Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cardiovasc Res ; 118(17): 3346-3359, 2023 01 18.
Article in English | MEDLINE | ID: mdl-35325057

ABSTRACT

AIMS: (Ultra) Small superparamagnetic iron oxide nanoparticles, (U)SPIO, are widely used as magnetic resonance imaging contrast media and assumed to be safe for clinical applications in cardiovascular disease. As safety tests largely relied on normolipidaemic models, not fully representative of the clinical setting, we investigated the impact of (U)SPIOs on disease-relevant endpoints in hyperlipidaemic models of atherosclerosis. METHODS AND RESULTS: RAW264.7 foam cells, exposed in vitro to ferumoxide (dextran-coated SPIO), ferumoxtran (dextran-coated USPIO), or ferumoxytol [carboxymethyl (CM) dextran-coated USPIO] (all 1 mg Fe/mL) showed increased apoptosis and reactive oxygen species accumulation for ferumoxide and ferumoxtran, whereas ferumoxytol was tolerated well. Pro-apoptotic (TUNEL+) and pro-oxidant activity of ferumoxide (0.3 mg Fe/kg) and ferumoxtran (1 mg Fe/kg) were confirmed in plaque, spleen, and liver of hyperlipidaemic ApoE-/- (n = 9/group) and LDLR-/- (n = 9-16/group) mice that had received single IV injections compared with saline-treated controls. Again, ferumoxytol treatment (1 mg Fe/kg) failed to induce apoptosis or oxidative stress in these tissues. Concomitant antioxidant treatment (EUK-8/EUK-134) largely prevented these effects in vitro (-68%, P < 0.05) and in plaques from LDLR-/- mice (-60%, P < 0.001, n = 8/group). Repeated ferumoxtran injections of LDLR-/- mice with pre-existing atherosclerosis enhanced plaque inflammation and apoptosis but did not alter plaque size. Strikingly, carotid artery plaques of endarterectomy patients who received ferumoxtran (2.6 mg Fe/kg) before surgery (n = 9) also showed five-fold increased apoptosis (18.2 vs. 3.7%, respectively; P = 0.004) compared with controls who did not receive ferumoxtran. Mechanistically, neither coating nor particle size seemed accountable for the observed cytotoxicity of ferumoxide and ferumoxtran. CONCLUSIONS: Ferumoxide and ferumoxtran, but not ferumoxytol, induced apoptosis of lipid-laden macrophages in human and murine atherosclerosis, potentially impacting disease progression in patients with advanced atherosclerosis.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Humans , Mice , Animals , Contrast Media , Dextrans/pharmacology , Foam Cells/pathology , Atherosclerosis/diagnostic imaging , Atherosclerosis/drug therapy , Atherosclerosis/pathology , Ferrosoferric Oxide/pharmacology , Magnetic Resonance Imaging/methods , Macrophages/pathology , Apoptosis , Oxides/pharmacology
2.
Sci Rep ; 9(1): 14547, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31601924

ABSTRACT

The anti-apoptotic protein myeloid cell leukemia 1 (Mcl-1) plays an important role in survival and differentiation of leukocytes, more specifically of neutrophils. Here, we investigated the impact of myeloid Mcl-1 deletion in atherosclerosis. Western type diet fed LDL receptor-deficient mice were transplanted with either wild-type (WT) or LysMCre Mcl-1fl/fl (Mcl-1-/-) bone marrow. Mcl-1 myeloid deletion resulted in enhanced apoptosis and lipid accumulation in atherosclerotic plaques. In vitro, Mcl-1 deficient macrophages also showed increased lipid accumulation, resulting in increased sensitivity to lipid-induced cell death. However, plaque size, necrotic core and macrophage content were similar in Mcl-1-/- compared to WT mice, most likely due to decreased circulating and plaque-residing neutrophils. Interestingly, Mcl-1-/- peritoneal foam cells formed up to 45% more multinucleated giant cells (MGCs) in vitro compared to WT, which concurred with an increased MGC presence in atherosclerotic lesions of Mcl-1-/- mice. Moreover, analysis of human unstable atherosclerotic lesions also revealed a significant inverse correlation between MGC lesion content and Mcl-1 gene expression, coinciding with the mouse data. Taken together, these findings suggest that myeloid Mcl-1 deletion leads to a more apoptotic, lipid and MGC-enriched phenotype. These potentially pro-atherogenic effects are however counteracted by neutropenia in circulation and plaque.


Subject(s)
Apoptosis , Giant Cells/cytology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , 3T3 Cells , Animals , Atherosclerosis/genetics , Atherosclerosis/metabolism , Cell Differentiation , Gene Deletion , Humans , Immunohistochemistry , Lipids/chemistry , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Neutrophils/metabolism , Phenotype , Plaque, Atherosclerotic/metabolism
3.
Sci Rep ; 7(1): 3086, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28596542

ABSTRACT

Proapoptotic Bcl-2 family member Bim is particularly relevant for deletion of autoreactive and activated T and B cells, implicating Bim in autoimmunity. As atherosclerosis is a chronic inflammatory process with features of autoimmune disease, we investigated the impact of hematopoietic Bim deficiency on plaque formation and parameters of plaque stability. Bim -/- or wild type bone marrow transplanted ldlr -/- mice were fed a Western type diet (WTD) for 5 or 10 weeks, after which they were immunophenotyped and atherosclerotic lesions were analyzed. Bim -/- transplanted mice displayed splenomegaly and overt lymphocytosis. CD4+ and CD8+ T cells were more activated (increased CD69 and CD71 expression, increased interferon gamma production). B cells were elevated by 147%, with a shift towards the pro-atherogenic IgG-producing B2 cell phenotype, resulting in a doubling of anti-oxLDL IgG1 antibody titers in serum of bim -/- mice. Bim -/- mice displayed massive intraplaque accumulation of Ig complexes and of lesional T cells, although this did not translate in changes in plaque size or stability features (apoptotic cell and macrophage content). The surprising lack in plaque phenotype despite the profound pro-atherogenic immune effects may be attributable to the sharp reduction of serum cholesterol levels in WTD fed bim -/- mice.


Subject(s)
Atherosclerosis/genetics , Autoimmune Diseases/etiology , Bcl-2-Like Protein 11/deficiency , Inflammation/etiology , Leukocytes/immunology , Leukocytes/metabolism , Receptors, LDL/deficiency , Animals , Apoptosis/genetics , Autoimmune Diseases/pathology , Bcl-2-Like Protein 11/genetics , Bone Marrow Transplantation , Disease Models, Animal , Hyperlipidemias , Immunity, Humoral , Immunoglobulins/immunology , Inflammation/pathology , Lymphocyte Count , Mice , Mice, Knockout , Receptors, LDL/genetics , Splenomegaly , Th1 Cells/immunology , Th1 Cells/metabolism
4.
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
5.
Atherosclerosis ; 235(1): 196-203, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24858338

ABSTRACT

AIM: Neuropeptide Y is an abundantly expressed neurotransmitter capable of modulating both immune and metabolic responses related to the development of atherosclerosis. NPY receptors are expressed by a number of vascular wall cell types, among which mast cells. However, the direct effects of NPY on atherosclerotic plaque development and progression remain to be investigated. In this study we thus aimed to determine whether NPY is expressed in atherosclerotic plaques and to establish its role in atherosclerotic plaque development. METHODS AND RESULTS: NPY expression was seen to be increased up to 2-fold in unstable human endarterectomy plaques, as compared to stable plaques, and to be significantly upregulated during lesion progression in apoE(-/-) mice. In apoE(-/-) mice focal overexpression of NPY in the carotid artery significantly increased atherosclerotic plaque size compared to controls, while plaque composition was unaffected. Interestingly, perivascular mast cell activation was significantly higher in the NPY-overexpressing mice, suggesting that NPY may impact plaque progression in part via mast cell activation. Furthermore, in vitro NPY-induced murine mast cell activation resulted in the release of pro-atherogenic mediators including IL-6 and tryptase. CONCLUSIONS: Our data show that NPY expression is increased during atherogenesis and in particular in unstable plaques. Furthermore, perivascular overexpression of NPY promoted plaque development and perivascular mast cell activation, suggestive of a role for NPY-induced mast cell activation in lesion progression.


Subject(s)
Atherosclerosis/blood , Mast Cells/cytology , Neuropeptide Y/blood , Plaque, Atherosclerotic/pathology , Animals , Apolipoproteins E/blood , Disease Progression , Gene Expression Profiling , Gene Expression Regulation , HEK293 Cells , Humans , Immunohistochemistry , Inflammation/blood , Interleukin-6/blood , Lentivirus/genetics , Mice , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Plaque, Atherosclerotic/blood , Tryptases/blood
6.
PLoS One ; 8(5): e63360, 2013.
Article in English | MEDLINE | ID: mdl-23700419

ABSTRACT

AIMS: Altered sphingosine 1-phosphate (S1P) homeostasis and signaling is implicated in various inflammatory diseases including atherosclerosis. As S1P levels are tightly controlled by S1P lyase, we investigated the impact of hematopoietic S1P lyase (Sgpl1(-/-)) deficiency on leukocyte subsets relevant to atherosclerosis. METHODS AND RESULTS: LDL receptor deficient mice that were transplanted with Sgpl1(-/-) bone marrow showed disrupted S1P gradients translating into lymphopenia and abrogated lymphocyte mitogenic and cytokine response as compared to controls. Remarkably however, Sgpl1(-/-) chimeras displayed mild monocytosis, due to impeded stromal retention and myelopoiesis, and plasma cytokine and macrophage expression patterns, that were largely compatible with classical macrophage activation. Collectively these two phenotypic features of Sgpl1 deficiency culminated in diminished atherogenic response. CONCLUSIONS: Here we not only firmly establish the critical role of hematopoietic S1P lyase in controlling S1P levels and T cell trafficking in blood and lymphoid tissue, but also identify leukocyte Sgpl1 as critical factor in monocyte macrophage differentiation and function. Its, partly counterbalancing, pro- and anti-inflammatory activity spectrum imply that intervention in S1P lyase function in inflammatory disorders such as atherosclerosis should be considered with caution.


Subject(s)
Aldehyde-Lyases/deficiency , Atherosclerosis/enzymology , Plaque, Atherosclerotic/enzymology , Receptors, LDL/deficiency , Aldehyde-Lyases/genetics , Animals , Atherosclerosis/immunology , Atherosclerosis/pathology , Bone Marrow Cells/enzymology , Cell Differentiation , Female , Hematopoiesis , Lymphocyte Count , Lymphopenia/enzymology , Lymphopenia/immunology , Lysophospholipids/metabolism , Macrophages/enzymology , Macrophages/immunology , Macrophages/physiology , Mice , Mice, Knockout , Neutrophils/enzymology , Phenotype , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/pathology , Receptors, LDL/genetics , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Spleen/metabolism
7.
FASEB J ; 27(1): 265-76, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23047899

ABSTRACT

Leukocyte chemotaxis is deemed instrumental in initiation and progression of atherosclerosis. It is mediated by G-protein-coupled receptors (e.g., CCR2 and CCR5), the activity of which is controlled by G-protein-coupled receptor kinases (GRKs). In this study, we analyzed the effect of hematopoietic deficiency of a potent regulator kinase of chemotaxis (GRK2) on atherogenesis. LDL receptor-deficient (LDLr(-/-)) mice with heterozygous hematopoietic GRK2 deficiency, generated by bone marrow transplantation (n=15), displayed a dramatic attenuation of plaque development, with 79% reduction in necrotic core and increased macrophage content. Circulating monocytes decreased and granulocytes increased in GRK2(+/-) chimeras, which could be attributed to diminished granulocyte colony-forming units in bone marrow. Collectively, these data pointed to myeloid cells as major mediators of the impaired atherogenic response in GRK2(+/-) chimeras. LDLr(-/-) mice with macrophage/granulocyte-specific GRK2 deficiency (LysM-Cre GRK2(flox/flox); n=8) failed to mimic the aforementioned phenotype, acquitting these cells as major responsible subsets for GRK2 deficiency-associated atheroprotection. To conclude, even partial hematopoietic GRK2 deficiency prevents atherosclerotic lesion progression beyond the fatty streak stage, identifying hematopoietic GRK2 as a potential target for intervention in atherosclerosis.


Subject(s)
Atherosclerosis/pathology , G-Protein-Coupled Receptor Kinase 2/genetics , Receptors, LDL/genetics , Animals , Apoptosis , Atherosclerosis/genetics , Female , Flow Cytometry , Mice , Mice, Knockout , Phagocytosis
SELECTION OF CITATIONS
SEARCH DETAIL
...