Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.097
Filter
1.
Arterioscler Thromb Vasc Biol ; 44(6): 1407-1418, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38695168

ABSTRACT

BACKGROUND: LCAT (lecithin cholesterol acyl transferase) catalyzes the conversion of unesterified, or free cholesterol, to cholesteryl ester, which moves from the surface of HDL (high-density lipoprotein) into the neutral lipid core. As this iterative process continues, nascent lipid-poor HDL is converted to a series of larger, spherical cholesteryl ester-enriched HDL particles that can be cleared by the liver in a process that has been termed reverse cholesterol transport. METHODS: We conducted a randomized, placebocontrolled, crossover study in 5 volunteers with atherosclerotic cardiovascular disease, to examine the effects of an acute increase of recombinant human (rh) LCAT via intravenous administration (300-mg loading dose followed by 150 mg at 48 hours) on the in vivo metabolism of HDL APO (apolipoprotein)A1 and APOA2, and the APOB100-lipoproteins, very low density, intermediate density, and low-density lipoproteins. RESULTS: As expected, recombinant human LCAT treatment significantly increased HDL-cholesterol (34.9 mg/dL; P≤0.001), and this was mostly due to the increase in cholesteryl ester content (33.0 mg/dL; P=0.014). This change did not affect the fractional clearance or production rates of HDL-APOA1 and HDL-APOA2. There were also no significant changes in the metabolism of APOB100-lipoproteins. CONCLUSIONS: Our results suggest that an acute increase in LCAT activity drives greater flux of cholesteryl ester through the reverse cholesterol transport pathway without significantly altering the clearance and production of the main HDL proteins and without affecting the metabolism of APOB100-lipoproteins. Long-term elevations of LCAT might, therefore, have beneficial effects on total body cholesterol balance and atherogenesis.


Subject(s)
Apolipoprotein A-II , Apolipoprotein A-I , Cholesterol, HDL , Cross-Over Studies , Phosphatidylcholine-Sterol O-Acyltransferase , Recombinant Proteins , Humans , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Male , Apolipoprotein A-I/blood , Middle Aged , Cholesterol, HDL/blood , Apolipoprotein A-II/blood , Female , Cholesterol Esters/blood , Cholesterol Esters/metabolism , Atherosclerosis/drug therapy , Atherosclerosis/enzymology , Atherosclerosis/blood , Apolipoprotein B-100/blood , Aged , Adult , Lipoproteins/blood , Lipoproteins/metabolism
2.
Atherosclerosis ; 392: 117519, 2024 May.
Article in English | MEDLINE | ID: mdl-38581737

ABSTRACT

BACKGROUND AND AIMS: Atherosclerosis is the primary underlying cause of myocardial infarction and stroke, which are the major causes of death globally. Heparanase (Hpse) is a pro-inflammatory extracellular matrix degrading enzyme that has been implicated in atherogenesis. However, to date the precise roles of Hpse in atherosclerosis and its mechanisms of action are not well defined. This study aims to provide new insights into the contribution of Hpse in different stages of atherosclerosis in vivo. METHODS: We generated Hpse gene-deficient mice on the atherosclerosis-prone apolipoprotein E gene knockout (ApoE-/-) background to investigate the impact of Hpse gene deficiency on the initiation and progression of atherosclerosis after 6 and 14 weeks high-fat diet feeding, respectively. Atherosclerotic lesion development, blood serum profiles, lesion composition and aortic immune cell populations were evaluated. RESULTS: Hpse-deficient mice exhibited significantly reduced atherosclerotic lesion burden in the aortic sinus and aorta at both time-points, independent of changes in plasma cholesterol levels. A significant reduction in the necrotic core size and an increase in smooth muscle cell content were also observed in advanced atherosclerotic plaques of Hpse-deficient mice. Additionally, Hpse deficiency reduced circulating and aortic levels of VCAM-1 at the initiation and progression stages of disease and circulating MCP-1 levels in the initiation but not progression stage. Moreover, the aortic levels of total leukocytes and dendritic cells in Hpse-deficient ApoE-/- mice were significantly decreased compared to control ApoE-/-mice at both disease stages. CONCLUSIONS: This study identifies Hpse as a key pro-inflammatory enzyme driving the initiation and progression of atherosclerosis and highlighting the potential of Hpse inhibitors as novel anti-inflammatory treatments for cardiovascular disease.


Subject(s)
Aorta , Atherosclerosis , Disease Models, Animal , Disease Progression , Glucuronidase , Mice, Knockout, ApoE , Plaque, Atherosclerotic , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/enzymology , Atherosclerosis/metabolism , Glucuronidase/deficiency , Glucuronidase/genetics , Glucuronidase/metabolism , Aorta/pathology , Aorta/metabolism , Aorta/enzymology , Aortic Diseases/pathology , Aortic Diseases/genetics , Aortic Diseases/enzymology , Aortic Diseases/metabolism , Diet, High-Fat , Apolipoproteins E/genetics , Apolipoproteins E/deficiency , Mice, Inbred C57BL , Male , Vascular Cell Adhesion Molecule-1/metabolism , Mice , Mice, Knockout , Sinus of Valsalva/pathology , Necrosis
3.
Arterioscler Thromb Vasc Biol ; 44(6): 1393-1406, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38660804

ABSTRACT

BACKGROUND: Low-dose aspirin is widely used for the secondary prevention of cardiovascular disease. The beneficial effects of low-dose aspirin are attributable to its inhibition of platelet Cox (cyclooxygenase)-1-derived thromboxane A2. Until recently, the use of the Pf4 (platelet factor 4) Cre has been the only genetic approach to generating megakaryocyte/platelet ablation of Cox-1 in mice. However, Pf4-ΔCre displays ectopic expression outside the megakaryocyte/platelet lineage, especially during inflammation. The use of the Gp1ba (glycoprotein 1bα) Cre promises a more specific, targeted approach. METHODS: To evaluate the role of Cox-1 in platelets, we crossed Pf4-ΔCre or Gp1ba-ΔCre mice with Cox-1flox/flox mice to generate platelet Cox-1-/- mice on normolipidemic and hyperlipidemic (Ldlr-/-; low-density lipoprotein receptor) backgrounds. RESULTS: Ex vivo platelet aggregation induced by arachidonic acid or adenosine diphosphate in platelet-rich plasma was inhibited to a similar extent in Pf4-ΔCre Cox-1-/-/Ldlr-/- and Gp1ba-ΔCre Cox-1-/-/Ldlr-/- mice. In a mouse model of tail injury, Pf4-ΔCre-mediated and Gp1ba-ΔCre-mediated deletions of Cox-1 were similarly efficient in suppressing platelet prostanoid biosynthesis. Experimental thrombogenesis and attendant blood loss were similar in both models. However, the impact on atherogenesis was divergent, being accelerated in the Pf4-ΔCre mice while restrained in the Gp1ba-ΔCres. In the former, accelerated atherogenesis was associated with greater suppression of PGI2 biosynthesis, a reduction in the lipopolysaccharide-evoked capacity to produce PGE2 (prostaglandin E) and PGD2 (prostanglandin D), activation of the inflammasome, elevated plasma levels of IL-1ß (interleukin), reduced plasma levels of HDL-C (high-density lipoprotein receptor-cholesterol), and a reduction in the capacity for reverse cholesterol transport. By contrast, in the latter, plasma HDL-C and α-tocopherol were elevated, and MIP-1α (macrophage inflammatory protein-1α) and MCP-1 (monocyte chemoattractant protein 1) were reduced. CONCLUSIONS: Both approaches to Cox-1 deletion similarly restrain thrombogenesis, but a differential impact on Cox-1-dependent prostanoid formation by the vasculature may contribute to an inflammatory phenotype and accelerated atherogenesis in Pf4-ΔCre mice.


Subject(s)
Blood Platelets , Cyclooxygenase 1 , Disease Models, Animal , Integrases , Mice, Inbred C57BL , Mice, Knockout , Platelet Aggregation , Platelet Factor 4 , Receptors, LDL , Animals , Blood Platelets/metabolism , Blood Platelets/drug effects , Blood Platelets/enzymology , Cyclooxygenase 1/metabolism , Cyclooxygenase 1/genetics , Cyclooxygenase 1/deficiency , Platelet Aggregation/drug effects , Platelet Factor 4/genetics , Platelet Factor 4/metabolism , Integrases/genetics , Receptors, LDL/genetics , Receptors, LDL/deficiency , Male , Mice , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/enzymology , Atherosclerosis/prevention & control , Atherosclerosis/blood , Hyperlipidemias/blood , Hyperlipidemias/genetics , Hyperlipidemias/enzymology , Phenotype , Membrane Proteins , Platelet Glycoprotein GPIb-IX Complex
4.
Thromb Haemost ; 124(6): 584-594, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38109905

ABSTRACT

OBJECTIVE: Aldehyde dehydrogenase 4A1 (ALDH4A1) was recently reported to be a novel autoantigen of atherosclerosis. However, its role in different phenotypes of acute coronary syndrome remains unclear. Herein, we planned to explore the circulating and regional expression of ALDH4A1 in patients with plaque rupture (PR) and plaque erosion (PE) determined by optical coherence tomography (OCT). METHODS AND RESULTS: After applying the inclusion and exclusion criteria, a prospective series of 312 patients with ST segment elevated myocardial infarction (STEMI), including 161 patients with PR and 151 patients with PE determined by OCT, were enrolled for plasma ALDH4A1 testing. In addition, ALDH4A1 was quantified using immunofluorescence in aspirated coronary thrombus samples obtained from 31 patients with PR and 25 patients with PE. In addition, we established an atherosclerosis mouse model and analyzed the distribution of ALDH4A1 expression in different mouse organs. Furthermore, we compared the level of ALDH4A1 in the spleen and carotid artery between Apoe-/- and C57 mice. The results showed that the plasma level of ALDH4A1 was significantly higher in STEMI patients with PE than in those with PR (4.6 ng/mL [2.2-8.7] vs. 3.5 ng/mL [1.6-5.6] p = 0.005). The expression of ALDH4A1 in aspirated coronary thrombi was also significantly higher in patients with PE than in those with PR (mean gray value: 32.0 [23.6-40.6] vs. 16.8 [14.0-24.5], p < 0.001). In animal models, the expression of ALDH4A1 is much higher in the spleen than in other organs, and the level of ALDH4A1 is significantly elevated in the spleen and carotid artery of Apoe-/- mice compared with C57 mice. CONCLUSION: The high levels of ALDH4A1 in the plasma and aspirated coronary thrombi independently correlated with PE in patients with STEMI. These results suggested that ALDH4A1 is involved in the mechanism of PE and serves as a promising biomarker and treatment target for patients with PE.


Subject(s)
Autoantigens , Mice, Inbred C57BL , Plaque, Atherosclerotic , ST Elevation Myocardial Infarction , Animals , Humans , Male , Autoantigens/immunology , Middle Aged , Female , ST Elevation Myocardial Infarction/blood , Aged , Mice , Biomarkers/blood , Prospective Studies , Mice, Knockout, ApoE , Disease Models, Animal , Spleen/pathology , Carotid Arteries/pathology , Carotid Arteries/diagnostic imaging , Aldehyde Dehydrogenase/metabolism , Coronary Artery Disease/immunology , Coronary Artery Disease/blood , Rupture, Spontaneous , Atherosclerosis/enzymology
5.
Commun Biol ; 5(1): 132, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35169231

ABSTRACT

Atherosclerosis is a chronic inflammatory condition in which macrophages play a major role. Janus kinase 2 (JAK2) is a pivotal molecule in inflammatory and metabolic signaling, and Jak2V617F activating mutation has recently been implicated with enhancing clonal hematopoiesis and atherosclerosis. To determine the essential in vivo role of macrophage (M)-Jak2 in atherosclerosis, we generate atherosclerosis-prone ApoE-null mice deficient in M-Jak2. Contrary to our expectation, these mice exhibit increased plaque burden with no differences in macrophage proliferation, recruitment or bone marrow clonal expansion. Notably, M-Jak2-deficient bone marrow derived macrophages show a significant defect in cholesterol efflux. Pharmacologic JAK2 inhibition with ruxolitinib also leads to defects in cholesterol efflux and accelerates atherosclerosis. Liver X receptor agonist abolishes the efflux defect and attenuates the accelerated atherosclerosis that occurs with M-Jak2 deficiency. Macrophages of individuals with the Jak2V617F mutation show increased efflux which is normalized when treated with a JAK2 inhibitor. Together, M-Jak2-deficiency leads to accelerated atherosclerosis primarily through defects in cholesterol efflux from macrophages.


Subject(s)
Atherosclerosis , Cholesterol , Janus Kinase 2 , Animals , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Cholesterol/metabolism , Janus Kinase 2/deficiency , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL
6.
Cardiovasc Res ; 118(2): 638-653, 2022 01 29.
Article in English | MEDLINE | ID: mdl-33599243

ABSTRACT

AIMS: Pathological arterial remodelling including neointimal hyperplasia and atherosclerosis is the main underlying cause for occluding arterial diseases. Cezanne is a novel deubiquitinating enzyme, functioning as a NF-кB negative regulator, and plays a key role in renal inflammatory response and kidney injury induced by ischaemia. Here we attempted to examine its pathological role in vascular smooth muscle cell (VSMC) pathology and arterial remodelling. METHODS AND RESULTS: Cezanne expression levels were consistently induced by various atherogenic stimuli in VSMCs, and in remodelled arteries upon injury. Functionally, VSMCs over-expressing wild-type Cezanne, but not the mutated catalytically-inactive Cezanne (C209S), had an increased proliferative ability and mobility, while the opposite was observed in VSMCs with Cezanne knockdown. Surprisingly, we observed no significant effects of Cezanne on VSMC apoptosis, NF-κB signalling, or inflammation. RNA-sequencing and biochemical studies showed that Cezanne drives VSMC proliferation by regulating CCN family member 1 (CCN1) by targeting ß-catenin for deubiquitination. Importantly, local correction of Cezanne expression in the injured arteries greatly decreased VSMC proliferation, and prevented arterial inward remodelling. Interestingly, global Cezanne gene deletion in mice led to smaller atherosclerotic plaques, but with a lower level of plaque stability. Translating, we observed a similar role for Cezanne in human VSMCs, and higher expression levels of Cezanne in human atherosclerotic lesions. CONCLUSION: Cezanne is a key regulator of VSMC proliferation and migration in pathological arterial remodelling. Our findings have important implications for therapeutic targeting Cezanne signalling and VSMC pathology in vascular diseases.


Subject(s)
Atherosclerosis/enzymology , Endopeptidases/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Vascular Remodeling , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Apoptosis , Atherosclerosis/genetics , Atherosclerosis/pathology , Cell Movement , Cell Proliferation , Cells, Cultured , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/metabolism , Disease Models, Animal , Endopeptidases/genetics , Humans , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , NF-kappa B/metabolism , Neointima , Ubiquitination , beta Catenin/genetics
7.
Microvasc Res ; 140: 104276, 2022 03.
Article in English | MEDLINE | ID: mdl-34742813

ABSTRACT

PURPOSE: We previously reported that a calpain inhibitor (CAI) prevents the development of atherosclerosis in rats. This study aimed to investigate the effects of CAI (1 mg/kg) on atherosclerosis in apolipoprotein E knockout (ApoE KO) mice that were fed a high-fat diet (HFD) and explore the underlying mechanism by analyzing the expression of genes related to the uptake and efflux of cholesterol. METHODS: Atherosclerotic plaques were evaluated. The activity of calpain in the aorta and that of superoxide dismutase (SOD) in the serum were assessed. Lipid profiles in the serum and liver were examined. Serum oxidized low-density lipoprotein (oxLDL), malondialdehyde (MDA), tumor necrosis factor (TNF-α), and interleukin-6 (IL-6) levels were measured. The mRNA expressions of CD68, TNF-α, IL-6, CD36, scavenger receptor (SR-A), peroxisome proliferator-activated receptor gamma (PPAR-γ), liver-x-receptor alpha (LXR-α), and ATP-binding cassette transporter class A1 (ABCA1) in the aorta and peritoneal macrophages were also evaluated. RESULTS: CAI reduced calpain activity in the aorta. CAI also impeded atherosclerotic lesion formation and mRNA expression of CD68 in the aorta and peritoneal macrophages of ApoE KO mice compared with those of mice receiving HFD. However, CAI had no effect on body weight and lipid levels in both the serum and liver. CAI significantly decreased MDA, oxLDL, TNF-α, and IL-6 levels and increased SOD activity in the serum. Moreover, CAI significantly inhibited the mRNA expression of TNF-α and IL-6 genes in the aorta and peritoneal macrophages. In addition, CAI significantly downregulated the mRNA expression of scavenger receptors CD36 and SR-A and upregulated the expression of genes involved in the cholesterol efflux pathway, i.e., PPAR-γ, LXR-α, and ABCA1 in the aorta and peritoneal macrophages. CONCLUSIONS: CAI inhibited the development of atherosclerotic lesions in ApoE KO mice, and this effect might be related to the reduction of oxidative stress and inflammation and the improvement of cholesterol intake and efflux pathways.


Subject(s)
Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Calpain/antagonists & inhibitors , Cholesterol/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Leupeptins/pharmacology , Lipid Metabolism/drug effects , Macrophages, Peritoneal/drug effects , RNA, Messenger/metabolism , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Aorta/enzymology , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Calpain/metabolism , Disease Models, Animal , Gene Expression Regulation , Lipid Metabolism/genetics , Liver X Receptors/genetics , Liver X Receptors/metabolism , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/pathology , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , PPAR gamma/genetics , PPAR gamma/metabolism , Plaque, Atherosclerotic , RNA, Messenger/genetics , Scavenger Receptors, Class A/genetics , Scavenger Receptors, Class A/metabolism
8.
Microvasc Res ; 139: 104252, 2022 01.
Article in English | MEDLINE | ID: mdl-34520772

ABSTRACT

Soluble fms-like tyrosine kinase-1 (sFlt-1), a circulating antiangiogenic protein, is involved in the pathogenesis of atherosclerosis (AS), and the underlying mechanism is still unclear. Here, we attempted to investigate the mechanism of action of sFlt-1 in AS. Human umbilical vein endothelial cells (HUVECs) were treated with oxidized low density lipoprotein (ox-LDL) to induce cell injury. ox-LDL treatment increased LC3-II/LC3-I ratio, Beclin-1 expression and GFP-LC3 puncta in HUVECs, suggesting that ox-LDL may induce autophagic flux impairment in HUVECs. ox-LDL-treated HUVECs displayed a decrease of sFlt-1 levels. Moreover, ox-LDL treatment reduced cell proliferation and elevated apoptosis in HUVECs, which was abrogated by sFlt-1 overexpression. Up-regulation of sFlt-1 repressed the activity of PI3K/AKT/mTOR signaling pathway and enhanced autophagy in HUVECs following ox-LDL treatment. Additionally, sFlt-1 overexpression-mediated increase of autophagy in ox-LDL-treated HUVECs was abolished by 3-methyladenine (autophagy inhibitor). 3-methyladenine abrogated the impact of sFlt-1 overexpression on proliferation and apoptosis in ox-LDL-treated HUVECs. This work confirmed that overexpression of sFlt-1 activated autophagy by repressing PI3K/Akt/mTOR signaling pathway, and thus alleviated ox-LDL-induced injury of HUVECs. Therefore, this study suggests that sFlt-1 may be a potential target for AS treatment.


Subject(s)
Atherosclerosis/enzymology , Autophagy/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Lipoproteins, LDL/toxicity , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Apoptosis/drug effects , Atherosclerosis/genetics , Atherosclerosis/pathology , Beclin-1/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Human Umbilical Vein Endothelial Cells/pathology , Humans , Microtubule-Associated Proteins/metabolism , Signal Transduction , Up-Regulation , Vascular Endothelial Growth Factor Receptor-1/genetics
9.
Arterioscler Thromb Vasc Biol ; 42(1): 67-86, 2022 01.
Article in English | MEDLINE | ID: mdl-34809446

ABSTRACT

OBJECTIVE: PCSK9 (proprotein convertase subtilisin/kexin type 9) plays a critical role in cholesterol metabolism via the PCSK9-LDLR (low-density lipoprotein receptor) axis in the liver; however, evidence indicates that PCSK9 directly contributes to the pathogenesis of various diseases through mechanisms independent of its LDL-cholesterol regulation. The objective of this study was to determine how PCSK9 directly acts on vascular smooth muscle cells (SMCs), contributing to degenerative vascular disease. Approach and Results: We first examined the effects of PCSK9 on cultured human aortic SMCs. Overexpression of PCSK9 downregulated the expression of ApoER2 (apolipoprotein E receptor 2), a known target of PCSK9. Treatment with soluble recombinant human ApoER2 or the DNA synthesis inhibitor, hydroxyurea, inhibited PCSK9-induced polyploidization and other cellular responses of human SMCs. Treatment with antibodies against ApoER2 resulted in similar effects to those observed with PCSK9 overexpression. Inducible, SMC-specific knockout of Pcsk9 accelerated neointima formation in mouse carotid arteries and reduced age-related arterial stiffness. PCSK9 was expressed in SMCs of human atherosclerotic lesions and abundant in the "shoulder" regions of vulnerable atherosclerotic plaques. PCSK9 was also expressed in SMCs of abdominal aortic aneurysm, which was inversely related to the expression of smooth muscle α-actin. CONCLUSIONS: Our findings demonstrate that PCSK9 inhibits proliferation and induces polyploidization, senescence, and apoptosis, which may be relevant to various degenerative vascular diseases.


Subject(s)
Apoptosis , Atherosclerosis/enzymology , Cell Proliferation , Cellular Senescence , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Proprotein Convertase 9/metabolism , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/pathology , Neointima , Plaque, Atherosclerotic , Proprotein Convertase 9/genetics , Signal Transduction , Vascular Stiffness
10.
Cells ; 10(11)2021 10 20.
Article in English | MEDLINE | ID: mdl-34831028

ABSTRACT

Coronary artery disease caused by atherosclerosis is a major cause of morbidity and mortality around the world. Data from preclinical and clinical studies support the belief that atherosclerosis is an inflammatory disease that is mediated by innate and adaptive immune signaling mechanisms. This review sought to highlight the role of Rac-mediated inflammatory signaling in the mechanisms driving atherosclerotic calcification. In addition, current clinical treatment strategies that are related to targeting hypercholesterolemia as a critical risk factor for atherosclerotic vascular disease are addressed in relation to the effects on Rac immune signaling and the implications for the future of targeting immune responses in the treatment of calcific atherosclerosis.


Subject(s)
Atherosclerosis/enzymology , Atherosclerosis/immunology , Signal Transduction , rac GTP-Binding Proteins/metabolism , Amino Acid Sequence , Atherosclerosis/drug therapy , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Inflammation/complications , Inflammation/pathology , Models, Biological , rac GTP-Binding Proteins/chemistry
11.
Life Sci ; 284: 119935, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34508760

ABSTRACT

OBJECTIVE: Atherosclerotic vascular disease remains the principal cause of death and disability among patients with type 2 diabetes. Unfortunately, the problem is not adequately resolved by therapeutic strategies with currently available drugs or approaches that solely focus on optimal glycemic control. To identify the key contributors and better understand the mechanism of diabetic atherosclerotic vascular disease, we aimed to elucidate the key genetic characteristics and pathological pathways in atherosclerotic vascular disease through nonbiased bioinformatics analysis and subsequent experimental demonstration and exploration in diabetic atherosclerotic vascular disease. METHODS AND RESULTS: Sixty-eight upregulated and 23 downregulated genes were identified from the analysis of gene expression profiles (GSE30169 and GSE6584). A comprehensive bioinformatic assay further identified that ferroptosis, a new type of programmed cell death and HMOX1 (a gene that encodes heme oxygenase), were vital factors in atherosclerotic vascular disease. We further demonstrated that diabetes significantly increased ferroptosis and HMOX1 levels compared to normal controls. Importantly, the ferroptosis inhibitor ferrostatin-1 (Fer-1) effectively attenuated diabetic atherosclerosis, suggesting the causative role of ferroptosis in diabetic atherosclerosis development. At the cellular level, Fer-1 ameliorated high glucose high lipid-induced lipid peroxidation and downregulated ROS production. More importantly, HMOX1 knockdown attenuated Fe2+ overload, reduced iron content and ROS, and alleviated lipid peroxidation, which led to a reduction in ferroptosis in diabetic human endothelial cells. CONCLUSIONS: We demonstrated that HMOX1 upregulation is responsible for the increased ferroptosis in diabetic atherosclerosis development, suggesting that HMOX1 may serve as a potential therapeutic or drug development target for diabetic atherosclerosis.


Subject(s)
Atherosclerosis/enzymology , Atherosclerosis/genetics , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/genetics , Ferroptosis , Heme Oxygenase-1/genetics , Up-Regulation , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Atherosclerosis/complications , Atherosclerosis/pathology , Cyclohexylamines/pharmacology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Diet, High-Fat , Disease Progression , Feeding Behavior , Female , Ferroptosis/drug effects , Gene Expression Profiling , Glutathione/metabolism , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Iron Overload/complications , Lipid Peroxidation/drug effects , Male , Mice, Knockout , NADP/metabolism , Phenylenediamines/pharmacology , Protein Interaction Maps/drug effects , Protein Interaction Maps/genetics , Signal Transduction/drug effects , Up-Regulation/drug effects , Up-Regulation/genetics
12.
J Clin Invest ; 131(15)2021 08 02.
Article in English | MEDLINE | ID: mdl-34338228

ABSTRACT

Endothelial-mesenchymal transition (EndMT) is associated with various cardiovascular diseases and in particular with atherosclerosis and plaque instability. However, the molecular pathways that govern EndMT are poorly defined. Specifically, the role of epigenetic factors and histone deacetylases (HDACs) in controlling EndMT and the atherosclerotic plaque phenotype remains unclear. Here, we identified histone deacetylation, specifically that mediated by HDAC9 (a class IIa HDAC), as playing an important role in both EndMT and atherosclerosis. Using in vitro models, we found class IIa HDAC inhibition sustained the expression of endothelial proteins and mitigated the increase in mesenchymal proteins, effectively blocking EndMT. Similarly, ex vivo genetic knockout of Hdac9 in endothelial cells prevented EndMT and preserved a more endothelial-like phenotype. In vivo, atherosclerosis-prone mice with endothelial-specific Hdac9 knockout showed reduced EndMT and significantly reduced plaque area. Furthermore, these mice displayed a more favorable plaque phenotype, with reduced plaque lipid content and increased fibrous cap thickness. Together, these findings indicate that HDAC9 contributes to vascular pathology by promoting EndMT. Our study provides evidence for a pathological link among EndMT, HDAC9, and atherosclerosis and suggests that targeting of HDAC9 may be beneficial for plaque stabilization or slowing the progression of atherosclerotic disease.


Subject(s)
Atherosclerosis/enzymology , Endothelium/enzymology , Histone Deacetylases/metabolism , Plaque, Atherosclerotic/enzymology , Repressor Proteins/metabolism , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Endothelium/pathology , Histone Deacetylases/genetics , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Knockout, ApoE , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/pathology , Repressor Proteins/genetics
13.
Int. j. cardiovasc. sci. (Impr.) ; 34(4): 411-419, July-Aug. 2021. tab, graf
Article in English | LILACS | ID: biblio-1286822

ABSTRACT

Abstract Background Sex-specific pathology of coronary artery disease (CAD) has not been recognized. Women with obstructive or nonobstructive CAD associated with traditional risk factors have similar events; no studies have explored both populations in association with genetic markers. Objective To evaluate the DD genotype in overweight menopausal women and its association with CAD and traditional risk factors. Method This cross-sectional study included 356 menopausal women who underwent coronary angiography as CAD assessment. The patients' DNA was extracted and polymorphisms were detected with a single polymerase chain reaction assay. Two groups were formed based on luminal lesions (normal [n = 134] or pathological [n = 222]) with a cutoff value > 30%, considering overweight and age. The chi-square test, Student's t-test, and multivariate logistic regression were performed as appropriate (p < 0.05) using the following variables: overweight, diabetes, hypertension, dyslipidemia, smoking status, sedentary lifestyle, and a family history of CAD. Results The mean age of the sample was 63 + 8 years, and the mean BMI was 28 + 5 kg/m2. The DD genotype was slightly more prevalent in the pathological group (30.2% vs. 21.6%, p = 0.079), but this significantly changed when BMI > 25 was considered (33% vs. 18%, p = 0.012). In multivariate analysis with two threshold levels (> 50 and > 60 years), diabetes was significantly associated with CAD in both models (p = 0.021 vs. 0.009) but the genotype was only associated with younger age (p = 0.034). Conclusion These data support an association between atherosclerosis and the renin-angiotensin system in overweight menopausal women that is dependent on the age at which the ischemic event occurs.


Subject(s)
Humans , Female , Coronary Artery Disease/etiology , Genetic Markers , Atherosclerosis/enzymology , Menopause , Cross-Sectional Studies , Retrospective Studies , Diabetes Mellitus , Overweight , Heart Disease Risk Factors , Genotype
14.
Clin Sci (Lond) ; 135(15): 1845-1858, 2021 08 13.
Article in English | MEDLINE | ID: mdl-34269800

ABSTRACT

OBJECTIVE: The mechanisms involved in NOX5 activation in atherosclerotic processes are not completely understood. The present study tested the hypothesis that lysophosphatidylcholine (LPC), a proatherogenic component of oxLDL, induces endothelial calcium influx, which drives NOX5-dependent reactive oxygen species (ROS) production, oxidative stress, and endothelial cell dysfunction. APPROACH: Human aortic endothelial cells (HAEC) were stimulated with LPC (10-5 M, for different time points). Pharmacological inhibition of NOX5 (Melittin, 10-7 M) and NOX5 gene silencing (siRNA) was used to determine the role of NOX5-dependent ROS production in endothelial oxidative stress induced by LPC. ROS production was determined by lucigenin assay and electron paramagnetic spectroscopy (EPR), calcium transients by Fluo4 fluorimetry, and NOX5 activity and protein expression by pharmacological assays and immunoblotting, respectively. RESULTS: LPC increased ROS generation in endothelial cells at short (15 min) and long (4 h) stimulation times. LPC-induced ROS was abolished by a selective NOX5 inhibitor and by NOX5 siRNA. NOX1/4 dual inhibition and selective NOX1 inhibition only decreased ROS generation at 4 h. LPC increased HAEC intracellular calcium, important for NOX5 activation, and this was blocked by nifedipine and thapsigargin. Bapta-AM, selective Ca2+ chelator, prevented LPC-induced ROS production. NOX5 knockdown decreased LPC-induced ICAM-1 mRNA expression and monocyte adhesion to endothelial cells. CONCLUSION: These results suggest that NOX5, by mechanisms linked to increased intracellular calcium, is key to early LPC-induced endothelial oxidative stress and pro-inflammatory processes. Since these are essential events in the formation and progression of atherosclerotic lesions, the present study highlights an important role for NOX5 in atherosclerosis.


Subject(s)
Atherosclerosis/enzymology , Endothelial Cells/drug effects , Lysophosphatidylcholines/toxicity , NADPH Oxidase 5/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Atherosclerosis/pathology , Calcium/metabolism , Calcium Signaling , Cell Adhesion , Cells, Cultured , Coculture Techniques , Endothelial Cells/enzymology , Endothelial Cells/pathology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Monocytes/metabolism , NADPH Oxidase 5/antagonists & inhibitors , NADPH Oxidase 5/genetics , RNA Interference
15.
J Cardiovasc Pharmacol ; 78(5): e749-e760, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34321402

ABSTRACT

ABSTRACT: Atherosclerosis (AS) is a major risk factor for cardiovascular disease, in which circular RNAs play important regulatory roles. This research aimed to explore the biological role of circular RNA Sterol Regulatory Element Binding Transcription Factor Chaperone (circSCAP) (hsa_circ_0001292) in AS development. Real-time PCR or Western blot assay was conducted to analyze RNA or protein expression. Cell proliferation and apoptosis were analyzed by CCK-8 assay and flow cytometry. The levels of lipid accumulation-associated indicators and oxidative stress factors were detected using commercial kits. The levels of inflammatory cytokines were examined using enzyme-linked immunosorbent assay. Intermolecular interaction was verified by dual-luciferase reporter analysis or RNA pull-down analysis. CircSCAP and phosphodiesterase 3B (PDE3B) levels were elevated, whereas the miR-221-5p level was decreased in patients with AS and oxidized low-density lipoprotein (ox-LDL)-induced THP-1 cells. CircSCAP absence suppressed lipid deposition, inflammation, and oxidative stress in ox-LDL-induced THP-1 cells. MiR-221-5p was a target of circSCAP, and anti-miR-221-5p largely reversed si-circSCAP-induced effects in ox-LDL-induced THP-1 cells. PDE3B was a target of miR-221-5p, and PDE3B overexpression largely counteracted miR-221-5p accumulation-mediated effects in ox-LDL-induced THP-1 cells. NF-κB signaling pathway was regulated by circSCAP/miR-221-5p/PDE3B axis in ox-LDL-induced THP-1 cells. In conclusion, circSCAP facilitated lipid accumulation, inflammation, and oxidative stress in ox-LDL-induced THP-1 macrophages by regulating miR-221-5p/PDE3B axis.


Subject(s)
Atherosclerosis/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 3/biosynthesis , Lipoproteins, LDL/toxicity , Macrophages/drug effects , MicroRNAs/metabolism , RNA, Circular/metabolism , Apoptosis/drug effects , Atherosclerosis/genetics , Atherosclerosis/pathology , Case-Control Studies , Cell Proliferation/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Cytokines/metabolism , Enzyme Induction , Female , Humans , Inflammation Mediators/metabolism , Macrophages/enzymology , Macrophages/pathology , Male , MicroRNAs/genetics , Middle Aged , Oxidative Stress/drug effects , RNA, Circular/genetics , Signal Transduction , THP-1 Cells
16.
Aging (Albany NY) ; 13(11): 14892-14909, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34102609

ABSTRACT

BACKGROUND: Recent studies have demonstrated a key role of vascular smooth muscle cell (VSMC) dysfunction in atherosclerosis. Cyclin-dependent kinases 9 (CDK9), a potential biomarker of atherosclerosis, was significantly increased in coronary artery disease patient serum and played an important role in inflammatory diseases. This study was to explore the pharmacological role of CDK9 inhibition in attenuating atherosclerosis. METHODS: A small-molecule CDK9 inhibitor, LDC000067, was utilized to treat the high fat diet (HFD)-fed ApoE-/- mice and human VSMCs. RESULTS: The results showed that inflammation and phenotypic switching of VSMCs were observed in HFD-induced atherosclerosis in ApoE-/- mice, which were accompanied with increased CDK9 in the serum and atherosclerotic lesions where it colocalized with VSMCs. LDC000067 treatment significantly suppressed HFD-induced inflammation, proliferation and phenotypic switching of VSMCs, resulting in reduced atherosclerosis in the ApoE-/- mice, while had no effect on plasma lipids. Further in vitro studies confirmed that LDC000067 and siRNA-mediated CDK9 knockdown reversed ox-LDL-induced inflammation and phenotypic switching of VSMCs from a contractile phenotype to a synthetic phenotype via inhibiting NF-κB signaling pathway in human VSMCs. CONCLUSION: These results indicate that inhibition of CDK9 may be a novel therapeutic target for the prevention of atherosclerosis.


Subject(s)
Atherosclerosis/enzymology , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Inflammation/pathology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Animals , Apolipoproteins E/deficiency , Atherosclerosis/complications , Cyclin-Dependent Kinase 9/metabolism , Diet, High-Fat , Humans , Inflammation/complications , Lipoproteins, LDL , Male , Mice , Myocytes, Smooth Muscle/drug effects , NF-kappa B/metabolism , Phenotype , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/metabolism , Signal Transduction
17.
Int J Mol Sci ; 22(11)2021 May 30.
Article in English | MEDLINE | ID: mdl-34070931

ABSTRACT

It is now about 20 years since the first case of a gain-of-function mutation involving the as-yet-unknown actor in cholesterol homeostasis, proprotein convertase subtilisin/kexin type 9 (PCSK9), was described. It was soon clear that this protein would have been of huge scientific and clinical value as a therapeutic strategy for dyslipidemia and atherosclerosis-associated cardiovascular disease (CVD) management. Indeed, PCSK9 is a serine protease belonging to the proprotein convertase family, mainly produced by the liver, and essential for metabolism of LDL particles by inhibiting LDL receptor (LDLR) recirculation to the cell surface with the consequent upregulation of LDLR-dependent LDL-C levels. Beyond its effects on LDL metabolism, several studies revealed the existence of additional roles of PCSK9 in different stages of atherosclerosis, also for its ability to target other members of the LDLR family. PCSK9 from plasma and vascular cells can contribute to the development of atherosclerotic plaque and thrombosis by promoting platelet activation, leukocyte recruitment and clot formation, also through mechanisms not related to systemic lipid changes. These results further supported the value for the potential cardiovascular benefits of therapies based on PCSK9 inhibition. Actually, the passive immunization with anti-PCSK9 antibodies, evolocumab and alirocumab, is shown to be effective in dramatically reducing the LDL-C levels and attenuating CVD. While monoclonal antibodies sequester circulating PCSK9, inclisiran, a small interfering RNA, is a new drug that inhibits PCSK9 synthesis with the important advantage, compared with PCSK9 mAbs, to preserve its pharmacodynamic effects when administrated every 6 months. Here, we will focus on the major understandings related to PCSK9, from its discovery to its role in lipoprotein metabolism, involvement in atherothrombosis and a brief excursus on approved current therapies used to inhibit its action.


Subject(s)
Atherosclerosis/genetics , Cholesterol, LDL/metabolism , Dyslipidemias/genetics , Plaque, Atherosclerotic/genetics , Proprotein Convertase 9/genetics , Thrombosis/genetics , Antibodies, Monoclonal, Humanized/therapeutic use , Atherosclerosis/drug therapy , Atherosclerosis/enzymology , Atherosclerosis/pathology , Blood Platelets/drug effects , Blood Platelets/enzymology , Blood Platelets/pathology , Cholesterol, LDL/antagonists & inhibitors , Dyslipidemias/drug therapy , Dyslipidemias/enzymology , Dyslipidemias/pathology , Fibrinolytic Agents/therapeutic use , Gene Expression Regulation , Humans , Hypolipidemic Agents/therapeutic use , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , PCSK9 Inhibitors , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/enzymology , Plaque, Atherosclerotic/pathology , Platelet Activation/drug effects , Proprotein Convertase 9/biosynthesis , RNA, Small Interfering/therapeutic use , Receptors, LDL/genetics , Receptors, LDL/metabolism , Signal Transduction , Thrombosis/enzymology , Thrombosis/pathology , Thrombosis/prevention & control
18.
J Cardiovasc Pharmacol ; 77(5): 642-649, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33951699

ABSTRACT

ABSTRACT: Atherosclerosis (AS) is one of the most severe cardiovascular diseases involved in the phenotypic switching of vascular smooth muscle cells (VSMCs). Tryptanthrin is a natural product with broad biological activities. However, the effect of tryptanthrin on atherosclerotic progression is unclear. The aim of this study was to determine the role of tryptanthrin in AS and explore the potential mechanism. In vitro, primary VSMCs were stimulated with platelet-derived growth factor-BB (PDGF) to induce cell dedifferentiation. Treatment with tryptanthrin (5 µM or 10 µM) suppressed the proliferation and recovered the contractility of VSMCs in the presence of PDGF. The contractile proteins (α-smooth muscle actin, calponin, and SM22α) were increased, and the synthetic protein vimentin was decreased by tryptanthrin in PDGF-induced VSMCs. ApoE-/- mice fed with high-fat diet were used as an in vivo model of AS. Similarly, gavage administration of tryptanthrin (50 mg/kg or 100 mg/kg) attenuated VSMC phenotypic changes from a contractile to a synthetic state in aortic tissues of AS mice. The serum lipid level, atherosclerotic plaque formation, and arterial intimal hyperplasia were attenuated by tryptanthrin. Furthermore, tryptanthrin increased the expression levels of phosphorylated AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC) both in vitro and in vivo. Administration of compound C, an AMPK inhibitor, reversed the inhibitory effect of tryptanthrin on VSMC dedifferentiation in vitro. Thus, we demonstrate that tryptanthrin protects against AS progression through the inhibition of VSMC switching from a contractile to a pathological synthetic phenotype by the activation of AMPK/ACC pathway. It provides novel insights into AS prevention and treatment.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Acetyl-CoA Carboxylase/metabolism , Atherosclerosis/drug therapy , Cell Plasticity/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Quinazolines/pharmacology , Animals , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Becaplermin/pharmacology , Cells, Cultured , Disease Models, Animal , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Neointima , Phenotype , Phosphorylation , Plaque, Atherosclerotic , Signal Transduction
20.
PLoS One ; 16(5): e0246600, 2021.
Article in English | MEDLINE | ID: mdl-33983975

ABSTRACT

Atherosclerotic vascular disease resulting from unstable plaques is the leading cause of morbidity and mortality in subjects with type 2 diabetes (T2D), and thus a major therapeutic goal is to discover T2D drugs that can also promote atherosclerotic plaque stability. Genetic or pharmacologic inhibition of mitogen-activated protein kinase-activated protein kinase-2 (MAPKAPK2 or MK2) in obese mice improves glucose homeostasis and enhances insulin sensitivity. We developed two novel orally active small-molecule inhibitors of MK2, TBX-1 and TBX-2, and tested their effects on metabolism and atherosclerosis in high-fat Western diet (WD)-fed Ldlr-/- mice. Ldlr-/- mice were first fed the WD to allow atherosclerotic lesions to become established, and the mice were then treated with TBX-1 or TBX-2. Both compounds improved glucose metabolism and lowered plasma cholesterol and triglyceride, without an effect on body weight. Most importantly, the compounds decreased lesion area, lessened plaque necrosis, and increased fibrous cap thickness in the aortic root lesions of the mice. Thus, in a preclinical model of high-fat feeding and established atherosclerosis, MK2 inhibitors improved metabolism and also enhanced atherosclerotic plaque stability, suggesting potential for further clinical development to address the epidemic of T2D associated with atherosclerotic vascular disease.


Subject(s)
Atherosclerosis/enzymology , Atherosclerosis/pathology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Plaque, Atherosclerotic/enzymology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Allosteric Regulation/drug effects , Animals , Aorta/pathology , Atherosclerosis/blood , Glucose/metabolism , Homeostasis/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Lipids/blood , Male , Mice, Inbred C57BL , Necrosis , Plaque, Atherosclerotic/blood , Plaque, Atherosclerotic/pathology , Protein Kinase Inhibitors/pharmacokinetics , Protein Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...