Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.386
Filter
1.
Atherosclerosis ; 397: 118556, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39222595

ABSTRACT

BACKGROUND AND AIMS: Atherosclerotic cardiovascular disease complicated by diabetes mellitus (DM) is the leading cause of death in diabetic patients, and it is strongly associated with macrophages and inflammasomes. It has been found that activation of NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome is closely associated with phosphatidylinositol 4-phosphate (PI4P) on the trans-Golgi. However, how PI4P and NLRP3 regulate macrophage function and its role in diabetic atherosclerotic plaques is unclear. METHODS: The expression of Pi4p and Nlrp3-inflammasome-related proteins in atherosclerosis in apolipoprotein E-deficient (Apoe-/-) and Apoe-/- DM mice was investigated. Then, Pi4p levels were affected by shRNA-Pi4kb or cDNA-Sac1 plasmid to investigate the effects of changes in Pi4p-related metabolic enzymes on macrophage function. Finally, genetically modified macrophages were injected into diabetic Apoe-/- mice to explore the effects on atherosclerosis. RESULTS: DM promoted plaque progression in atherosclerotic mice and increased expression of Pi4p and Nlrp3 in plaques. In addition, impaired macrophage function induced by high glucose was reversed by transfected shRNA-Pi4kb or cDNA-Sac1 plasmid. Furthermore, decreased levels of Pi4p reduced plaque area in diabetic Apoe-/- mice. CONCLUSIONS: Our data suggests that Pi4p/Nlrp3 in macrophages play an important role in the exacerbation of atherosclerosis in diabetic mice. Pi4p-related metabolizing enzymes (PI4KB and SAC1) may be a potential therapeutic strategy for diabetic atherosclerosis, and macrophage therapy is also a potential treatment.


Subject(s)
Atherosclerosis , Diabetes Mellitus, Experimental , Disease Progression , Macrophages , Mice, Knockout, ApoE , NLR Family, Pyrin Domain-Containing 3 Protein , Plaque, Atherosclerotic , Signal Transduction , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Macrophages/metabolism , Atherosclerosis/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , Diabetes Mellitus, Experimental/metabolism , Mice , Mice, Inbred C57BL , Male , Inflammasomes/metabolism , Apolipoproteins E/genetics , Apolipoproteins E/deficiency
2.
Cell Death Dis ; 15(9): 689, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39304654

ABSTRACT

Recent research has explored the potential of the demethylating drug 5-azacytidine (Aza) as therapy for a range of diseases. However, the therapeutic efficacy of Aza for patients of atherosclerosis remains unclear. This study investigates the therapeutic application of Aza to atherosclerosis in order to elucidate the underlying mechanisms. We generated induced Tregs (iTregs) from CD4+ T cells by using Aza in vitro, and this was followed by the intravenous infusion of iTregs for the treatment of atherosclerosis. The adoptive transfer of Aza-iTreg significantly increased peripheral blood Treg cells, suppressed inflammation, and attenuated atherosclerosis in ApoE-/- mice. Furthermore, we observed a notable demethylation of the Forkhead box P3 (Foxp3)-regulatory T cell-specific demethylated region (TSDR) and an upregulation of Foxp3 expression in the CD4+ T cells in the spleen of the ApoE-/- mice following the transfer of Aza- iTregs. We also demonstrated that Aza converted naive CD4+ T cells into Tregs by DNA methyltransferase 1 (Dnmt1)-mediated Foxp3-TSDR demethylation and the upregulation of Foxp3 expression. Conversely, the overexpression of Dnmt1 in the CD4+ T cells attenuated the Aza-induced Foxp3-TSDR demethylation and upregulation of Foxp3 expression. Our results reveal that Aza converts naive CD4+ T cells into functional Tregs by inhibiting Dnmt1, and the transfer of Aza-iTregs suppresses atherosclerosis in mice.


Subject(s)
Atherosclerosis , Azacitidine , CD4-Positive T-Lymphocytes , Epigenesis, Genetic , Forkhead Transcription Factors , T-Lymphocytes, Regulatory , Animals , Atherosclerosis/drug therapy , Atherosclerosis/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Azacitidine/pharmacology , Mice , Forkhead Transcription Factors/metabolism , Epigenesis, Genetic/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/drug effects , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Mice, Inbred C57BL , Apolipoproteins E/metabolism , Apolipoproteins E/genetics , Male , Humans
3.
Sci Rep ; 14(1): 21371, 2024 09 12.
Article in English | MEDLINE | ID: mdl-39266573

ABSTRACT

Atherosclerosis (AS) is a major cause of cardiovascular diseases that may lead to mortality. This study aimed to evaluate the therapeutic potential of tetrandrine in high cholesterol diet (HCD)-induced atherosclerosis, in rats, via modulation of miR-34a, as well as, Wnt5a/Ror2/ABCA1/NF-κB pathway and to compare its efficacy with atorvastatin. Induction of AS, in male rats, was done via IP administration of vitamin D3 (70 U/Kg for 3 days) together with HCD. At the end of the 9th week, rats were treated with atorvastatin at a dose of 20 mg/kg, and tetrandrine at different doses of (18.75, and 31.25 mg/kg) for 22 days. Serum inflammatory cytokines and lipid profile, liver oxidative stress parameters, and aortic tissue Wnt5a, Ror2, ABCA1, NF-κB, miR-34a levels were assessed in all experimental groups. Histopathological and Immunohistochemical assessments of aortic tissue sections were done. Results showed that tetrandrine treatment reverted the inflammatory and oxidative stress state together with reducing the serum lipids via modulating miR-34a, and Wnt5a/Ror2/ABCA1/NF-κB pathway. Moreover, it reverted the histopathological abnormalities observed in AS rats. Tetrandrine beneficial effects, in both doses, were comparable to that of atorvastatin, in most of the discussed parameters. These findings praise tetrandrine as a promising agent for management of atherosclerosis.


Subject(s)
ATP Binding Cassette Transporter 1 , Atherosclerosis , Benzylisoquinolines , MicroRNAs , NF-kappa B , Wnt-5a Protein , Animals , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Atherosclerosis/pathology , Wnt-5a Protein/metabolism , Rats , NF-kappa B/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Male , Benzylisoquinolines/pharmacology , Benzylisoquinolines/therapeutic use , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter 1/genetics , Oxidative Stress/drug effects , Cholecalciferol/pharmacology , Signal Transduction/drug effects , Rats, Wistar , Diet, High-Fat/adverse effects , Cholesterol, Dietary/adverse effects
4.
Int J Mol Sci ; 25(17)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39273324

ABSTRACT

Several lines of evidence have linked the intestinal bacterium Helicobacter cinaedi with the pathogenesis of atherosclerosis, identifying the Cinaedi Antigen Inflammatory Protein (CAIP) as a key virulence factor. Oxidative stress and inflammation are crucial in sustaining the atherosclerotic process and oxidized LDL (oxLDL) uptake. Primary human macrophages and endothelial cells were pre-incubated with 10 µM diphenyl iodonium salt (DPI) and stimulated with 20 µg/mL CAIP. Lectin-like oxLDL receptor (LOX-1) expression was evaluated by FACS analysis, reactive oxygen species (ROS) production was measured using the fluorescent probe H2DCF-DA, and cytokine release was quantified by ELISA assay. Foam cells formation was assessed by Oil Red-O staining, and phosphorylation of p38 and ERK1/2 MAP kinases and NF-κB pathway activation were determined by Western blot. This study demonstrated that CAIP triggered LOX-1 over-expression and increased ROS production in both macrophages and endothelial cells. Blocking ROS abrogated LOX-1 expression and reduced LDL uptake and foam cells formation. Additionally, CAIP-mediated pro-inflammatory cytokine release was significantly affected by ROS inhibition. The signaling pathway induced by CAIP-induced oxidative stress led to p38 MAP kinase phosphorylation and NF-κB activation. These findings elucidate the mechanism of action of CAIP, which heightens oxidative stress and contributes to the atherosclerotic process in H. cinaedi-infected patients.


Subject(s)
Atherosclerosis , Helicobacter Infections , Helicobacter , Lipoproteins, LDL , Macrophages , Reactive Oxygen Species , Scavenger Receptors, Class E , Humans , Reactive Oxygen Species/metabolism , Atherosclerosis/metabolism , Atherosclerosis/microbiology , Atherosclerosis/pathology , Macrophages/metabolism , Macrophages/microbiology , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Scavenger Receptors, Class E/metabolism , Lipoproteins, LDL/metabolism , Helicobacter/pathogenicity , Endothelial Cells/metabolism , Endothelial Cells/microbiology , NF-kappa B/metabolism , Foam Cells/metabolism , Cytokines/metabolism , Oxidative Stress , p38 Mitogen-Activated Protein Kinases/metabolism , Bacterial Proteins/metabolism , MAP Kinase Signaling System , Cells, Cultured , Signal Transduction
5.
Int J Mol Sci ; 25(17)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39273484

ABSTRACT

Hypercholesterolemia forms the background of several cardiovascular pathologies. LDL receptor-knockout (LDLR-KO) mice kept on a high-fat diet (HFD) develop high cholesterol levels and atherosclerosis (AS). Cannabinoid type 1 receptors (CB1Rs) induce vasodilation, although their role in cardiovascular pathologies is still controversial. We aimed to reveal the effects of CB1Rs on vascular function and remodeling in hypercholesterolemic AS-prone LDLR-KO mice. Experiments were performed on a newly established LDLR and CB1R double-knockout (KO) mouse model, in which KO and wild-type (WT) mice were kept on an HFD or a control diet (CD) for 5 months. The vascular functions of abdominal aorta rings were tested with wire myography. The vasorelaxation effects of acetylcholine (Ach, 1 nM-1 µM) were obtained after phenylephrine precontraction, which was repeated with inhibitors of nitric oxide synthase (NOS) and cyclooxygenase (COX), Nω-nitro-L-arginine (LNA), and indomethacin (INDO), respectively. Blood pressure was measured with the tail-cuff method. Immunostaining of endothelial NOS (eNOS) was carried out. An HFD significantly elevated the cholesterol levels in the LDLR-KO mice more than in the corresponding WT mice (mean values: 1039 ± 162 mg/dL vs. 91 ± 18 mg/dL), and they were not influenced by the presence of the CB1R gene. However, with the defect of the CB1R gene, damage to the Ach relaxation ability was moderated. The blood pressure was higher in the LDLR-KO mice compared to their WT counterparts (systolic/diastolic values: 110/84 ± 5.8/6.8 vs. 102/80 ± 3.3/2.5 mmHg), which was significantly elevated with an HFD (118/96 ± 1.9/2 vs. 100/77 ± 3.4/3.1 mmHg, p < 0.05) but attenuated in the CB1R-KO HFD mice. The expression of eNOS was depressed in the HFD WT mice compared to those on the CD, but it was augmented if CB1R was knocked out. This newly established double-knockout mouse model provides a tool for studying the involvement of CB1Rs in the development of hypercholesterolemia and atherosclerosis. Our results indicate that knocking out the CB1R gene significantly attenuates vascular damage in hypercholesterolemic mice.


Subject(s)
Disease Models, Animal , Hypercholesterolemia , Mice, Knockout , Receptor, Cannabinoid, CB1 , Receptors, LDL , Vasodilation , Animals , Hypercholesterolemia/metabolism , Hypercholesterolemia/genetics , Hypercholesterolemia/pathology , Mice , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB1/genetics , Receptors, LDL/genetics , Receptors, LDL/metabolism , Receptors, LDL/deficiency , Vasodilation/drug effects , Diet, High-Fat/adverse effects , Male , Nitric Oxide Synthase Type III/metabolism , Atherosclerosis/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/etiology , Vascular Remodeling/drug effects , Mice, Inbred C57BL , Acetylcholine/pharmacology
6.
Cardiovasc Diabetol ; 23(1): 331, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39252021

ABSTRACT

BACKGROUND: Visceral adipose tissue in individuals with obesity is an independent cardiovascular risk indicator. However, it remains unclear whether adipose tissue influences common cardiovascular diseases, such as atherosclerosis, through its secreted exosomes. METHODS: The exosomes secreted by adipose tissue from diet-induced obesity mice were isolated to examine their impact on the progression of atherosclerosis and the associated mechanism. Endothelial apoptosis and the proliferation and migration of vascular smooth muscle cells (VSMCs) within the atherosclerotic plaque were evaluated. Statistical significance was analyzed using GraphPad Prism 9.0 with appropriate statistical tests. RESULTS: We demonstrate that adipose tissue-derived exosomes (AT-EX) exacerbate atherosclerosis progression by promoting endothelial apoptosis, proliferation, and migration of VSMCs within the plaque in vivo. MicroRNA-132/212 (miR-132/212) was detected within AT-EX cargo. Mechanistically, miR-132/212-enriched AT-EX exacerbates palmitate acid-induced endothelial apoptosis via targeting G protein subunit alpha 12 and enhances platelet-derived growth factor type BB-induced VSMC proliferation and migration by targeting phosphatase and tensin homolog in vitro. Importantly, melatonin decreases exosomal miR-132/212 levels, thereby mitigating the pro-atherosclerotic impact of AT-EX. CONCLUSION: These data uncover the pathological mechanism by which adipose tissue-derived exosomes regulate the progression of atherosclerosis and identify miR-132/212 as potential diagnostic and therapeutic targets for atherosclerosis.


Subject(s)
Apoptosis , Atherosclerosis , Cell Movement , Cell Proliferation , Disease Models, Animal , Disease Progression , Exosomes , Mice, Inbred C57BL , MicroRNAs , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Plaque, Atherosclerotic , Animals , MicroRNAs/metabolism , MicroRNAs/genetics , Exosomes/metabolism , Exosomes/pathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/genetics , Cell Proliferation/drug effects , Apoptosis/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/drug effects , Cell Movement/drug effects , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , Male , Signal Transduction , Cells, Cultured , Obesity/metabolism , Obesity/pathology , Mice, Knockout, ApoE , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelial Cells/drug effects , Aortic Diseases/pathology , Aortic Diseases/metabolism , Aortic Diseases/genetics , Becaplermin/pharmacology , Becaplermin/metabolism , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Mice , Humans
7.
Sci Signal ; 17(855): eadt2301, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39316664

ABSTRACT

Early exposure to cholesterol primes later atherosclerosis through phenotypic changes to resident-like arterial macrophages.


Subject(s)
Atherosclerosis , Macrophages , Plaque, Atherosclerotic , Humans , Plaque, Atherosclerotic/pathology , Plaque, Atherosclerotic/metabolism , Macrophages/metabolism , Macrophages/cytology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Animals , Cholesterol/metabolism
8.
ACS Appl Mater Interfaces ; 16(37): 49660-49672, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39240784

ABSTRACT

Atherosclerosis is a persistent inflammatory condition of the blood vessels associated with abnormalities in lipid metabolism. Development of biomimetic nanoplatforms provides an effective strategy. Herein, inspired by the peptide CLIKKPF spontaneously coupling to phosphatidylserine (PS) on the inner leaflet of cell membranes specifically, MM@NPs were constructed by macrophage membrane spontaneous encapsulation of cyclodextrin-based nanoparticles modified with the peptide CLIKKPF and loaded with the hydrophobic compound resveratrol. MM@NPs could be specifically phagocytized by the activated endothelium with the overexpressed VCAM-1 for enhancing target delivery into the pathological lesion. Additionally, for the ApoE-/- mice, MM@NPs provide comprehensive treatment efficiency in reducing oxidant stress, alleviating the inherent inflammation, and decreasing cholesterol deposition, subsequently resulting in the atherosclerotic plaque regression. Therefore, MM@NPs could be one possible candidate for improving lipid metabolism and inflammation in atherosclerosis.


Subject(s)
Atherosclerosis , Cyclodextrins , Inflammation , Lipid Metabolism , Macrophages , Nanoparticles , Animals , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Atherosclerosis/pathology , Mice , Macrophages/metabolism , Macrophages/drug effects , Cyclodextrins/chemistry , Cyclodextrins/pharmacology , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Lipid Metabolism/drug effects , Nanoparticles/chemistry , RAW 264.7 Cells , Resveratrol/chemistry , Resveratrol/pharmacology , Nanomedicine , Cell Membrane/metabolism , Cell Membrane/drug effects , Humans
9.
Atherosclerosis ; 397: 118567, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39243663

ABSTRACT

BACKGROUND AND AIMS: Mast cell-derived heparin proteoglycans (HEP-PG) can be mimicked by bioconjugates carrying antithrombotic and anti-inflammatory properties. The dual antiplatelet and anticoagulant (APAC) construct administered, either locally or intravenously (i.v.), targets activated endothelium, its adhesion molecules, and subendothelial matrix proteins, all relevant to atherogenesis. We hypothesized that APAC influences cellular interactions in atherosclerotic lesion development and studied APAC treatment during the initiation and progression of experimental atherosclerosis. METHODS: Male western-type diet-fed Apoe-/- mice were equipped with perivascular carotid artery collars to induce local atherosclerosis. In this model, mRNA expression of adhesion molecules including ICAM-1, VCAM-1, P-Selectin, and Platelet Factor 4 (PF4) are upregulated upon lesion development. From day 1 (prevention) or from 2.5 weeks after lesion initiation (treatment), mice were administered 0.2 mg/kg APAC i.v. or control vehicle three times weekly for 2.5 weeks. At week 5 after collar placement, mice were sacrificed, and lesion morphology was microscopically assessed. RESULTS: APAC treatment did not affect body weight or plasma total cholesterol levels during the experiments. In the prevention setting, APAC reduced carotid artery plaque size and volume by over 50 %, aligning with decreased plaque macrophage area and collagen content. During the treatment setting, APAC reduced macrophage accumulation and necrotic core content, and improved markers of plaque stability. CONCLUSIONS: APAC effectively reduced early atherosclerotic lesion development and improved markers of plaque inflammation in advanced atherosclerosis. Thus, APAC may have potential to alleviate the progression of atherosclerosis.


Subject(s)
Anticoagulants , Carotid Artery Diseases , Disease Models, Animal , Mice, Knockout, ApoE , Plaque, Atherosclerotic , Platelet Aggregation Inhibitors , Animals , Male , Carotid Artery Diseases/prevention & control , Carotid Artery Diseases/pathology , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/drug therapy , Anticoagulants/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Mice , Proteoglycans , Carotid Arteries/pathology , Carotid Arteries/drug effects , Mice, Inbred C57BL , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Atherosclerosis/metabolism , Apolipoproteins E/genetics , Platelet Factor 4 , Vascular Cell Adhesion Molecule-1/metabolism , Heparin/analogs & derivatives
10.
FASEB J ; 38(18): e70069, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39315853

ABSTRACT

Coronary plaque rupture remains the prominent mechanism of myocardial infarction. Accurate identification of rupture-prone plaque may improve clinical management. This study assessed the discriminatory performance of electrochemical impedance spectroscopy (EIS) in human cardiac explants to detect high-risk atherosclerotic features that portend rupture risk. In this single-center, prospective study, n = 26 cardiac explants were collected for EIS interrogation of the three major coronary arteries. Vessels in which advancement of the EIS catheter without iatrogenic plaque disruption was rendered impossible were not assessed. N = 61 vessels underwent EIS measurement and histological analyses. Plaques were dichotomized according to previously established high rupture-risk parameter thresholds. Diagnostic performance was determined via receiver operating characteristic areas-under-the-curve (AUC). Necrotic cores were identified in n = 19 vessels (median area 1.53 mm2) with a median fibrous cap thickness of 62 µm. Impedance was significantly greater in plaques with necrotic core area ≥1.75 mm2 versus <1.75 mm2 (19.8 ± 4.4 kΩ vs. 7.2 ± 1.0 kΩ, p = .019), fibrous cap thickness ≤65 µm versus >65 µm (19.1 ± 3.5 kΩ vs. 6.5 ± 0.9 kΩ, p = .004), and ≥20 macrophages per 0.3 mm-diameter high-power field (HPF) versus <20 macrophages per HPF (19.8 ± 4.1 kΩ vs. 10.2 ± 0.9 kΩ, p = .002). Impedance identified necrotic core area ≥1.75 mm2, fibrous cap thickness ≤65 µm, and ≥20 macrophages per HPF with AUCs of 0.889 (95% CI: 0.716-1.000) (p = .013), 0.852 (0.646-1.000) (p = .025), and 0.835 (0.577-1.000) (p = .028), respectively. Further, phase delay discriminated severe stenosis (≥70%) with an AUC of 0.767 (0.573-0.962) (p = .035). EIS discriminates high-risk atherosclerotic features that portend plaque rupture in human coronary artery disease and may serve as a complementary modality for angiography-guided atherosclerosis evaluation.


Subject(s)
Coronary Artery Disease , Coronary Vessels , Dielectric Spectroscopy , Plaque, Atherosclerotic , Humans , Coronary Artery Disease/pathology , Dielectric Spectroscopy/methods , Male , Female , Plaque, Atherosclerotic/pathology , Plaque, Atherosclerotic/diagnostic imaging , Middle Aged , Prospective Studies , Aged , Coronary Vessels/pathology , Atherosclerosis/pathology , Risk Factors
11.
Atherosclerosis ; 397: 118582, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39260002

ABSTRACT

BACKGROUND AND AIMS: Lysyl oxidase (LOX) catalyzes the crosslinking of collagen and elastin to maintain tensile strength and structural integrity of the vasculature. Excessive LOX activity increases vascular stiffness and the severity of occlusive diseases. Herein, we investigated the mechanisms by which LOX controls atherogenesis and osteogenic differentiation of vascular smooth muscle cells (SMC) in hyperlipidemic mice. METHODS: Gene inactivation of Lox in SMC was achieved in conditional knockout mice after tamoxifen injections. Atherosclerosis burden and vascular calcification were assessed in hyperlipidemic conditional [Loxf/fMyh11-CreERT2ApoE-/-] and sibling control mice [Loxwt/wtMyh11-CreERT2ApoE-/-]. Mechanistic studies were performed with primary aortic SMC from Lox mutant and wild type mice. RESULTS: Inactivation of Lox in SMCs decreased > 70 % its RNA expression and protein level in the aortic wall and significantly reduced LOX activity without compromising vascular structure and function. Moreover, LOX deficiency protected mice against atherosclerotic burden (13 ± 2 versus 23 ± 1 %, p < 0.01) and plaque calcification (5 ± 0.4 versus 11.8 ± 3 %, p < 0.05) compared to sibling controls. Interestingly, gene inactivation of Lox in SMCs preserved the contractile phenotype of vascular SMC under hyperlipidemic conditions as demonstrated by single-cell RNA sequencing and immunofluorescence. Mechanistically, the absence of LOX in SMC prevented excessive collagen crosslinking and the subsequent activation of the pro-osteogenic FAK/ß-catenin signaling axis. CONCLUSIONS: Lox inactivation in SMC protects mice against atherosclerosis and plaque calcification by reducing SMC modulation and FAK/ß-catenin signaling.


Subject(s)
Atherosclerosis , Disease Models, Animal , Hyperlipidemias , Mice, Knockout , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Plaque, Atherosclerotic , Protein-Lysine 6-Oxidase , Vascular Calcification , Animals , Protein-Lysine 6-Oxidase/metabolism , Protein-Lysine 6-Oxidase/genetics , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/metabolism , Atherosclerosis/genetics , Atherosclerosis/enzymology , Atherosclerosis/pathology , Atherosclerosis/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/metabolism , Vascular Calcification/genetics , Vascular Calcification/pathology , Vascular Calcification/enzymology , Vascular Calcification/prevention & control , Vascular Calcification/metabolism , Hyperlipidemias/genetics , Hyperlipidemias/enzymology , Hyperlipidemias/complications , Hyperlipidemias/metabolism , Mice , Osteogenesis , Cells, Cultured , Aortic Diseases/pathology , Aortic Diseases/genetics , Aortic Diseases/enzymology , Aortic Diseases/prevention & control , Aortic Diseases/metabolism , Aorta/pathology , Aorta/enzymology , Aorta/metabolism , Male , Mice, Inbred C57BL , beta Catenin/metabolism , Signal Transduction , Extracellular Matrix Proteins
12.
Atherosclerosis ; 397: 118570, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39276419

ABSTRACT

BACKGROUND AND AIMS: CCN4/WISP-1 regulates various cell behaviours that contribute to atherosclerosis progression, including cell adhesion, migration, proliferation and survival. We therefore hypothesised that CCN4 regulates the development and progression of atherosclerotic plaques. METHODS: We used a high fat fed ApoE-/- mouse model to study atherosclerotic plaque progression in the brachiocephalic artery and aortic root. In protocol 1, male ApoE-/- mice with established plaques were given a CCN4 helper-dependent adenovirus to see the effect of treatment with CCN4, while in protocol 2 male CCN4-/-ApoE-/- were compared to CCN4+/+ApoE-/- mice to assess the effect of CCN4 deletion on plaque progression. RESULTS: CCN4 overexpression resulted in reduced occlusion of the brachiocephalic artery with less apoptosis, fewer macrophages, and attenuated lipid core size. The amount of plaque found on the aortic root was also reduced. CCN4 deficiency resulted in increased apoptosis and occlusion of the brachiocephalic artery as well as increased plaque in the aortic root. Additionally, in vitro cells from CCN4-/-ApoE-/- mice had higher apoptotic levels. CCN4 deficiency did not significantly affect blood cholesterol levels or circulating myeloid cell populations. CONCLUSIONS: We conclude that in an atherosclerosis model the most important action of CCN4 is the effect on cell apoptosis. CCN4 provides pro-survival signals and leads to reduced cell death, lower macrophage number, smaller lipid core size and reduced atherosclerotic plaque burden. As such, the pro-survival effect of CCN4 is worthy of further investigation, in a bid to find a therapeutic for atherosclerosis.


Subject(s)
Apoptosis , Atherosclerosis , Disease Models, Animal , Mice, Knockout, ApoE , Plaque, Atherosclerotic , Animals , Male , Atherosclerosis/pathology , Atherosclerosis/metabolism , Atherosclerosis/genetics , CCN Intercellular Signaling Proteins/metabolism , CCN Intercellular Signaling Proteins/genetics , Mice, Inbred C57BL , Brachiocephalic Trunk/pathology , Brachiocephalic Trunk/metabolism , Mice , Macrophages/metabolism , Disease Progression , Diet, High-Fat , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Aorta/pathology , Aorta/metabolism
13.
Nat Commun ; 15(1): 8034, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39271657

ABSTRACT

Atherosclerosis is an inflammatory disorder responsible for cardiovascular disease. Reactivation of efferocytosis, the phagocytic removal of cells by macrophages, has emerged as a translational target for atherosclerosis. Systemic blockade of the key 'don't-eat-me' molecule, CD47, triggers the engulfment of apoptotic vascular tissue and potently reduces plaque burden. However, it also induces red blood cell clearance, leading to anemia. To overcome this, we previously developed a macrophage-specific nanotherapy loaded with a chemical inhibitor that promotes efferocytosis. Because it was found to be safe and effective in murine studies, we aimed to advance our nanoparticle into a porcine model of atherosclerosis. Here, we demonstrate that production can be scaled without impairing nanoparticle function. At an early stage of disease, we find our nanotherapy reduces apoptotic cell accumulation and inflammation in the atherosclerotic lesion. Notably, this therapy does not induce anemia, highlighting the translational potential of targeted macrophage checkpoint inhibitors.


Subject(s)
Anemia , Atherosclerosis , CD47 Antigen , Disease Models, Animal , Inflammation , Macrophages , Nanoparticles , Phagocytosis , Animals , Atherosclerosis/drug therapy , Atherosclerosis/pathology , Macrophages/drug effects , Macrophages/metabolism , Nanoparticles/chemistry , CD47 Antigen/metabolism , CD47 Antigen/antagonists & inhibitors , Swine , Inflammation/pathology , Phagocytosis/drug effects , Apoptosis/drug effects , Humans , Plaque, Atherosclerotic/pathology , Mice , Male
14.
Adv Exp Med Biol ; 1460: 575-594, 2024.
Article in English | MEDLINE | ID: mdl-39287865

ABSTRACT

Lipotoxicity can mediate endothelial dysfunction in obesity. Altered endothelial cell phenotype during the pathobiological course of the lipotoxicity may lead to hemostatic abnormalities, which is a hallmark of several hematological disorders. Impaired hemostasis could also be directly related to numerous metabolic diseases such as hypertension, diabetes, and atherosclerosis. On the other hand, the local hematopoietic bone marrow (BM) renin-angiotensin system (RAS) contributes to the development of atherosclerosis via acting on the lipotoxicity processes. Local BM RAS, principally an autocrine/paracrine/intracrine hematological system, is located at the crossroads of cellular regulation, molecular interactions, and lipotoxicity-mediated vascular endothelial dysfunction. The positive regulatory role of plasma LDL on AT1 receptor-mediated hematopoietic stem cell (HSC) differentiation and the production of pro-atherogenic monocytes have been described. LDL-regulated HSC function may explain in part hypercholesterolemia-induced inflammation as well as the anti-inflammatory and anti-atherosclerotic effects of AT1 receptor blockers. The role of local adipose tissue RAS is directly related to the pathogenesis of metabolic derangements in obesity. There may be a crosstalk between local BM RAS and local adipose tissue RAS at the genomics and transcriptomics levels. This chapter aims to review hematological alterations propagating the pathological influences of lipotoxicity on the vascular endothelium.


Subject(s)
Hematologic Diseases , Obesity , Renin-Angiotensin System , Humans , Obesity/metabolism , Obesity/complications , Hematologic Diseases/metabolism , Hematologic Diseases/pathology , Hematologic Diseases/etiology , Renin-Angiotensin System/physiology , Animals , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/etiology
15.
Ann Med ; 56(1): 2407525, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39316662

ABSTRACT

INTRODUCTION: Atherosclerosis (AS) is a chronic inflammatory disease characterized by lipid accumulation, inflammation and apoptosis of the arterial wall. This study evaluated the effects of lysophosphatidylinositol (LPI) on endothelial cells activation and autophagy in AS. METHODS: qRT-PCR and Western blotting were done to verify the expression of ICAM1, GPR55 and SOD2. RNA-Seq was performed and screened for the different expressions of long noncoding RNAs (lncRNAs), combining bioinformatics analysis to elucidate the mechanism by which lncRNA functions. RESULTS: qRT-PCR and Western blotting results showed that LPI increased GPR55 and ICAM1 expression. RNA-Seq analysis and qRT-PCR results showed that LPI increased the expression of LINC01235, LINC00520 and LINC01963; LINC01235 was the most obvious. Mechanistically, bioinformatic analysis demonstrated that LINC01235 inhibited autophagy through sponging miR-224-3p. And miRNA-224-3p targeted RABEP1. CONCLUSIONS: LPI promoted endothelial cell activation. LPI induced the expression of LINC01235 and LINC01235 inhibited autophagy through miR-224-3p/RABEP1. Collectively, this study first reveals the function of LINC01235, which may serve as a potential therapeutic target in AS.


Subject(s)
Atherosclerosis , Autophagy , Lysophospholipids , MicroRNAs , RNA, Long Noncoding , Receptors, Cannabinoid , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Autophagy/drug effects , Autophagy/genetics , Humans , Atherosclerosis/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , Receptors, Cannabinoid/metabolism , Receptors, Cannabinoid/genetics , MicroRNAs/metabolism , MicroRNAs/genetics , Lysophospholipids/metabolism , Lysophospholipids/pharmacology , Endothelial Cells/metabolism , Intercellular Adhesion Molecule-1/metabolism , Intercellular Adhesion Molecule-1/genetics , Human Umbilical Vein Endothelial Cells/metabolism
16.
J Biosci ; 492024.
Article in English | MEDLINE | ID: mdl-39234946

ABSTRACT

Atherosclerosis is a common cardiovascular disease in which the arteries are thickened due to buildup of plaque. This study aims to identify programmed cell death (PCD)-related biomarkers and explore the crucial regulatory mechanisms of atherosclerosis. Gene expression profiles of atherosclerosis and control groups from GSE20129 and GSE23746 were obtained. Necroptosis was elevated in atherosclerosis. Weighted gene coexpression network analysis (WGCNA) was conducted in GSE23746 and GSE56045 to identify PCD-related modules and to perform enrichment analysis. Two necroptosis-related genes (IRF9 and STAT1) were identified and considered as biomarkers. Enrichment analysis showed that these gene modules were mainly related to immune response regulation. In addition, single-cell RNA sequencing data from GSE159677 were obtained and the characteristic cell types of atherosclerosis were identified. A total of 11 immune cell types were identified through UMAP dimension reduction. Most immune cells were mainly enriched in plaque samples, and STAT1 and IRF9 were primarily expressed in T-cells and macrophages. Moreover, the roles of IRF9 and STAT1 were assessed and found to be significantly upregulated in atherosclerosis, which was associated with increased risk of atherosclerosis. This study provides a molecular feature of atherosclerosis, offering an important basis for further research on its pathological mechanisms and the search for new therapeutic targets.


Subject(s)
Atherosclerosis , Biomarkers , Interferon-Stimulated Gene Factor 3, gamma Subunit , STAT1 Transcription Factor , T-Lymphocytes , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Humans , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Biomarkers/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Gene Regulatory Networks , Gene Expression Profiling , Gene Expression Regulation , Macrophages/immunology , Macrophages/metabolism , Transcriptome/genetics , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/pathology , Plaque, Atherosclerotic/immunology
17.
Lipids Health Dis ; 23(1): 279, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39227809

ABSTRACT

BACKGROUND: NOD-like receptor protein 3 (NLRP3) inflammasome activation is indispensable for atherogenesis. Mitophagy has emerged as a potential strategy to counteract NLRP3 inflammasome activation triggered by impaired mitochondria. Our previous research has indicated that dihydromyricetin, a natural flavonoid, can mitigate NLRP3-mediated endothelial inflammation, suggesting its potential to treat atherosclerosis. However, the precise underlying mechanisms remain elusive. This study sought to investigate whether dihydromyricetin modulates endothelial mitophagy and inhibits NLRP3 inflammasome activation to alleviate atherogenesis, along with the specific mechanisms involved. METHODS: Apolipoprotein E-deficient mice on a high-fat diet were administered daily oral gavages of dihydromyricetin for 14 weeks. Blood samples were procured to determine the serum lipid profiles and quantify proinflammatory cytokine concentrations. Aortas were harvested to evaluate atherosclerotic plaque formation and NLRP3 inflammasome activation. Concurrently, in human umbilical vein endothelial cells, Western blotting, flow cytometry, and quantitative real-time PCR were employed to elucidate the mechanistic role of mitophagy in the modulation of NLRP3 inflammasome activation by dihydromyricetin. RESULTS: Dihydromyricetin administration significantly attenuated NLRP3 inflammasome activation and vascular inflammation in mice on a high-fat diet, thereby exerting a pronounced inhibitory effect on atherogenesis. Both in vivo and in vitro, dihydromyricetin treatment markedly enhanced mitophagy. This enhancement in mitophagy ameliorated the mitochondrial damage instigated by saturated fatty acids, thereby inhibiting the activation and nuclear translocation of NF-κB. Consequently, concomitant reductions in the transcript levels of NLRP3 and interleukin-1ß (IL-1ß), alongside decreased activation of NLRP3 inflammasome and IL-1ß secretion, were discerned. Notably, the inhibitory effects of dihydromyricetin on the activation of NF-κB and subsequently the NLRP3 inflammasome were determined to be, at least in part, contingent upon its capacity to promote mitophagy. CONCLUSION: This study suggested that dihydromyricetin may function as a modulator to promote mitophagy, which in turn mitigates NF-κB activity and subsequent NLRP3 inflammasome activation, thereby conferring protection against atherosclerosis.


Subject(s)
Atherosclerosis , Diet, High-Fat , Flavonols , Human Umbilical Vein Endothelial Cells , Inflammasomes , Mitophagy , NLR Family, Pyrin Domain-Containing 3 Protein , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mitophagy/drug effects , Animals , Flavonols/pharmacology , Atherosclerosis/drug therapy , Atherosclerosis/prevention & control , Atherosclerosis/pathology , Atherosclerosis/metabolism , Inflammasomes/metabolism , Inflammasomes/drug effects , Mice , Humans , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Diet, High-Fat/adverse effects , Male , Mice, Inbred C57BL , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Mitochondria/drug effects , Mitochondria/metabolism
18.
Mol Med ; 30(1): 137, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39227813

ABSTRACT

N6-methyladenosine (m6A) modification stands out among various RNA modifications as the predominant form within eukaryotic cells, influencing numerous cellular processes implicated in disease development. m6A modification has gained increasing attention in the development of atherosclerosis and has become a research hotspot in recent years. Programmed cell death (PCD), encompassing apoptosis, autophagy, pyroptosis, ferroptosis, and necroptosis, plays a pivotal role in atherosclerosis pathogenesis. In this review, we delve into the intricate interplay between m6A modification and diverse PCD pathways, shedding light on their complex association during the onset and progression of atherosclerosis. Clarifying the relationship between m6A and PCD in atherosclerosis is of great significance to provide novel strategies for cardiovascular disease treatment.


Subject(s)
Adenosine , Apoptosis , Atherosclerosis , Humans , Adenosine/analogs & derivatives , Adenosine/metabolism , Atherosclerosis/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , Animals , Apoptosis/genetics , Autophagy/genetics , RNA/genetics , RNA/metabolism
19.
J Cell Mol Med ; 28(17): e70064, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39248527

ABSTRACT

Cathepsin B (CTSB) is a member of the cysteine protease family, primarily responsible for degrading unnecessary organelles and proteins within the acidic milieu of lysosomes to facilitate recycling. Recent research has revealed that CTSB plays a multifaceted role beyond its function as a proteolytic enzyme in lysosomes. Importantly, recent data suggest that CTSB has significant impacts on different cardiac pathological conditions, such as atherosclerosis (AS), myocardial infarction, hypertension, heart failure and cardiomyopathy. Especially in the context of AS, preclinical models and clinical sample imaging data indicate that the cathepsin activity-based probe can reliably image CTSB activity in foam cells and atherosclerotic plaques; concurrently, it allows synchronous diagnostic and therapeutic interventions. However, our knowledge of CTSB in cardiovascular disease is still in the early stage. This paper aims to provide a comprehensive review of the significance of CTSB in cardiovascular physiology and pathology, with the objective of laying a theoretical groundwork for the development of drugs targeting CTSB.


Subject(s)
Cardiovascular Diseases , Cathepsin B , Humans , Cathepsin B/metabolism , Cardiovascular Diseases/metabolism , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology
20.
Sci Rep ; 14(1): 20684, 2024 09 05.
Article in English | MEDLINE | ID: mdl-39237581

ABSTRACT

Heme released from damaged and senescent red blood cells (RBCs) may contribute to oxidant-mediated cell injury. One of the recently investigated physiological processes, essential in preventing the inflammatory impact of labile heme, is its uptake from the bloodstream by endothelial cells (ECs). In this study, we investigated heme uptake by ECs starting from the model studies on the in vitro cellular level, through the endothelium layer on the ex vivo murine aortic tissues. As the cellular model, Human Aortic Endothelial Cells (HAECs) were chosen, and the concentration of labile heme was adjusted so to avoid the excessive toxic effect of the labile heme. We utilized label-free Raman imaging with two different excitation wavelengths to capture the uptake process in situ and characterize the oxidation state of the iron ion in the intercalated heme. The phenomenon of heme uptake was demonstrated in both, the healthy control C57Bl/6J and FVB animals, as well as in mice with developed atherosclerosis (ApoE/LDLR-/- mice). In the presented work, we presented for the first time Raman-based evidence on the heme uptake process by endothelial cells in both, in vitro and ex vivo systems.


Subject(s)
Endothelial Cells , Heme , Spectrum Analysis, Raman , Animals , Heme/metabolism , Spectrum Analysis, Raman/methods , Endothelial Cells/metabolism , Mice , Humans , Mice, Inbred C57BL , Atherosclerosis/metabolism , Atherosclerosis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL