Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 215
Filter
1.
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
2.
Arterioscler Thromb Vasc Biol ; 44(6): 1346-1364, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38660806

ABSTRACT

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


Subject(s)
Adipose Tissue, White , Atherosclerosis , Disease Models, Animal , Metabolic Syndrome , Mice, Inbred C57BL , Proteasome Endopeptidase Complex , Receptors, LDL , Animals , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Atherosclerosis/drug therapy , Atherosclerosis/immunology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Metabolic Syndrome/drug therapy , Metabolic Syndrome/immunology , Adipose Tissue, White/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/pathology , Receptors, LDL/genetics , Receptors, LDL/deficiency , Proteasome Endopeptidase Complex/metabolism , Male , Proteasome Inhibitors/pharmacology , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Aortic Diseases/prevention & control , Aortic Diseases/pathology , Aortic Diseases/genetics , Aortic Diseases/enzymology , Aortic Diseases/immunology , Aortic Diseases/metabolism , Macrophages/drug effects , Macrophages/metabolism , Macrophages/immunology , Plaque, Atherosclerotic , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Mice, Knockout, ApoE , Mice , Energy Metabolism/drug effects , Oligopeptides
3.
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
4.
Clin Sci (Lond) ; 135(21): 2483-2502, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34643227

ABSTRACT

Vascular calcification is highly prevalent in chronic kidney disease (CKD), and is characterized by transdifferentiation from contractile vascular smooth muscle cells (VSMCs) into an osteogenic phenotype. However, no effective and therapeutic option to prevent vascular calcification is yet available. Dihydromyricetin (DMY), a bioactive flavonoid isolated from Ampelopsis grossedentata, has been found to inhibit VSMCs proliferation and the injury-induced neointimal formation. However, whether DMY has an effect on osteogenic differentiation of VSMCs and vascular calcification is still unclear. In the present study, we sought to investigate the effect of DMY on vascular calcification in CKD and the underlying mechanism. DMY treatment significantly attenuated calcium/phosphate-induced calcification of rat and human VSMCs in a dose-dependent manner, as shown by Alizarin Red S staining and calcium content assay, associated with down-regulation of osteogenic markers including type I collagen (COL I), Runt-related transcription factor 2 (RUNX2), bone morphogenetic protein 2 (BMP2) and osteocalcin (OCN). These results were further confirmed in aortic rings ex vivo. Moreover, DMY ameliorated vascular calcification in rats with CKD. Additionally, we found that AKT signaling was activated during vascular calcification, whereas significantly inhibited by DMY administration. DMY treatment significantly reversed AKT activator-induced vascular calcification. Furthermore, inhibition of AKT signaling efficiently attenuated calcification, which was similar to that after treatment with DMY alone, and DMY had a better inhibitory effect on calcification as compared with AKT inhibitor. The present study demonstrated that DMY has a potent inhibitory role in vascular calcification partially by inhibiting AKT activation, suggesting that DMY may act as a promising therapeutic candidate for patients suffering from vascular calcification.


Subject(s)
Aortic Diseases/prevention & control , Flavonols/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Osteogenesis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Renal Insufficiency, Chronic/drug therapy , Vascular Calcification/prevention & control , Animals , Aorta/drug effects , Aorta/enzymology , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/etiology , Aortic Diseases/pathology , Cells, Cultured , Disease Models, Animal , Humans , Male , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/enzymology , Renal Insufficiency, Chronic/pathology , Signal Transduction , Vascular Calcification/enzymology , Vascular Calcification/etiology , Vascular Calcification/pathology
5.
Physiol Res ; 70(4): 533-542, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34062069

ABSTRACT

Valsartan has the potential to attenuate neointimal hyperplasia and to suppress the inflammatory response. This study aimed to evaluate the role of valsartan in neointimal hyperplasia and the toll-like receptor 4 (TLR4)-nitric oxide synthase (NOS) pathway in the balloon-injured rat aorta.Forty-eight Wistar rats were randomly allocated to three groups: sham control (control), balloon-injured group (surgery), and balloon-injured+valsartan-treated group (valsartan). Rats were killed at 14 and 28 days after balloon-injury, and then the aortic tissues were collected for morphometric analysis as well as for measurements of the mRNA or protein expression of angiotensin II, angiotensin II type 1 (AT1) receptor, angiotensin II type 2 (AT2) receptor, TLR4, endothelial nitric oxide synthase (eNOS), inducible NOS (iNOS), serine/arginine-rich splicing factor 1(SRSF1) and extracellular signal regulated kinase (ERK). Valsartan at a dose of 20 mg/kg/day markedly decreased neointimal hyperplasia in the aorta of balloon-injured rats, and significantly reduced the mRNA or protein expression of TLR4, AT1 receptor, SRSF1 and phosphorylated-ERK (p-ERK) as well as the aortic levels of iNOS (all p < 0.05). Moreover, valsartan increased the eNOS level and AT2 receptor mRNA and protein expression levels (all p < 0.05). Valsartan prevented neointimal hyperplasia and inhibited SRSF1 expression and the TLR4-iNOS-ERK-AT1 receptor pathway in the balloon-injured rat aorta.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Aorta/drug effects , Aortic Diseases/drug therapy , Extracellular Signal-Regulated MAP Kinases/metabolism , Neointima , Nitric Oxide Synthase Type II/metabolism , Receptor, Angiotensin, Type 1/metabolism , Serine-Arginine Splicing Factors/metabolism , Toll-Like Receptor 4/metabolism , Valsartan/pharmacology , Vascular System Injuries/drug therapy , Animals , Aorta/enzymology , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/pathology , Disease Models, Animal , Hyperplasia , Male , Phosphorylation , Rats, Wistar , Receptor, Angiotensin, Type 1/genetics , Signal Transduction , Toll-Like Receptor 4/genetics , Vascular System Injuries/enzymology , Vascular System Injuries/genetics , Vascular System Injuries/pathology
7.
Arterioscler Thromb Vasc Biol ; 41(6): e338-e353, 2021 06.
Article in English | MEDLINE | ID: mdl-33792343
8.
J Cell Physiol ; 236(6): 4750-4763, 2021 06.
Article in English | MEDLINE | ID: mdl-33615471

ABSTRACT

Excessively high cholesterol content in the blood leads to nonalcohol fatty liver disease (NAFLD) and arteriosclerosis. Although there are increasing publications and patent applications to lower blood cholesterol with small chemical molecules, limited effective drugs can be available in clinic. It is necessary to uncover new targets and drugs to alleviate high cholesterol. Esterase D (ESD) is abundant in liver and it remains unknown about its role in cholesterol metabolism. Here we reported that small chemical molecule fluorescigenic pyrazoline derivative 5 (FPD5), a new ESD activator, could effectively reverse high blood cholesterol level and prevent fatty liver and arteriosclerosis in apoE-/- mice fed the high-fat diet. We also observed that FPD5 could reduce oxidized low density lipoprotein (oxLDL)-induced formation of foam cells. To further investigate the mechanism of FPD5 action on blood cholesterol modulation, we found that ESD trigged by FPD5 was aggregated in lysosome and interacted with Jun activation domain binding protein 1 (JAB1). ESD served as a deacetylase to remove Thr89 acetylation of JAB1 and increased its activity; thus, promoting the ATP-binding cassette transporters A1 (ABCA1) to accelerate cholesterol efflux. Our findings demonstrate that FPD5 decreases blood cholesterol level to ameliorate NAFLD and arteriosclerosis through ESD/JAB1/ABCA1 pathway, and ESD functions as a novel nonclassical deacetylase that hydrolyzes serine/threonine acetyl group. Our findings not only highlight that FPD5 may be a pioneer drug for alleviating blood cholesterol but also indicate that ESD is a potential drug target that promotes cholesterol metabolism.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Anticholesteremic Agents/pharmacology , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , COP9 Signalosome Complex/metabolism , Cholesterol/blood , Enzyme Inhibitors/pharmacology , Foam Cells/drug effects , Peptide Hydrolases/metabolism , Thiolester Hydrolases/antagonists & inhibitors , Acetylation , Animals , Aortic Diseases/blood , Aortic Diseases/enzymology , Aortic Diseases/pathology , Atherosclerosis/blood , Atherosclerosis/enzymology , Atherosclerosis/pathology , Biomarkers/blood , Diet, High-Fat , Disease Models, Animal , Down-Regulation , Foam Cells/enzymology , Foam Cells/pathology , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/prevention & control , Plaque, Atherosclerotic , Protein Processing, Post-Translational , RAW 264.7 Cells , Thiolester Hydrolases/metabolism
9.
Cardiovasc Res ; 117(4): 1060-1069, 2021 03 21.
Article in English | MEDLINE | ID: mdl-32402085

ABSTRACT

AIMS: Fibroblast activation protein (FAP) is upregulated at sites of tissue remodelling including chronic arthritis, solid tumours, and fibrotic hearts. It has also been associated with human coronary atherosclerotic plaques. Yet, the causal role of FAP in atherosclerosis remains unknown. To investigate the cause-effect relationship of endogenous FAP in atherogenesis, we assessed the effects of constitutive Fap deletion on plaque formation in atherosclerosis-prone apolipoprotein E (Apoe) or low-density lipoprotein receptor (Ldlr) knockout mice. METHODS AND RESULTS: Using en face analyses of thoraco-abdominal aortae and aortic sinus cross-sections, we demonstrate that Fap deficiency decreased plaque formation in two atherosclerotic mouse models (-46% in Apoe and -34% in Ldlr knockout mice). As a surrogate of plaque vulnerability fibrous cap thickness was used; it was increased in Fap-deficient mice, whereas Sirius red staining demonstrated that total collagen content remained unchanged. Using polarized light, atherosclerotic lesions from Fap-deficient mice displayed increased FAP targets in terms of enhanced collagen birefringence in plaques and increased pre-COL3A1 expression in aortic lysates. Analyses of the Stockholm Atherosclerosis Gene Expression data revealed that FAP expression was increased in human atherosclerotic compared to non-atherosclerotic arteries. CONCLUSIONS: Our data provide causal evidence that constitutive Fap deletion decreases progression of experimental atherosclerosis and increases features of plaque stability with decreased collagen breakdown. Thus, inhibition of FAP expression or activity may not only represent a promising therapeutic target in atherosclerosis but appears safe at the experimental level for FAP-targeted cancer therapies.


Subject(s)
Aorta/enzymology , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Endopeptidases/deficiency , Membrane Proteins/deficiency , Vascular Remodeling , Animals , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Case-Control Studies , Collagen/genetics , Collagen/metabolism , Disease Models, Animal , Endopeptidases/genetics , Fibrosis , Gene Deletion , Humans , Lipids/blood , Male , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout, ApoE , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Plaque, Atherosclerotic , Proteome , Receptors, LDL/deficiency , Receptors, LDL/genetics , Transcriptome
10.
Cardiovasc Res ; 117(5): 1295-1308, 2021 04 23.
Article in English | MEDLINE | ID: mdl-32667970

ABSTRACT

AIMS: Atherosclerotic vascular disease has an inflammatory pathogenesis. Heme from intraplaque haemorrhage may drive a protective and pro-resolving macrophage M2-like phenotype, Mhem, via AMPK and activating transcription factor 1 (ATF1). The antidiabetic drug metformin may also activate AMPK-dependent signalling. Hypothesis: Metformin systematically induces atheroprotective genes in macrophages via AMPK and ATF1, thereby suppresses atherogenesis. METHODS AND RESULTS: Normoglycaemic Ldlr-/- hyperlipidaemic mice were treated with oral metformin, which profoundly suppressed atherosclerotic lesion development (P < 5 × 10-11). Bone marrow transplantation from AMPK-deficient mice demonstrated that metformin-related atheroprotection required haematopoietic AMPK [analysis of variance (ANOVA), P < 0.03]. Metformin at a clinically relevant concentration (10 µM) evoked AMPK-dependent and ATF1-dependent increases in Hmox1, Nr1h2 (Lxrb), Abca1, Apoe, Igf1, and Pdgf, increases in several M2-markers and decreases in Nos2, in murine bone marrow macrophages. Similar effects were seen in human blood-derived macrophages, in which metformin-induced protective genes and M2-like genes, suppressible by si-ATF1-mediated knockdown. Microarray analysis comparing metformin with heme in human macrophages indicated that the transcriptomic effects of metformin were related to those of heme, but not identical. Metformin-induced lesional macrophage expression of p-AMPK, p-ATF1, and downstream M2-like protective effects. CONCLUSION: Metformin activates a conserved AMPK-ATF1-M2-like pathway in mouse and human macrophages, and results in highly suppressed atherogenesis in hyperlipidaemic mice via haematopoietic AMPK.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Activating Transcription Factor 1/metabolism , Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Macrophages/drug effects , Metformin/pharmacology , Plaque, Atherosclerotic , AMP-Activated Protein Kinases/genetics , Activating Transcription Factor 1/genetics , Animals , Aorta/enzymology , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation , Humans , Macrophages/enzymology , Macrophages/pathology , Mice, Knockout , Phenotype , Phosphorylation , Receptors, LDL/genetics , Receptors, LDL/metabolism , Signal Transduction
11.
Vascul Pharmacol ; 133-134: 106777, 2020.
Article in English | MEDLINE | ID: mdl-32750408

ABSTRACT

Atherosclerosis is a systemic chronic inflammatory disease. Many antioxidants including alpha-lipoic acid (LA), a product of lipoic acid synthase (Lias), have proven to be effective for treatment of this disease. However, the question remains whether LA regulates the immune response as a protective mechanism against atherosclerosis. We initially investigated whether enhanced endogenous antioxidant can retard the development of atherosclerosis via immunomodulation. To explore the impact of enhanced endogenous antioxidant on the retardation of atherosclerosis via immune regulation, our laboratory has recently created a double mutant mouse model, using apolipoprotein E-deficient (Apoe-/-) mice crossbred with mice overexpressing lipoic acid synthase gene (LiasH/H), designated as LiasH/HApoe-/- mice. Their littermates, Lias+/+Apoe-/- mice, served as a control. Distinct redox environments between the two strains of mice have been established and they can be used to facilitate identification of antioxidant targets in the immune response. At 6 months of age, LiasH/HApoe-/- mice had profoundly decreased atherosclerotic lesion size in the aortic sinus compared to their Lias+/+Apoe-/- littermates, accompanied by significantly enhanced numbers of regulatory T cells (Tregs) and anti-oxidized LDL autoantibody in the vascular system, and reduced T cell infiltrates in aortic walls. Our results represent a novel exploration into an environment with increased endogenous antioxidant and its ability to alleviate atherosclerosis, likely through regulation of the immune response. These outcomes shed light on a new therapeutic strategy using antioxidants to lessen atherosclerosis.


Subject(s)
Aorta/enzymology , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Plaque, Atherosclerotic , Sulfurtransferases/biosynthesis , Animals , Aorta/immunology , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/immunology , Aortic Diseases/pathology , Atherosclerosis/enzymology , Atherosclerosis/immunology , Atherosclerosis/pathology , Autoantibodies/blood , Disease Models, Animal , Enzyme Induction , Lipoproteins, LDL/immunology , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Oxidation-Reduction , Oxidative Stress , Sulfurtransferases/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
12.
Arterioscler Thromb Vasc Biol ; 40(5): 1155-1167, 2020 05.
Article in English | MEDLINE | ID: mdl-32212851

ABSTRACT

OBJECTIVES: During the advancement of atherosclerosis, plaque cellularity is governed by the influx of monocyte-derived macrophages and their turnover via apoptotic and nonapoptotic forms of cell death. Previous reports have demonstrated that programmed necrosis, or necroptosis, of plaque macrophages contribute to necrotic core formation. Knockdown or inhibition of the necrosome components RIPK1 (receptor-interacting protein kinase 1) and RIPK3 (receptor-interacting protein kinase 3) slow atherogenesis, and activation of the terminal step of necroptosis, MLKL (mixed lineage kinase domain-like protein), has been demonstrated in advanced human atherosclerotic plaques. However, whether MLKL directly contributes to lesion development and necrotic core formation has not been investigated. Approaches and Results: MLKL expression was knocked down in atherogenic Apoe-knockout mice via the administration of antisense oligonucleotides. During atherogenesis, Mlkl knockdown decreased both programmed cell death and the necrotic core in the plaque. However, total lesion area remained unchanged. Furthermore, treatment with the MLKL antisense oligonucleotide unexpectedly reduced circulating cholesterol levels compared with control antisense oligonucleotide but increased the accumulation of lipids within the plaque and in vitro in macrophage foam cells. MLKL colocalized with the late endosome and multivesicular bodies in peritoneal macrophages incubated with atherogenic lipoproteins. Transfection with MLKL antisense oligonucleotide increased lipid localization with the multivesicular bodies, suggesting that upon Mlkl knockdown, lipid trafficking becomes defective leading to enhanced lipid accumulation in macrophages. CONCLUSIONS: These studies confirm the requirement for MLKL as the executioner of necroptosis, and as such a significant contributor to the necrotic core during atherogenesis. We also identified a previously unknown role for MLKL in regulating endosomal trafficking to facilitate lipid handling in macrophages during atherogenesis.


Subject(s)
Aortic Diseases/enzymology , Atherosclerosis/enzymology , Cholesterol/metabolism , Foam Cells/enzymology , Macrophages, Peritoneal/enzymology , Plaque, Atherosclerotic , Protein Kinases/deficiency , Animals , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Disease Models, Animal , Endosomes/metabolism , Female , Foam Cells/pathology , Macrophages, Peritoneal/pathology , Male , Mice, Knockout, ApoE , Necroptosis , Necrosis , Oligonucleotides, Antisense/administration & dosage , Protein Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction
13.
BMC Cardiovasc Disord ; 20(1): 133, 2020 03 13.
Article in English | MEDLINE | ID: mdl-32169038

ABSTRACT

BACKGROUND: Previous studies have indicated that the JAK/STAT signaling pathway is involved in modulating arterial adventitia inflammation response. In this study, we designed experiments to further investigate the effect of JAK2/STAT3/SOCS3 signaling in rabbit atherosclerosis process. METHODS: Atherosclerosis was induced in the abdominal arteries of rabbits by balloon injury of the aorta supplemented by the atherogenic diet. Simultaneously, in the process of atherosclerosis, animals underwent either ruxolitinib treatment or not for 12 weeks. At the end of the experimental period, all rabbits were sacrificed. The plaque areas in abdominal artery, the lipid burden of plaque and the calcium burden of plaque were detected by H&E staining, Oil Red O staining and Alizarin Red staining, respectively. In addition, rabbit plasma lipids and inflammatory cytokines were measured by biochemical test kits or ELISA kits. Finally, the expression and phosphorylation levels of JAK2/STAT3/SOCS3 pathway-related proteins were detected by RT-qPCR, western blot and immunohistochemistry assays. RESULTS: H&E staining and CT scan analysis showed that rabbit atherosclerosis model was constructed successfully. Ruxolitinib, an inhibitor of the Janus kinase 2 (JAK2), substantially reduced the area of atherosclerotic plaques in rabbits treated with high fat diet and balloon injury of the aorta. Moreover, ruxolitinib significantly decreased IL-6, IL-1ß, IFN-γ and TNF-α, but increased IL-10 and IL-17 levels in plasma of atherosclerotic rabbits. Additionally, ruxolitinib reduced plasma TC, TG and LDL-C contents and AIP value, while enhanced HDL-C level in atherosclerotic rabbits. Furthermore, we found that JAK2 and STAT3 phosphorylation were up-regulated in rabbits with atherosclerosis when compared with those of the control group, followed by the expression of SOCS3 was also increased due to the activation of JAK2 and STAT3. Interestingly, ruxolitinib could inactivate JAK2 and STAT3 pathway and decrease SOCS3 expression. CONCLUSION: Taken together, the inhibition of JAK2/STAT3/SOCS3 signaling pathway may be a novel method for the clinical treatment of artery atherosclerosis.


Subject(s)
Aorta, Abdominal/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Janus Kinase 2/antagonists & inhibitors , Janus Kinase Inhibitors/pharmacology , Plaque, Atherosclerotic , Pyrazoles/pharmacology , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Animals , Aorta, Abdominal/enzymology , Aorta, Abdominal/pathology , Aortic Diseases/blood , Aortic Diseases/enzymology , Aortic Diseases/pathology , Atherosclerosis/blood , Atherosclerosis/enzymology , Atherosclerosis/pathology , Cytokines/blood , Disease Models, Animal , Inflammation Mediators/blood , Janus Kinase 2/metabolism , Lipids/blood , Male , Nitriles , Phosphorylation , Pyrimidines , Rabbits , Signal Transduction
14.
Cardiovasc Drugs Ther ; 34(2): 145-152, 2020 04.
Article in English | MEDLINE | ID: mdl-32086626

ABSTRACT

OBJECTIVE: Increased myelopoiesis has been linked to risk of atherosclerotic cardiovascular disease (ACD). Excessive myelopoiesis can be driven by dyslipidemia and cholesterol accumulation in hematopoietic stem and progenitor cells (HSPC) and may involve increased signaling via Janus kinase 2 (JAK2). Constitutively activating JAK2 mutants drive biased myelopoiesis and promote development of myeloproliferative neoplasms (MPN) or clonal hematopoiesis, conditions associated with increased risk of ACD. JAK2 inhibitors have been developed as a therapy for MPNs. The potential for JAK2 inhibitors to protect against atherosclerosis has not been tested. We therefore assessed the impact of JAK2 inhibition on atherogenesis. METHODS: A selective JAK2 inhibitor TG101348 (fedratinib) or vehicle was given to high-fat high-cholesterol Western diet (WD)-fed wild-type (WT) or Apoe-/- mice. Hematopoietic cell profiles, cell proliferation, and atherosclerosis in WT or Apoe-/- mice were assessed. RESULTS: TG101348 selectively reversed neutrophilia, monocytosis, HSPC, and granulocyte-macrophage progenitor (GMP) expansion in Apoe-/- mice with decreased cellular phosphorylated STAT5 and ERK1/2 and reduced cell cycling and BrdU incorporation in HSPCs, indicating inhibition of JAK/STAT signaling and cell proliferation. Ten-week WD feeding allowed the development of marked aortic atherosclerosis in Apoe-/- mice which was substantially reduced by TG101348. CONCLUSIONS: Selective JAK2 inhibition reduces atherogenesis by suppressing excessive myelopoiesis in hypercholesterolemic Apoe-/- mice. These findings suggest selective JAK2 inhibition as a potential therapeutic approach to decrease ACD risk in patients with increased myelopoiesis and leukocytosis.


Subject(s)
Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Hematopoietic Stem Cells/drug effects , Janus Kinase 2/antagonists & inhibitors , Janus Kinase Inhibitors/pharmacology , Myelopoiesis/drug effects , Pyrrolidines/pharmacology , Sulfonamides/pharmacology , Animals , Aorta/enzymology , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Female , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/pathology , Janus Kinase 2/metabolism , Leukocytosis/enzymology , Leukocytosis/prevention & control , Mice, Inbred C57BL , Mice, Knockout, ApoE , Plaque, Atherosclerotic , Signal Transduction
15.
J Cardiovasc Pharmacol ; 75(4): 321-332, 2020 04.
Article in English | MEDLINE | ID: mdl-31895870

ABSTRACT

Artemisinin is an endoperoxide sesquiterpene lactone from Artemisia annua L with multiple beneficial effects, including anti-inflammation, antioxidant, and vascular protection. Recent studies have found that inflammation along with autophagy deficiency in macrophages is the possible reason for foam cell accumulation in the intima, which leads to atherosclerotic plaque formation. The primary aims of this study were to explore the inhibiting effect of artemisinin on atherosclerosis in high-fat diet-fed ApoE mice and investigate the probable mechanism. Artemisinin (50 and 100 mg/kg, intragastric administration) treatment effectively inhibited foamy macrophage transformation and decreased atherosclerotic plaque formation in atherosclerotic mice. Moreover, artemisinin promoted AMP-activated protein kinase (AMPK) activation, inhibited mammalian target of rapamycin (mTOR) and uncoordinated-51-like kinase 1 (ULK1) phosphorylation, and increased LC-3II accumulation and P62 degradation, thereby enhancing macrophage autophagy. Besides, the inhibiting effect of artemisinin on mTOR and ULK1 phosphorylation could be abrogated by AMPK knockdown, suggesting AMPK was the essential target of artemisinin on promoting macrophage autophagy. Our study indicated that artemisinin alleviated atherosclerotic lesions by accelerating macrophage autophagy through the AMPK/mTOR/ULK1 pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Anti-Inflammatory Agents/pharmacology , Aorta/drug effects , Aortic Diseases/prevention & control , Artemisinins/pharmacology , Atherosclerosis/prevention & control , Autophagy/drug effects , Foam Cells/drug effects , TOR Serine-Threonine Kinases/metabolism , Animals , Aorta/enzymology , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Autophagy-Related Protein-1 Homolog/metabolism , Diet, High-Fat , Disease Models, Animal , Foam Cells/enzymology , Foam Cells/pathology , Lipoproteins, LDL/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Microtubule-Associated Proteins/metabolism , Phosphorylation , Plaque, Atherosclerotic , RAW 264.7 Cells , Sequestosome-1 Protein/metabolism , Signal Transduction
16.
J Pathol ; 250(1): 30-41, 2020 01.
Article in English | MEDLINE | ID: mdl-31509234

ABSTRACT

Medial arterial calcification (MAC) is a major complication of chronic kidney disease (CKD) and an indicator of poor prognosis. Aortic overexpression of tissue-nonspecific alkaline phosphatase (TNAP) accelerates MAC formation. The present study aimed to assess whether a TNAP inhibitor, SBI-425, protects against MAC and improves survival probability in a CKD-mineral and bone disorder (MBD) mouse model. CKD-MBD mice were divided in three groups: vehicle, SBI-10, and SBI-30. They were fed a 0.2% adenine and 0.8% phosphorus diet from 14 to 20 weeks of age to induce CKD, followed by a high-phosphorus (0.2% adenine and 1.8% phosphorus) diet for another 6 weeks. At 14-20 weeks of age, mice in the SBI-10 and SBI-30 groups were given 10 and 30 mg/kg SBI-425 by gavage once a day, respectively, while vehicle-group mice were given distilled water as vehicle. Control mice were fed a standard chow (0.8% phosphorus) between the ages of 8 and 20 weeks. Computed tomography imaging, histology, and aortic tissue calcium content revealed that, compared to vehicle animals, SBI-425 nearly halted the formation of MAC. Mice in the control, SBI-10 and SBI-30 groups exhibited 100% survival, which was significantly better than vehicle-treated mice (57.1%). Aortic mRNA expression of Alpl, encoding TNAP, as well as plasma and aortic tissue TNAP activity, were suppressed by SBI-425 administration, whereas plasma pyrophosphate increased. We conclude that a TNAP inhibitor successfully protected the vasculature from MAC and improved survival rate in a mouse CKD-MBD model, without causing any adverse effects on normal skeletal formation and residual renal function. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Alkaline Phosphatase/antagonists & inhibitors , Aorta/drug effects , Aortic Diseases/prevention & control , Chronic Kidney Disease-Mineral and Bone Disorder/drug therapy , Enzyme Inhibitors/pharmacology , Niacinamide/analogs & derivatives , Sulfonamides/pharmacology , Vascular Calcification/prevention & control , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Aorta/enzymology , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/etiology , Aortic Diseases/pathology , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Chronic Kidney Disease-Mineral and Bone Disorder/enzymology , Chronic Kidney Disease-Mineral and Bone Disorder/pathology , Disease Models, Animal , Gene Expression Regulation , Male , Mice, Inbred C57BL , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Niacinamide/pharmacology , Osteoblasts/drug effects , Osteoblasts/enzymology , Osteoblasts/pathology , Time Factors , Vascular Calcification/enzymology , Vascular Calcification/etiology , Vascular Calcification/pathology
17.
Proc Natl Acad Sci U S A ; 116(47): 23698-23704, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31690656

ABSTRACT

Pyrophosphate deficiency may explain the excessive vascular calcification found in children with Hutchinson-Gilford progeria syndrome (HGPS) and in a mouse model of this disease. The present study found that hydrolysis products of ATP resulted in a <9% yield of pyrophosphate in wild-type blood and aortas, showing that eNTPD activity (ATP → phosphate) was greater than eNPP activity (ATP → pyrophosphate). Moreover, pyrophosphate synthesis from ATP was reduced and pyrophosphate hydrolysis (via TNAP; pyrophosphate → phosphate) was increased in both aortas and blood obtained from mice with HGPS. The reduced production of pyrophosphate, together with the reduction in plasma ATP, resulted in marked reduction of plasma pyrophosphate. The combination of TNAP inhibitor levamisole and eNTPD inhibitor ARL67156 increased the synthesis and reduced the degradation of pyrophosphate in aortas and blood ex vivo, suggesting that these combined inhibitors could represent a therapeutic approach for this devastating progeroid syndrome. Treatment with ATP prevented vascular calcification in HGPS mice but did not extend longevity. By contrast, combined treatment with ATP, levamisole, and ARL67156 prevented vascular calcification and extended longevity by 12% in HGPS mice. These findings suggest a therapeutic approach for children with HGPS.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Alkaline Phosphatase/physiology , Aortic Diseases/prevention & control , Apyrase/antagonists & inhibitors , Calcinosis/prevention & control , Diphosphates/metabolism , Levamisole/therapeutic use , Progeria/drug therapy , Pyrophosphatases/antagonists & inhibitors , Adenosine Triphosphate/therapeutic use , Alkaline Phosphatase/antagonists & inhibitors , Animals , Antigens, CD/physiology , Aortic Diseases/enzymology , Apyrase/deficiency , Apyrase/physiology , Calcinosis/enzymology , Disease Models, Animal , Gene Knock-In Techniques , Humans , Lamin Type A/genetics , Longevity/drug effects , Male , Mice , Mice, Transgenic , Myocytes, Smooth Muscle/metabolism , Phosphoric Diester Hydrolases/deficiency , Phosphoric Diester Hydrolases/physiology , Progeria/genetics , Progeria/metabolism , Progeria/pathology , Pyrophosphatases/deficiency , Pyrophosphatases/physiology , RNA Interference , RNA, Small Interfering/pharmacology , Real-Time Polymerase Chain Reaction
18.
Atherosclerosis ; 291: 78-86, 2019 12.
Article in English | MEDLINE | ID: mdl-31704554

ABSTRACT

BACKGROUND AND AIMS: Atherosclerosis is a chronic inflammatory disorder mediated by macrophage activation. MicroRNA-21 (miR-21) is a key regulator in the macrophage inflammatory response. However, the functional role of miR-21 in atherogenesis is far from clear. METHODS AND RESULTS: Here, we report that miR-21 is significantly upregulated in mouse atherosclerotic plaques and peripheral monocytes from patients with coronary artery disease. Compared with miR-21+/+apoE-/- mice (apoE-/- mice), miR-21-/-apoE-/- (double knockout, DKO) mice showed less atherosclerotic lesions, reduced presence of macrophages, decreased smooth muscle cells(SMC) and collagen content in the aorta. We further explored the role of miR-21 in macrophage activation in vitro. Bone marrow-derived macrophages (BMDMs) from DKO mice not only exhibit impaired function of migration induced by chemokine (C-C motif) ligand 2 (CCL2) but also a weakened macrophage-endothelium interaction activated by tumor necrosis factor-α (TNF-α). However, atherogenic inflammatory cytokine secretion was not affected by miR-21 in vitro or in vivo. Additionally, miR-21 knockdown in BMDMs directly derepressed the expression of dual specificity protein phosphatase 8 (Dusp-8), a previously validated miR-21 target in cardiac fibroblasts, which negatively regulates mitogen-activated protein kinase (MAPK) signaling, particularly the p38-and c-Jun N-terminal kinase (JNK)-related signaling pathways. CONCLUSIONS: These data demonstrate that inhibition of miR-21 may restrict the formation of atherosclerotic plaques partly by regulating macrophage migration and adhesion, while, reduced SMCs and collagen content in plaques may lead to a less stable phenotype with the progression of atherosclerosis. Thus, the absence of miR-21 reduces atherosclerotic lesions but may not represent all benefit in atherosclerosis development.


Subject(s)
Aorta/enzymology , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Chemotaxis , Dual-Specificity Phosphatases/metabolism , Macrophage Activation , Macrophages/enzymology , MicroRNAs/metabolism , Animals , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cell Adhesion , Disease Models, Animal , Dual-Specificity Phosphatases/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophages/pathology , Male , Mice , Mice, Knockout, ApoE , MicroRNAs/genetics , Plaque, Atherosclerotic , RAW 264.7 Cells , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
19.
Vascul Pharmacol ; 121: 106582, 2019 10.
Article in English | MEDLINE | ID: mdl-31437529

ABSTRACT

BACKGROUND: The contentious effects of estrogen therapy on the risk of postmenopausal cardiovascular disease (CVD) indicate that this type of atherosclerosis is not solely induced by estrogen deficiency. Other sex hormones such as elevated luteinizing hormone (LH) may also affect CVD risk in this population. We therefore explored the relationship between LH and atherosclerosis in ovariectomized (OVX) female mice. METHODS: Aortic atherosclerotic lesions were assessed in OVX ApoE knock out (ApoE-/-) female mice administered with LH. Human umbilical vascular endothelial cells (HUVECs) were cultured as cell model. The influence of LH on NO release, phosphorylated endothelial nitric oxide synthase (eNOS) and Akt levels were evaluated. Immunoprecipitation and lentiviral particle transfection were applied to assess the role of Gαq on PI3K activity. RESULTS: LH increased the atherosclerotic lesion area and carotid artery intima-media thickness (IMT) in OVX ApoE-/- female mice. High levels of LH attenuated vasodilation induced by Ach and inhibited NO release from HUVECs. These effects were related to the findings that LH enhanced interaction between Gαq and p110α, which subsequently inhibited PI3K activity and suppressed the phosphorylation of Akt and eNOS. CONCLUSIONS: Elevated LH promotes atherosclerosis formation in OVX ApoE-/- female mice. This effect may be mediated by inhibiting endothelial NO synthesis via PI3K/Akt signaling pathway.


Subject(s)
Aorta/drug effects , Aortic Diseases/chemically induced , Atherosclerosis/chemically induced , Class I Phosphatidylinositol 3-Kinases/metabolism , Luteinizing Hormone/toxicity , Nitric Oxide/metabolism , Plaque, Atherosclerotic , Proto-Oncogene Proteins c-akt/metabolism , Animals , Aorta/enzymology , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Female , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Mice, Knockout, ApoE , Nitric Oxide Synthase Type III/metabolism , Ovariectomy , Phosphorylation , Signal Transduction
20.
J Cardiovasc Pharmacol ; 74(2): 152-161, 2019 08.
Article in English | MEDLINE | ID: mdl-31306371

ABSTRACT

BACKGROUND AND AIMS: High-fat diet (HFD) is reported to induce atherosclerosis and insulin resistance. Macrophage lipid accumulation has been implicated as key mediators during the development of HFD-induced atherosclerosis. Traditional Chinese formula, which has long been used to improve disorder of glucose and lipid metabolism of patients, is now gradually being used as complementary therapy. This study aimed to investigate the effect of Danhong injection (DHI), a Chinese medicine used for the treatment of coronary artery disease, on atherosclerosis and its underlying mechanisms. METHODS AND RESULTS: We observed the effects of DHI on HFD-induced atherosclerosis in a mice model, macrophage lipid accumulation in an ox-LDL-stimulated macrophage model, and the role of PI3K/AKT insulin pathway in the process of DHI ameliorating atherosclerosis. The data demonstrated that DHI attenuated atherosclerosis by ameliorating blood lipids, reducing the atherosclerotic index and atherosclerotic plaque area in HFD-induced atherosclerotic mice, and inhibiting TC levels in an ox-LDL-induced macrophage model. By estimating the levels of serum insulin resistance-related indexes and protein expression of GLUT-4, DHI treatment dramatically inhibited the levels of fasting serum NEFA and fasting serum insulin and promoted the protein expression of GLUT-4 in aortas of the HFD-induced atherosclerotic mice. Moreover, according to the hints provided by microarray-based transcriptional profiling, the results demonstrated that DHI treatment also promoted the activation of PI3K/AKT insulin signaling pathway induced by IRS-1 in aortas of HFD-induced atherosclerotic mice. Furthermore, in an ox-LDL-induced macrophage model, the activation of PI3k/AKT signaling pathway also effectively functioned in the process of DHI inhibiting macrophage lipid accumulation. CONCLUSIONS: These results highlight that DHI treatment attenuates atherosclerosis and macrophage lipid accumulation by promoting the activation of PI3K/AKT insulin signaling pathway. It provides new insights into the molecular mechanism of DHI and its therapeutic potential in the treatment of atherosclerosis.


Subject(s)
Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Drugs, Chinese Herbal/administration & dosage , Hypolipidemic Agents/administration & dosage , Insulin/metabolism , Lipids/blood , Macrophages/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Aorta/enzymology , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Diet, High-Fat , Disease Models, Animal , Gene Expression Regulation , Injections, Intraperitoneal , Insulin/genetics , Macrophages/enzymology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Phosphatidylinositol 3-Kinase/genetics , Plaque, Atherosclerotic , Proto-Oncogene Proteins c-akt/genetics , RAW 264.7 Cells , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...