongly reduced circulating leukocyte numbers and inflammatory monocytes. In addition, they decreased CD4+ T cell activation and increased IL-10+ CD4+ T cell numbers. Interestingly, both IL-10+ and IL-10- B cells exacerbated serum cholesterol levels and resulted in fatty livers, which potentially masked the beneficial effects of IL-10+ B cells on atherosclerosis. CONCLUSIONS: These findings underscore the strong immune-regulating function of IL-10+ B cells and provide additional incentives to explore effective strategies that expand IL-10+ B cells in atherosclerosis.


Subject(s)
Atherosclerosis/immunology , B-Lymphocytes, Regulatory/immunology , Interleukin-10/metabolism , Receptors, LDL/genetics , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/cytology , Cholesterol/blood , Immune System , Inflammation , Leukocytes/cytology , Liver/metabolism , Male , Mice , Mice, Knockout , Monocytes/cytology
9.
Atherosclerosis ; 280: 132-139, 2019 01.
Article in English | MEDLINE | ID: mdl-30513408

ABSTRACT

BACKGROUND AND AIMS: The development of atherosclerosis is tightly regulated by the innate and adaptive immune system. Communication between these two compartments occurs, among others, upon presentation of lipid antigens to the NKT cell population by CD1d-expressing antigen-presenting cells. Recent evidence states that also mast cells express CD1d and can directly communicate with NKT cells. However, no such relationship has been reported in atherosclerosis. Here, we aimed to elucidate in vivo the CD1d-mediated interaction between mast cells and NKT cells upon atherosclerosis progression. METHODS: We adoptively transferred CD1d-/- or control mast cells to mast cell-deficient apoE-/-KitW-sh/W-sh mice and subsequently placed the animals on a Western-type diet for 10 weeks. RESULTS: At the end of the Western-type diet period, the aortic root of CD1d-/- mast cell-reconstituted mice displayed increased plaque size, with less collagen deposition and higher intraplaque CD4+ T cells, as compared to control mice. In addition, T cells inside the aortic arch showed higher pro-inflammatory cytokine production in the form of IFNγ, TNFα and IL-17. Finally, T-bet expression was found elevated in both CD4+ and CD8+ circulating T cells. CONCLUSIONS: This study is the first to illustrate that disruption of the CD1d communication pathway between mast cells and NKT cells aggravates atherosclerosis, through a shift towards pro-inflammatory T cell responses. This ability of mast cell action during plaque progression sheds new light on their role in atherosclerosis.


Subject(s)
Antigens, CD1d/metabolism , Atherosclerosis/immunology , CD4-Positive T-Lymphocytes/cytology , Killer Cells, Natural/cytology , Mast Cells/cytology , Natural Killer T-Cells/cytology , Plaque, Atherosclerotic/immunology , Animals , Aorta, Thoracic/metabolism , Bone Marrow Cells/cytology , CD8-Positive T-Lymphocytes/cytology , Female , Inflammation , Interferon-gamma/immunology , Interleukin-17/immunology , Lymphocyte Activation , Mast Cells/metabolism , Mice , Tumor Necrosis Factor-alpha/immunology
10.
PLoS One ; 13(1): e0190962, 2018.
Article in English | MEDLINE | ID: mdl-29346401

ABSTRACT

An abdominal aortic aneurysm (AAA) is a dilatation of the abdominal aorta leading to serious complications and mostly to death. AAA development is associated with an accumulation of inflammatory cells in the aorta including NKT cells. An important factor in promoting the recruitment of these inflammatory cells into tissues and thereby contributing to the development of AAA is angiotensin II (Ang II). We demonstrate that a deficiency in CD1d dependent NKT cells under hyperlipidemic conditions (LDLr-/-CD1d-/- mice) results in a strong decline in the severity of angiotensin II induced aneurysm formation when compared with LDLr-/- mice. In addition, we show that Ang II amplifies the activation of NKT cells both in vivo and in vitro. We also provide evidence that type I NKT cells contribute to AAA development by inducing the expression of matrix degrading enzymes in vSMCs and macrophages, and by cytokine dependently decreasing vSMC viability. Altogether, these data prove that CD1d-dependent NKT cells contribute to AAA development in the Ang II-mediated aneurysm model by enhancing aortic degradation, establishing that therapeutic applications which target NKT cells can be a successful way to prevent AAA development.


Subject(s)
Antigens, CD1d/genetics , Aortic Aneurysm, Abdominal/prevention & control , Receptors, LDL/genetics , Angiotensin II/administration & dosage , Animals , Apoptosis/immunology , Flow Cytometry , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/pathology , NIH 3T3 Cells , Natural Killer T-Cells/immunology , Real-Time Polymerase Chain Reaction
11.
Front Immunol ; 9: 2937, 2018.
Article in English | MEDLINE | ID: mdl-30619297

ABSTRACT

Macroautophagy (or autophagy) is a conserved cellular process in which cytoplasmic cargo is targeted for lysosomal degradation. Autophagy is crucial for the functional integrity of different subsets of T cells in various developmental stages. Since atherosclerosis is an inflammatory disease of the vessel wall which is partly characterized by T cell mediated autoimmunity, we investigated how advanced atherosclerotic lesions develop in mice with T cells that lack autophagy-related protein 7 (Atg7), a protein required for functional autophagy. Mice with a T cell-specific knock-out of Atg7 (Lck-Cre Atg7f/f) had a diminished naïve CD4+ and CD8+ T cell compartment in the spleen and mediastinal lymph node as compared to littermate controls (Atg7f/f). Lck-Cre Atg7f/f and Atg7f/f mice were injected intravenously with rAAV2/8-D377Y-mPCSK9 and fed a Western-type diet to induce atherosclerosis. While Lck-Cre Atg7f/f mice had equal serum Proprotein Convertase Subtilisin/Kexin type 9 levels as compared to Atg7f/f mice, serum cholesterol levels were significantly diminished in Lck-Cre Atg7f/f mice. Histological analysis of the liver revealed less steatosis, and liver gene expression profiling showed decreased expression of genes associated with hepatic steatosis in Lck-Cre Atg7f/f mice as compared to Atg7f/f mice. The level of hepatic CD4+ and CD8+ T cells was greatly diminished but both CD4+ and CD8+ T cells showed a relative increase in their IFNγ and IL-17 production upon Atg7 deficiency. Atg7 deficiency furthermore reduced the hepatic NKT cell population which was decreased to < 0.1% of the lymphocyte population. Interestingly, T cell-specific knock-out of Atg7 decreased the mean atherosclerotic lesion size in the tri-valve area by over 50%. Taken together, T cell-specific deficiency of Atg7 resulted in a decrease in hepatic steatosis and limited inflammatory potency in the (naïve) T cell compartment in peripheral lymphoid tissues, which was associated with a strong reduction in experimental atherosclerosis.


Subject(s)
Atherosclerosis/immunology , Autophagy/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Diet, Western/adverse effects , Fatty Liver/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Atherosclerosis/etiology , Atherosclerosis/metabolism , Autophagy/genetics , Autophagy-Related Protein 7/deficiency , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cytokines/immunology , Cytokines/metabolism , Fatty Liver/etiology , Fatty Liver/metabolism , Female , Liver/immunology , Liver/metabolism , Liver/pathology , Mice, Knockout , Mice, Transgenic
12.
Eur J Pharmacol ; 816: 47-57, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-28363745

ABSTRACT

Despite life-style advice and the prescription of cholesterol-lowering and anti-thrombotic drugs, cardiovascular diseases are still the leading cause of death worldwide. Therefore, there is an urgent need for new therapeutic strategies focussing on atherosclerosis, the major underlying pathology of cardiovascular diseases characterized by an accumulation of lipids in an inflamed arterial/vessel wall. CD1d-restricted lipid-sensing natural killer T (NKT) cells, bridging the innate and adaptive immunity, and CD1d-expressing antigen-presenting cells are detected in atherosclerotic lesions of mice and humans. In this review we will summarize studies that point to a critical role for NKT cells in the pathogenesis of atherosclerosis and other cardiovascular diseases by the secretion of pro-atherogenic cytokines and cytotoxins. These pro-atherogenic NKT cells are potential targets for new therapeutic strategies in the prevention and treatment of cardiovascular diseases. Additionally, proteins transferring lipids during atherosclerosis, which are also important in the loading of lipids onto CD1d and possible endogenous ligands responsible for the activation of NKT cells during atherosclerosis will be discussed.


Subject(s)
Cardiovascular Diseases/immunology , Natural Killer T-Cells , Animals , Atherosclerosis/immunology , Humans , Lipoproteins/metabolism , Lymphocyte Activation , Mice , Natural Killer T-Cells/cytology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism
13.
Sci Rep ; 7: 42688, 2017 02 16.
Article in English | MEDLINE | ID: mdl-28202909

ABSTRACT

Toll-like receptors (TLR) provide a critical link between innate and adaptive immunity, both important players in atherosclerosis. Since evidence for the role of TLR5 is lacking, we aimed to establish this in the immune axis of atherosclerosis. We assessed the effect of the TLR5-specific ligand Flagellin on macrophage maturation and T-cell polarisation. Next, we generated TLR5-/-LDLr-/- chimeras to study the effect of hematopoietic TLR5 deficiency on atherosclerosis formation. Flagellin stimulation did not influence wildtype or TLR5-/- macrophage maturation. Only in wildtype macrophages, Flagellin exposure increased MCP-1 and IL6 expression. Flagellin alone reduced T-helper 1 proliferation, which was completely overruled in the presence of T-cell receptor activation. In vivo, hematopoietic TLR5 deficiency attenuated atherosclerotic lesion formation by ≈25% (1030*103 ± 63*103 vs. 792*103 ± 61*103 µm2; p = 0.013) and decreased macrophage area (81.3 ± 12.0 vs. 44.2 ± 6.6 µm2; p = 0.011). In TLR5-/- chimeric mice, we observed lower IL6 plasma levels (36.4 ± 5.6 vs. 15.1 ± 2.2 pg/mL; p = 0.003), lower (activated) splenic CD4+ T-cell content (32.3 ± 2.1 vs. 21.0 ± 1.2%; p = 0.0018), accompanied by impaired T-cell proliferative responses. In conclusion, hematopoietic TLR5 deficiency inhibits atherosclerotic lesion formation by attenuated macrophage accumulation and defective T-cell responsiveness.


Subject(s)
Atherosclerosis/etiology , Atherosclerosis/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Macrophages/immunology , Macrophages/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Toll-Like Receptor 5/deficiency , Animals , Atherosclerosis/blood , Atherosclerosis/pathology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Movement/genetics , Cell Movement/immunology , Cytokines/metabolism , Disease Models, Animal , Inflammation Mediators/metabolism , Leukocyte Count , Mice , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , T-Lymphocyte Subsets/cytology
14.
Sci Rep ; 6: 37585, 2016 11 24.
Article in English | MEDLINE | ID: mdl-27883026

ABSTRACT

Lysophosphatidic acid (LPA) is a natural lysophospholipid present at high concentrations within lipid-rich atherosclerotic plaques. Upon local accumulation in the damaged vessels, LPA can act as a potent activator for various types of immune cells through its specific membrane receptors LPA1/3. LPA elicits chemotactic, pro-inflammatory and apoptotic effects that lead to atherosclerotic plaque progression. In this study we aimed to inhibit LPA signaling by means of LPA1/3 antagonism using the small molecule Ki16425. We show that LPA1/3 inhibition significantly impaired atherosclerosis progression. Treatment with Ki16425 also resulted in reduced CCL2 production and secretion, which led to less monocyte and neutrophil infiltration. Furthermore, we provide evidence that LPA1/3 blockade enhanced the percentage of non-inflammatory, Ly6Clow monocytes and CD4+ CD25+ FoxP3+ T-regulatory cells. Finally, we demonstrate that LPA1/3 antagonism mildly reduced plasma LDL cholesterol levels. Therefore, pharmacological inhibition of LPA1/3 receptors may prove a promising approach to diminish atherosclerosis development.


Subject(s)
Atherosclerosis/drug therapy , Receptors, Lysophosphatidic Acid/genetics , Animals , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/pathology , Cholesterol, LDL/blood , Disease Models, Animal , Endocytosis/genetics , Humans , Isoxazoles/administration & dosage , Lysophospholipids/genetics , Mice , Propionates/administration & dosage , Receptors, LDL/deficiency , Receptors, LDL/genetics , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Signal Transduction/genetics
15.
Cardiovasc Res ; 111(3): 252-61, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27234908

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.


Subject(s)
Adoptive Transfer , Antigens, Ly/metabolism , Atherosclerosis/prevention & control , CD11b Antigen/metabolism , Myeloid-Derived Suppressor Cells/transplantation , Receptors, LDL/deficiency , Animals , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Cell Proliferation , Cells, Cultured , Coculture Techniques , Diet, Western , Disease Models, Animal , Female , Genetic Predisposition to Disease , Interferon-gamma/metabolism , Lymphocyte Activation , Male , Mice, Knockout , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Nitric Oxide/metabolism , Phenotype , Receptors, LDL/genetics , Spleen/immunology , Spleen/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism
16.
J Immunol ; 194(5): 2208-18, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25653425

ABSTRACT

Modulation of immune responses may form a powerful approach to treat atherosclerosis. It was shown that clearance of apoptotic cells results in tolerance induction to cleared Ags by dendritic cells (DCs); however, this seems impaired in atherosclerosis because Ag-specific tolerance is lacking. This could result, in part, from decreased emigration of DCs from atherosclerotic lesions because of the high-cholesterol environment. Nonetheless, local induction of anti-inflammatory responses by apoptotic cell clearance seems to dampen atherosclerosis, because inhibition of apoptotic cell clearance worsens atherosclerosis. In this study, we assessed whether i.v. administration of oxLDL-induced apoptotic DCs (apop(ox)-DCs) and, as a control, unpulsed apoptotic DCs could modulate atherosclerosis by inducing tolerance. Adoptive transfer of apop(ox)-DCs into low-density lipoprotein receptor knockout mice either before or during feeding of a Western-type diet resulted in increased numbers of CD103(+) tolerogenic splenic DCs, with a concomitant increase in regulatory T cells. Interestingly, both types of apoptotic DCs induced an immediate 40% decrease in Ly-6C(hi) monocyte numbers and a 50% decrease in circulating CCL2 levels, but only apop(ox)-DC treatment resulted in long-term effects on monocytes and CCL2 levels. Although initial lesion development was reduced by 40% in both treatment groups, only apop(ox)-DC treatment prevented lesion progression by 28%. Moreover, progressed lesions of apop(ox)-DC-treated mice showed a robust 45% increase in collagen content, indicating an enhanced stability of lesions. Our findings clearly show that apoptotic DC treatment significantly decreases lesion development, but only apop(ox)-DCs can positively modulate lesion progression and stability. These findings may translate into a safe treatment for patients with established cardiovascular diseases using patient-derived apop(ox)-DCs.


Subject(s)
Atherosclerosis/therapy , Dendritic Cells/immunology , Immunotherapy, Adoptive , Lipoproteins, LDL/pharmacology , Plaque, Atherosclerotic/therapy , Adaptive Immunity , Animals , Antigens, Ly/genetics , Antigens, Ly/immunology , Apoptosis , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Collagen/genetics , Collagen/immunology , Dendritic Cells/drug effects , Dendritic Cells/pathology , Dendritic Cells/transplantation , Gene Expression Regulation , Immune Tolerance , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/pathology , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/pathology , Primary Cell Culture , Receptors, LDL , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
17.
Angew Chem Int Ed Engl ; 53(41): 10975-8, 2014 Oct 06.
Article in English | MEDLINE | ID: mdl-25163608

ABSTRACT

The ubiquitously expressed mannose-6-phosphate receptors (MPRs) are a promising class of receptors for targeted compound delivery into the endolysosomal compartments of a variety of cell types. The development of a synthetic, multivalent, mannose-6-phosphate (M6P) glycopeptide-based MPR ligand is described. The conjugation of this ligand to fluorescent DCG-04, an activity-based probe for cysteine cathepsins, enabled fluorescent readout of its receptor-targeting properties. The resulting M6P-cluster-BODIPY-DCG-04 probe was shown to efficiently label cathepsins in cell lysates as well as in live cells. Furthermore, the introduction of the 6-O-phosphates leads to a completely altered uptake profile in COS and dendritic cells compared to a mannose-containing ligand. Competition with mannose-6-phosphate abolished all uptake of the probe in COS cells, and we conclude that the mannose-6-phosphate cluster targets the MPR and ensures the targeted delivery of cargo bound to the cluster into the endolysosomal pathway.


Subject(s)
Cathepsins/metabolism , Endosomes/metabolism , Receptor, IGF Type 2/chemistry , Animals , Boron Compounds/chemistry , COS Cells , Cathepsins/chemistry , Chlorocebus aethiops , Dendritic Cells/cytology , Dendritic Cells/metabolism , Fluorescent Dyes/chemistry , Glycopeptides/chemical synthesis , Glycopeptides/chemistry , Leucine/analogs & derivatives , Leucine/chemistry , Ligands , Mannosephosphates/chemistry , Mice , Protein Binding
18.
J Mol Cell Cardiol ; 74: 44-52, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24816217

ABSTRACT

AIMS: The SDF-1α/CXCR4 dyad was previously shown by us and others to be instrumental in intimal hyperplasia as well as early stage atherosclerosis. We here sought to investigate its impact on clinically relevant stages of atherosclerosis in mouse and man. METHODS AND RESULTS: Immunohistochemical analysis of CXCR4 expression in human atherosclerotic lesions revealed a progressive accumulation of CXCR4(+) cells during plaque progression. To address causal involvement of CXCR4 in advanced stages of atherosclerosis we reconstituted LDLr(-/-) mice with autologous bone marrow infected with lentivirus encoding SDF-1α antagonist or CXCR4 degrakine, which effects proteasomal degradation of CXCR4. Functional CXCR4 blockade led to progressive plaque expansion with disease progression, while also promoting intraplaque haemorrhage. Moreover, CXCR4 knockdown was seen to augment endothelial adhesion of neutrophils. Concordant with this finding, inhibition of CXCR4 function increased adhesive capacity and reduced apoptosis of neutrophils and resulted in hyperactivation of circulating neutrophils. Compatible with a role of the neutrophil CXCR4 in end-stage atherosclerosis, CXCR4 expression by circulating neutrophils was lowered in patients with acute cardiovascular syndromes. CONCLUSION: In conclusion, CXCR4 contributes to later stages of plaque progression by perturbing neutrophil function.


Subject(s)
Atherosclerosis/genetics , Hemorrhage/genetics , Neutrophils/metabolism , Plaque, Atherosclerotic/genetics , Receptors, CXCR4/genetics , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Cell Adhesion , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Disease Progression , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Gene Expression Regulation , Genetic Vectors , Hemorrhage/metabolism , Hemorrhage/pathology , Humans , Lentivirus/genetics , Lentivirus/metabolism , Mice , Mice, Knockout , Neutrophils/pathology , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Proteasome Endopeptidase Complex/metabolism , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Receptors, LDL/deficiency , Receptors, LDL/genetics , Signal Transduction
19.
PLoS One ; 8(12): e83134, 2013.
Article in English | MEDLINE | ID: mdl-24376654

ABSTRACT

OBJECTIVE: Co-stimulatory and co-inhibitory molecules are mainly expressed on T cells and antigen presenting cells and strongly orchestrate adaptive immune responses. Whereas co-stimulatory molecules enhance immune responses, signaling via co-inhibitory molecules dampens the immune system, thereby showing great therapeutic potential to prevent cardiovascular diseases. Signaling via co-inhibitory T cell immunoglobulin and ITIM domain (TIGIT) directly inhibits T cell activation and proliferation, and therefore represents a novel therapeutic candidate to specifically dampen pro-atherogenic T cell reactivity. In the present study, we used an agonistic anti-TIGIT antibody to determine the effect of excessive TIGIT-signaling on atherosclerosis. METHODS AND RESULTS: TIGIT was upregulated on CD4(+) T cells isolated from mice fed a Western-type diet in comparison with mice fed a chow diet. Agonistic anti-TIGIT suppressed T cell activation and proliferation both in vitro and in vivo. However, agonistic anti-TIGIT treatment of LDLr(-/-) mice fed a Western-type diet for 4 or 8 weeks did not affect atherosclerotic lesion development in comparison with PBS and Armenian Hamster IgG treatment. Furthermore, elevated percentages of dendritic cells were observed in the blood and spleen of agonistic anti-TIGIT-treated mice. Additionally, these cells showed an increased activation status but decreased IL-10 production. CONCLUSIONS: Despite the inhibition of splenic T cell responses, agonistic anti-TIGIT treatment does not affect initial atherosclerosis development, possibly due to increased activity of dendritic cells.


Subject(s)
Antibodies/pharmacology , Atherosclerosis/pathology , CD4-Positive T-Lymphocytes/drug effects , Receptors, Immunologic/genetics , Receptors, LDL/genetics , Animals , Atherosclerosis/genetics , Atherosclerosis/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cell Proliferation/drug effects , Cricetulus , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/pathology , Female , Immunoglobulin G/pharmacology , Interleukin-10/genetics , Interleukin-10/immunology , Lymphocyte Activation/drug effects , Mice , Mice, Knockout , Receptors, Immunologic/agonists , Receptors, Immunologic/immunology , Receptors, LDL/deficiency , Receptors, LDL/immunology , Spleen/drug effects , Spleen/immunology , Spleen/pathology
20.
J Immunol ; 191(9): 4573-80, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24068673

ABSTRACT

Patients suffering from cardiovascular disease have well-established atherosclerotic lesions, rendering lesion regression of therapeutic interest. The OX40 (TNFRSF4)-OX40 ligand (OX40L; TNFSF4) pathway is important for the proliferation and survival of T cells, stimulates B cells, and is associated with cardiovascular disease. We hypothesized that interference with the OX40-OX40L pathway, in combination with decreases in cholesterol, may induce regression of atherosclerosis. LDLr(-/-) mice were fed a Western-type diet for 10 wk, after which they received chow diet and were treated with anti-OX40L or PBS for 10 wk. A significant regression of lesions was observed in the aorta and aortic arch of anti-OX40L-treated mice compared with control mice. Interference of the OX40-OX40L pathway reduced Th2 responses, as shown by decreases in GATA-3 and IL-4 levels. Also, IgE levels were decreased, as demonstrated by reduced mast cell presence and activation. Notably, IL-5 production by T and B1 cells was increased, thus enhancing atheroprotective oxidized low-density lipoprotein-specific IgM production. The increase in IL-5 production and IgM was mediated by IL-33 production by APCs upon OX40L blockade. We conclude that interruption of the OX40-OX40L signaling pathway, combined with decreases in dietary cholesterol, induces the regression of atherosclerosis through induction of IL-5-producing T cells and oxidized low-density lipoprotein-specific IgM and reductions in Th2 and mast cells.


Subject(s)
Atherosclerosis/metabolism , Cholesterol/metabolism , Membrane Glycoproteins/metabolism , Receptors, LDL/deficiency , Receptors, OX40/metabolism , Tumor Necrosis Factors/metabolism , Animals , Antibodies, Monoclonal/immunology , Antigen-Presenting Cells/metabolism , Cell Proliferation , Cells, Cultured , GATA3 Transcription Factor/metabolism , Immunoglobulin E/blood , Immunoglobulin M/immunology , Interleukin-33 , Interleukin-4/metabolism , Interleukin-5/biosynthesis , Interleukins/biosynthesis , Lymphocyte Activation/immunology , Male , Mast Cells/immunology , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , OX40 Ligand , Receptors, LDL/genetics , Th2 Cells/immunology , Tumor Necrosis Factors/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...