Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 32
1.
Tissue Cell ; 86: 102290, 2024 Feb.
Article En | MEDLINE | ID: mdl-38103473

Accelerating the repair of damaged endothelium can effectively inhibit the progression of atherosclerosis (AS). Transient receptor potential channel TRPM4 is a non-selective cation channel activated by internal Ca2+, which is expressed in endothelial cells. This study aimed to reveal the potential role of TRPM4 in AS along with the mechanism. Human coronary artery endothelial cells (HCAECs) induced by ox-LDL was regarded as an in vitro model. The impacts of TRPM4 knockdown on cellular inflammation response, oxidative stress, normal endothelial function and lipid peroxidation were evaluated. Given that ferroptosis promotes AS progression, the effects of TRPM4 on intracellular iron ions and ferroptosis-related proteins was determined. Afterwards, HCAECs were treated with ferroptosis inducer erastin, and the influence of ferroptosis in the cellular model was revealed. TRPM4 was elevated in response to ox-LDL treatment in HCAECs. TRPM4 knockdown reduced the inflammation response, oxidative stress and lipid peroxidation caused by ox-LDL, and maintained the normal function of HCAECs. Erastin treatment destroyed the impacts of TRPM4 knockdown that are beneficial for cells to resist ox-LDL, showing the enhancement of the above adverse factors. Together, this study found that TRPM4 knockdown reduced ox-LDL-induced inflammation, oxidative stress, and dysfunction in HCAECs, possibly via a mechanism involving Fe2+ and ferroptosis-related proteins.


Ferroptosis , TRPM Cation Channels , Humans , Receptors, LDL/metabolism , Receptors, Oxidized LDL/metabolism , Endothelial Cells/metabolism , Scavenger Receptors, Class E/metabolism , Cells, Cultured , Lipoproteins, LDL/pharmacology , Lipoproteins, LDL/metabolism , Coronary Vessels/metabolism , Proteins/metabolism , Inflammation/metabolism , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism
2.
J Mol Cell Cardiol ; 162: 110-118, 2022 01.
Article En | MEDLINE | ID: mdl-34555408

It is well known that lectin-like oxidized low-density lipoprotein (ox-LDL) and its receptor LOX-1, angiotensin II (AngII) and its type 1 receptor (AT1-R) play an important role in the development of cardiac hypertrophy. However, the molecular mechanism is not clear. In this study, we found that ox-LDL-induced cardiac hypertrophy was suppressed by inhibition of LOX-1 or AT1-R but not by AngII inhibition. These results suggest that the receptors LOX-1 and AT1-R, rather than AngII, play a key role in the role of ox-LDL. The same results were obtained in mice lacking endogenous AngII and their isolated cardiomyocytes. Ox-LDL but not AngII could induce the binding of LOX-1 and AT1-R; inhibition of LOX-1 or AT1-R but not AngII could abolish the binding of these two receptors. Overexpression of wild type LOX-1 with AT1-R enhanced ox-LDL-induced binding of two receptors and phosphorylation of ERKs, however, transfection of LOX-1 dominant negative mutant (lys266ala / lys267ala) or an AT1-R mutant (glu257ala) not only reduced the binding of two receptors but also inhibited the ERKs phosphorylation. Phosphorylation of ERKs induced by ox-LDL in LOX-1 and AT1-R-overexpression cells was abrogated by an inhibitor of Gq protein rather than Jak2, Rac1 or RhoA. Genetically, an AT1-R mutant lacking Gq protein coupling ability inhibited ox-LDL induced ERKs phosphorylation. Furthermore, through bimolecular fluorescence complementation analysis, we confirmed that ox-LDL rather than AngII stimulation induced the direct binding of LOX-1 and AT1-R. We conclude that direct binding of LOX-1 and AT1-R and the activation of downstream Gq protein are important mechanisms of ox-LDL-induced cardiomyocyte hypertrophy.


Angiotensin II , Scavenger Receptors, Class E , Angiotensin II/metabolism , Angiotensin II/pharmacology , Animals , Cells, Cultured , Lipoproteins, LDL/metabolism , Mice , Myocytes, Cardiac/metabolism , Receptors, LDL/metabolism , Receptors, Oxidized LDL/metabolism , Scavenger Receptors, Class E/genetics , Scavenger Receptors, Class E/metabolism
3.
Circulation ; 139(2): 243-255, 2019 01 08.
Article En | MEDLINE | ID: mdl-30586697

BACKGROUND: Although the role of Th17 and regulatory T cells in the progression of atherosclerosis has been highlighted in recent years, their molecular mediators remain elusive. We aimed to evaluate the association between the CD69 receptor, a regulator of Th17/regulatory T cell immunity, and atherosclerosis development in animal models and in patients with subclinical disease. METHODS: Low-density lipoprotein receptor-deficient chimeric mice expressing or not expressing CD69 on either myeloid or lymphoid cells were subjected to a high fat diet. In vitro functional assays with human T cells were performed to decipher the mechanism of the observed phenotypes. Expression of CD69 and NR4A nuclear receptors was evaluated by reverse transcription-polymerase chain reaction in 305 male participants of the PESA study (Progression of Early Subclinical Atherosclerosis) with extensive (n=128) or focal (n=55) subclinical atherosclerosis and without disease (n=122). RESULTS: After a high fat diet, mice lacking CD69 on lymphoid cells developed large atheroma plaque along with an increased Th17/regulatory T cell ratio in blood. Oxidized low-density lipoprotein was shown to bind specifically and functionally to CD69 on human T lymphocytes, inhibiting the development of Th17 cells through the activation of NR4A nuclear receptors. Participants of the PESA study with evidence of subclinical atherosclerosis displayed a significant CD69 and NR4A1 mRNA downregulation in peripheral blood leukocytes compared with participants without disease. The expression of CD69 remained associated with the risk of subclinical atherosclerosis in an adjusted multivariable logistic regression model (odds ratio, 0.62; 95% CI, 0.40-0.94; P=0.006) after adjustment for traditional risk factors, the expression of NR4A1, the level of oxidized low-density lipoprotein, and the counts of different leucocyte subsets. CONCLUSIONS: CD69 depletion from the lymphoid compartment promotes a Th17/regulatory T cell imbalance and exacerbates the development of atherosclerosis. CD69 binding to oxidized low-density lipoprotein on T cells induces the expression of anti-inflammatory transcription factors. Data from a cohort of the PESA study with subclinical atherosclerosis indicate that CD69 expression in PBLs inversely correlates with the presence of disease. The expression of CD69 remained an independent predictor of subclinical atherosclerosis after adjustment for traditional risk factors.


Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Atherosclerosis/prevention & control , Immunity, Cellular , Lectins, C-Type/metabolism , Lipoproteins, LDL/metabolism , Receptors, Oxidized LDL/metabolism , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , Adult , Animals , Antigens, CD/genetics , Antigens, Differentiation, T-Lymphocyte/genetics , Asymptomatic Diseases , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Disease Models, Animal , Female , Genetic Predisposition to Disease , Humans , Jurkat Cells , Lectins, C-Type/deficiency , Lectins, C-Type/genetics , Male , Mice, Knockout , Middle Aged , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Phenotype , Plaque, Atherosclerotic , Prospective Studies , Rats , Receptors, LDL/genetics , Receptors, LDL/metabolism , Risk Factors , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Th17 Cells/immunology , Th17 Cells/pathology
4.
Gynecol Endocrinol ; 34(3): 217-222, 2018 Mar.
Article En | MEDLINE | ID: mdl-28949260

OBJECTIVE: This research was conducted to assess the effects of coenzyme Q10 (CoQ10) intake on gene expression related to insulin, lipid and inflammation in subjects with polycystic ovary syndrome (PCOS). METHODS: This randomized double-blind, placebo-controlled trial was conducted on 40 subjects diagnosed with PCOS. Subjects were randomly allocated into two groups to intake either 100 mg CoQ10 (n = 20) or placebo (n = 20) per day for 12 weeks. Gene expression related to insulin, lipid and inflammation were quantified in blood samples of PCOS women with RT-PCR method. RESULTS: Results of RT-PCR shown that compared with the placebo, CoQ10 intake downregulated gene expression of oxidized low-density lipoprotein receptor 1 (LDLR) (p < 0.001) and upregulated gene expression of peroxisome proliferator-activated receptor gamma (PPAR-γ) (p = 0.01) in peripheral blood mononuclear cells of subjects with PCOS. In addition, compared to the placebo group, CoQ10 supplementation downregulated gene expression of interleukin-1 (IL-1) (p = 0.03), interleukin-8 (IL-8) (p = 0.001) and tumor necrosis factor alpha (TNF-α) (p < 0.001) in peripheral blood mononuclear cells of subjects with PCOS. CONCLUSIONS: Overall, CoQ10 intake for 12 weeks in PCOS women significantly improved gene expression of LDLR, PPAR-γ, IL-1, IL-8 and TNF-α.


Dietary Supplements , Gene Expression/drug effects , Inflammation/genetics , Insulin/genetics , Lipid Metabolism/genetics , Polycystic Ovary Syndrome/genetics , Ubiquinone/analogs & derivatives , Adult , Double-Blind Method , Female , Gene Expression Regulation/drug effects , Humans , Inflammation/metabolism , Insulin/metabolism , Interleukin-1/genetics , Interleukin-1/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Leukocytes, Mononuclear/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Polycystic Ovary Syndrome/metabolism , Receptors, Oxidized LDL/genetics , Receptors, Oxidized LDL/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Ubiquinone/administration & dosage
5.
PLoS One ; 10(8): e0133845, 2015.
Article En | MEDLINE | ID: mdl-26305474

Neointimal hyperplasia of vascular smooth muscle cells (VSMC) plays a critical role in atherosclerotic plaque formation and in-stent restenosis, but the underlying mechanisms are still incompletely understood. We performed a proteomics study to identify novel signaling molecules organizing the VSMC hyperplasia. The differential proteomics analysis in a balloon-induced injury model of rat carotid artery revealed that the expressions of 44 proteins are changed within 3 days post injury. The combination of cellular function assays and a protein network analysis further demonstrated that 27 out of 44 proteins constitute key signaling networks orchestrating the phenotypic change of VSMC from contractile to epithelial-like synthetic. Among the list of proteins, the in vivo validation specifically revealed that six proteins (Rab15, ITR, OLR1, PDHß, PTPε) are positive regulators for VSMC hyperplasia. In particular, the OLR1 played dual roles in the VSMC hyperplasia by directly mediating oxidized LDL-induced monocyte adhesion via NF-κB activation and by assisting the PDGF-induced proliferation/migration. Importantly, OLR1 and PDGFRß were associated in close proximity in the plasma membrane. Thus, this study elicits the protein network organizing the phenotypic change of VSMC in the vascular injury diseases such as atherosclerosis and discovers OLR1 as a novel molecular link between the proliferative and inflammatory responses of VSMCs.


Carotid Arteries/metabolism , Inflammation/metabolism , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Proteomics , Receptors, Oxidized LDL/metabolism , Scavenger Receptors, Class E/metabolism , Animals , Carotid Arteries/pathology , Cell Proliferation , Humans , Hyperplasia , Inflammation/pathology , Male , Models, Biological , Muscle, Smooth, Vascular/pathology , Neointima/metabolism , Neointima/pathology , Phenotype , Platelet-Derived Growth Factor/metabolism , Protein Interaction Maps , Proteome/metabolism , Rats, Sprague-Dawley , Signal Transduction , U937 Cells
6.
Free Radic Biol Med ; 87: 58-68, 2015 Oct.
Article En | MEDLINE | ID: mdl-26117323

Hydrogen (H(2)) attenuates the development of atherosclerosis in mouse models. We aimed to examine the effects of H(2) on atherosclerotic plaque stability. Low-density lipoprotein receptor-knockout (LDLR(-/-)) mice fed an atherogenic diet were dosed daily with H(2) and/or simvastatin. In vitro studies were carried out in an oxidized-LDL (ox-LDL)-stimulated macrophage-derived foam cell model treated with or without H(2). H(2) or simvastatin significantly enhanced plaque stability by increasing levels of collagen, as well as reducing macrophage and lipid levels in plaques. The decreased numbers of dendritic cells and increased numbers of regulatory T cells in plaques further supported the stabilizing effect of H(2) or simvastatin. Moreover, H(2) treatment decreased serum ox-LDL level and apoptosis in plaques with concomitant inhibition of endoplasmic reticulum stress (ERS) and reduction of reactive oxygen species (ROS) accumulation in the aorta. In vitro, like the ERS inhibitor 4-phenylbutyric acid, H(2) inhibited ox-LDL- or tunicamycin (an ERS inducer)-induced ERS response and cell apoptosis. In addition, like the ROS scavenger N-acetylcysteine, H(2) inhibited ox-LDL- or Cu(2+) (an ROS inducer)-induced reduction in cell viability and increase in cellular ROS. Also, H(2) increased Nrf2 (NF-E2-related factor-2, an important factor in antioxidant signaling) activation and Nrf2 small interfering RNA abolished the protective effect of H(2) on ox-LDL-induced cellular ROS production. The inhibitory effects of H(2) on the apoptosis of macrophage-derived foam cells, which take effect by suppressing the activation of the ERS pathway and by activating the Nrf2 antioxidant pathway, might lead to an improvement in atherosclerotic plaque stability.


Atherosclerosis/metabolism , NF-E2-Related Factor 2/biosynthesis , Plaque, Atherosclerotic/metabolism , Receptors, LDL/genetics , Receptors, Oxidized LDL/metabolism , Animals , Apoptosis/drug effects , Atherosclerosis/diet therapy , Atherosclerosis/genetics , Atherosclerosis/pathology , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Humans , Hydrogen/administration & dosage , Hydrogen/metabolism , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics , Plaque, Atherosclerotic/diet therapy , Plaque, Atherosclerotic/pathology , Reactive Oxygen Species/metabolism , Receptors, LDL/metabolism , Receptors, Oxidized LDL/genetics , Simvastatin/administration & dosage
7.
FASEB J ; 29(8): 3342-56, 2015 Aug.
Article En | MEDLINE | ID: mdl-25877213

The angiotensin II type 1 receptor (AT1) is a 7-transmembrane domain GPCR that when activated by its ligand angiotensin II, generates signaling events promoting vascular dysfunction and the development of cardiovascular disease. Here, we show that the single-transmembrane oxidized LDL (oxLDL) receptor (LOX-1) resides in proximity to AT1 on cell-surface membranes and that binding of oxLDL to LOX-1 can allosterically activate AT1-dependent signaling events. oxLDL-induced signaling events in human vascular endothelial cells were abolished by knockdown of AT1 and inhibited by AT1 blockade (ARB). oxLDL increased cytosolic G protein by 350% in Chinese hamster ovary (CHO) cells with genetically induced expression of AT1 and LOX-1, whereas little increase was observed in CHO cells expressing only LOX-1. Immunoprecipitation and in situ proximity ligation assay (PLA) assays in CHO cells revealed the presence of cell-surface complexes involving LOX-1 and AT1. Chimeric analysis showed that oxLDL-induced AT1 signaling events are mediated via interactions between the intracellular domain of LOX-1 and AT1 that activate AT1. oxLDL-induced impairment of endothelium-dependent vascular relaxation of vascular ring from mouse thoracic aorta was abolished by ARB or genetic deletion of AT1. These findings reveal a novel pathway for AT1 activation and suggest a new mechanism whereby oxLDL may be promoting risk for cardiovascular disease.


Lectins/metabolism , Lipoproteins, LDL/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptors, Oxidized LDL/metabolism , Animals , CHO Cells , COS Cells , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Cricetulus , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Humans , Signal Transduction/physiology
8.
J Mol Cell Cardiol ; 84: 36-44, 2015 Jul.
Article En | MEDLINE | ID: mdl-25871829

Chronic kidney disease (CKD), an independent risk factor for cardiovascular disease, is associated with abnormal lipoprotein metabolism. We examined whether electronegative low-density lipoprotein (LDL) is mechanistically linked to cardiac dysfunction in patients with early CKD. We compared echocardiographic parameters between patients with stage 2 CKD (n = 88) and normal controls (n = 89) and found that impaired relaxation was more common in CKD patients. Reduction in estimated glomerular filtration rate was an independent predictor of left ventricular relaxation dysfunction. We then examined cardiac function in a rat model of early CKD induced by unilateral nephrectomy (UNx) by analyzing pressure-volume loop data. The time constant of isovolumic pressure decay was longer and the maximal velocity of pressure fall was slower in UNx rats than in controls. When we investigated the mechanisms underlying relaxation dysfunction, we found that LDL from CKD patients and UNx rats was more electronegative than LDL from their respective controls and that LDL from UNx rats induced intracellular calcium overload in H9c2 cardiomyocytes in vitro. Furthermore, chronic administration of electronegative LDL, which signals through lectin-like oxidized LDL receptor-1 (LOX-1), induced relaxation dysfunction in wild-type but not LOX-1(-/-) mice. In in vitro and in vivo experiments, impaired cardiac relaxation was associated with increased calcium transient resulting from nitric oxide (NO)-dependent nitrosylation of SERCA2a due to increases in inducible NO synthase expression and endothelial NO synthase uncoupling. In conclusion, LDL becomes more electronegative in early CKD. This change disrupts SERCA2a-regulated calcium homeostasis, which may be the mechanism underlying cardiorenal syndrome.


Calcium/metabolism , Homeostasis , Lipoproteins, LDL/metabolism , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/physiopathology , Adult , Animals , Case-Control Studies , Demography , Female , Fibrosis , Heart , Humans , Male , Mice, Inbred C57BL , Models, Biological , Myocytes, Cardiac/metabolism , Nephrectomy , Nitric Oxide Synthase Type II/metabolism , Nitrosation , Rats, Sprague-Dawley , Receptors, Oxidized LDL/metabolism , Renal Insufficiency, Chronic/diagnostic imaging , Renin-Angiotensin System , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Ultrasonography , Up-Regulation , Vasodilation , tau Proteins/metabolism
9.
J Transl Med ; 12: 76, 2014 Mar 22.
Article En | MEDLINE | ID: mdl-24655356

BACKGROUND: In kidney transplantation, the prevalence of hypercholesterolemia as a co-morbidity factor known to affect graft function, is rising due to the increased number of older donors in response to organ shortage as well as to the hyperlipidemic effects of immunosuppressors in recipient. This study aimed to characterize the effects of hypercholesterolemia on renal graft outcome, investigating the role of oxidized low-density lipoprotein (OxLDL). METHODS: In vivo, we used a porcine preclinical model of renal auto-transplantation modulated by two experimental diets: a normal (n = 6) or a hyperlipidemic diet (n = 5) maintained during the 3 month follow-up after the surgical procedure. Kidney function and OxLDL levels were monitored as well as fibrosis, LOX-1 and TGF beta signaling pathways. In vitro, we used human artery endothelial cells subjected to OxLDL to investigate the TGF beta profibrotic pathway and the role of the scavenger receptor LOX-1. RESULTS: Hyperlipidemic diet-induced increase in plasma OxLDL levels at the time of surgery correlated with an increase in proteinuria 3 months after transplantation, associated with an early graft fibrosis combined with an activation of renal TGF beta signaling. These data suggest a direct involvement of OxLDL in the hyperlipidemic diet-induced activation of the pro-fibrotic TGF beta pathway which seems to be activated by LOX-1 signaling. These results were supported by studies with endothelial cells incubated in culture medium containing OxLDL promoting TGF beta expression inhibited by LOX-1 antibody. CONCLUSIONS: These results implicate OxLDL in the hyperlipidemic diet-promoted fibrosis in transplanted kidneys, suggesting LOX-1 as a potential therapeutic target and reinforce the need to control cholesterol levels in kidney transplant recipients.


Diet, High-Fat/adverse effects , Kidney Transplantation , Lipoproteins, LDL/blood , Animals , Antibodies, Blocking/pharmacology , Arteries/pathology , Cholesterol/blood , Creatinine/blood , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium/drug effects , Endothelium/metabolism , Endothelium/pathology , Fibrosis , Humans , Kidney/physiopathology , Kidney Function Tests , Male , Models, Animal , Proteinuria/blood , Proteinuria/complications , Proteinuria/pathology , Receptors, Oxidized LDL/metabolism , Signal Transduction/drug effects , Sus scrofa , Transforming Growth Factor beta/metabolism , Transplantation, Autologous , Vimentin/metabolism
10.
Zhonghua Yi Xue Za Zhi ; 93(42): 3388-93, 2013 Nov 12.
Article Zh | MEDLINE | ID: mdl-24418038

OBJECTIVE: To explore the effects of nicotinic acid intervention on vascular endothelial dysfunction mediated by oxidized low density lipoprotein (ox-LDL)/lectin-like oxidized low density lipoprotein receptor-1 (LOX-1) in diet-induced obese immature rats and its possible mechanism through detecting the expression levels of ox-LDL and LOX-1 in abdominal aorta. METHODS: A model of diet-induced obese immature rats was established by high-fat diet. And 30 immature rats were divided randomly and equally into control (n = 10), high-fat (n = 10) and drug control (n = 10) groups. At the end of 12 weeks, the levels of serum total cholesterol (TC), triglyceride (TG), LDL and high-density lipoprotein (HDL) were examined. The levels of ox-LDL, soluble intercellular adhesion molecule-1 (sICAM-1), endothelin and nitric oxide (NO) were detected. The gene and protein expressions of LOX-1 and ICAM-1 in abdominal aorta were detected. And the location protein expressions of LOX-1 and ICAM-1 were examined. RESULTS: High-fat diet induced hyperlipidemia and obesity in immature rats. The serum levels of TG, TC, LDL, ox-LDL and endothelin in high-fat and drug control groups were all higher than control group ((0.98 ± 0.12) and (0.69 ± 0.06) vs (0.49 ± 0.06) mmol/L, (2.11 ± 0.16) and (1.62 ± 0.12) vs (1.30 ± 0.12) mmol/L, (0.71 ± 0.04) and (0.50 ± 0.03) vs (0.30 ± 0.04) mmol/L, (44.2 ± 5.1) and (33.7 ± 2.1) vs (26.6 ± 2.9) µg/L, (187 ± 10) and (157 ± 6) vs (118 ± 7) pg/ml). The indices in high-fat group were higher than those in drug control group (all P < 0.01) . The levels of HDL and NO in high-fat and drug control groups were lower than those in control group (all P < 0.01); the levels of HDL and NO in high-fat group lower than those in drug control group (all P < 0.01). And the levels of LOX-1, ICAM-1 protein and mRNA in high-fat group were higher than those in drug control and control groups (all P < 0.01).ox-LDL was correlated positively with LOX-1, TC, TG, LDL, endothelin and ICAM-1 (r = 0.918, 0.867, 0.857, 0.834, 0.869, 0.644, all P < 0.01) , but negatively with NO and HDL (r = -0.823, -0.872, P < 0.01) . CONCLUSION: Early treatment of nicotinic acid can protect endothelial function through inducing therapeutic effects on hyperlipidemia and antioxidation and down-regulating the expression level of ox-LDL/LOX-1 in vascular endothelium.


Endothelium, Vascular/physiopathology , Hyperlipidemias/physiopathology , Lipoproteins, LDL/metabolism , Niacin/pharmacology , Obesity/metabolism , Animals , Endothelium, Vascular/metabolism , Hyperlipidemias/metabolism , Intercellular Adhesion Molecule-1/metabolism , Male , Nitric Oxide/metabolism , Obesity/physiopathology , Rats , Rats, Wistar , Receptors, Oxidized LDL/metabolism
11.
Curr Opin Lipidol ; 23(5): 439-45, 2012 Oct.
Article En | MEDLINE | ID: mdl-22777292

PURPOSE OF REVIEW: LOX-1 is a multiligand receptor implicated in endothelial dysfunction and atherosclerosis, although it was originally identified as an oxidized LDL receptor. In this review, the roles of various LOX-1 ligands and their interaction with LOX-1 are discussed to understand the pathophysiological significance of LOX-1. RECENT FINDINGS: LOX-1 knockout mice showed resistance of endothelium-dependent vasorelaxation against oxidized LDL and retardation of atherosclerosis progression. LOX-1 ligand reduction in mice also attenuated atherosclerosis progression. In a human cohort study, high concentration of apoB-containing LOX-1 ligands predicted the incidence of cardiovascular disease. Furthermore, modified HDL, which existed in high concentration in the plasma of coronary artery disease patients, was found to induce impairment of endothelial nitric oxide release via LOX-1. In addition to lipoproteins, LOX-1 was found to work as a C-reactive protein receptor providing a scaffold for the activation of the complement system. SUMMARY: LOX-1 is a unique molecule among the sensors of danger signals. LOX-1 is not only sensing danger signals such as modified LDL and heat shock protein, but also scaffolding other danger sensors including C-reactive protein and C1q, and directly commanding responses to danger signals by working as a cell adhesion molecule. Via these functions, LOX-1 might work as a surveillance molecule of vascular homeostasis.


Arteriosclerosis/physiopathology , Scavenger Receptors, Class E/metabolism , Signal Transduction , Animals , Apolipoproteins B/metabolism , C-Reactive Protein/metabolism , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Complement Activation , Disease Progression , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Humans , Ligands , Mice , Mice, Knockout , Receptors, Oxidized LDL/metabolism
12.
Microbes Infect ; 14(1): 43-9, 2012 Jan.
Article En | MEDLINE | ID: mdl-21911078

The association of Chlamydia pneumoniae and atherosclerosis has been well documented. Recently, it has been demonstrated that C. pneumoniae up-regulates expression of the lectin-like ox-LDL receptor (LOX-1) in endothelial cells. Many of the pro-atherogenic effects of ox-LDL occur through its activation and uptake by LOX-1. This class E scavenger receptor contains a carbohydrate-recognition domain common to the C type lectin family. Previously, we have demonstrated that the major outer membrane protein of the chlamydiae is glycosylated and glycan removal abrogates infectivity of C. pneumoniae for endothelial cells. In this study, we investigated whether C. pneumoniae binds to LOX-1. The results show that 1) infection of endothelial cells by C. pneumoniae is inhibited by ligands that bind to the LOX-1 receptor, but not by ligands binding to other scavenger receptors; 2) anti-LOX-1 antibody inhibits C. pneumoniae infectivity, while antibodies against other scavenger receptors do not; 3) anti-LOX-1 antibody inhibits attachment of C. pneumoniae to endothelial cells; and 4) C. pneumoniae co-localizes with LOX-1. These effects were not observed for Chlamydia trachomatis. In conclusion, C. pneumoniae binds to the LOX-1 receptor, which is known to promote atherosclerosis.


Atherosclerosis/metabolism , Atherosclerosis/microbiology , Chlamydophila pneumoniae/metabolism , Receptors, Oxidized LDL/metabolism , Scavenger Receptors, Class E/metabolism , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Atherosclerosis/pathology , Cell Line , Chlamydophila pneumoniae/pathogenicity , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Gene Expression Regulation, Bacterial , HeLa Cells , Humans , Lectins/metabolism , Scavenger Receptors, Class E/antagonists & inhibitors , Scavenger Receptors, Class E/immunology , Up-Regulation
13.
Zhonghua Yi Xue Za Zhi ; 91(31): 2216-20, 2011 Aug 23.
Article Zh | MEDLINE | ID: mdl-22094043

OBJECTIVE: Our previous studies found that 100 µg/ml oxidized low-density lipoprotein (ox-LDL) could up-regulate the autophagic level in human umbilical vein endothelial cells (HUVEC). The present study was conducted to observe the roles of oxidative stress and lectin-like oxidized low density lipoprotein-1 (LOX-1) in the ox-LDL-induced up-regulation of autophagy. METHODS: Prior to the ox-LDL exposure, LOX-1mAb, vitamin C and vitamin E were used to study the roles of LOX-1 and oxidative stress in the activation of autophagy. The contents of total-superoxide dismutase (T-SOD) and MDA (malondialdehyde) in the culture medium were detected with enzyme linked immunosorbent assay. Western blot was employed to detect the levels of autophagic marker microtubule-associated protein light chain 3 (MAP1-LC3)-II/LC3-I, beclin1 and lysosome associated membrane protein 2a (lamp2a). RESULTS: After the ox-LDL exposure, the down-regulated level of T-SOD [0.5 h (32.73 ± 1.09 vs 40.16 ± 1.28) U/ml, P < 0.01; 6 h (29.32 ± 1.56 vs 40.16 ± 1.28) U/ml, P < 0.01] and the up-regulated level of MDA [0.5 h (1.11 ± 0.04 vs 0.57 ± 0.05) nmol/ml, P < 0.01; 6 h (0.69 ± 0.03 vs 0.57 ± 0.05) nmol/ml, P < 0.05] in culture medium were also significant at 0.5 h and 6 h. The ox-LDL-induced increased ratio of LC3-II/LC3-I was reversed by the pretreatments of vitamin C and vitamin E (0.5 h, vitC: 3.11 ± 0.02 vs 4.31 ± 0.50, P < 0.05; vitE: 3.46 ± 0.19 vs 4.31 ± 0.50, P < 0.05; 6 h, vitC: 1.44 ± 0.05 vs 2.31 ± 0.16, P < 0.05), but not LOX-1mAb. LOX-1mAb decreased the ox-LDL-induced elevated level of lamp2a protein while vitamin C and vitamin E only inhibited the elevation of lamp2a at the timepoint of 6 h, but not 0.5 h. CONCLUSION: Oxidative stress, rather than LOX-1, plays an important role in the ox-LDL-induced up-regulation of autophagy in HUVEC. The formation of autolysosomes is associated with the LOX-1-mediated endocytosis of ox-LDL. Oxidative stress only plays a minor role in the formation of autolysosomes induced by the engulfed ox-LDL.


Autophagy , Human Umbilical Vein Endothelial Cells/metabolism , Lipoproteins, LDL/metabolism , Oxidative Stress , Receptors, Oxidized LDL/metabolism , Cell Line , Humans , Plant Lectins , Up-Regulation
14.
J Lipid Res ; 52(2): 299-307, 2011 Feb.
Article En | MEDLINE | ID: mdl-21078775

Palmitic acid (PA) upregulates oxidized LDL receptor-1 (LOX-1), a scavenger receptor responsible for uptake of oxidized LDL (oxLDL), and enhances oxLDL uptake in macrophages. However, the precise underlying mechanism remains to be elucidated. PA is known to induce endoplasmic reticulum (ER) stress in various cell types. Therefore, we investigated whether ER stress is involved in PA-induced LOX-1 upregulation. PA induced ER stress, as determined by phosphorylation of PERK, eIF2α, and JNK, as well as induction of CHOP in macrophage-like THP-1 cells. Inhibitors [4-phenylbutyric acid (PBA), sodium tauroursodeoxycholate (TUDCA), and salubrinal] and small interfering RNA (siRNA) for the ER stress response decreased PA-induced LOX-1 upregulation. Thapsigargin, an ER stress inducer, upregulated LOX-1, which was decreased by PBA and TUDCA. We next examined whether unsaturated FAs could counteract the effect of PA. Both oleic acid (OA) and linoleic acid (LA) suppressed PA-induced LOX-1. Activation of the ER stress response observed in the PA-treated cells was markedly attenuated when the cells were cotreated with OA or LA. In addition, OA and LA suppressed thapsigargin-induced LOX-1 upregulation with reduced activation of ER stress markers. Our results indicate that activation of ER stress is involved in PA-induced LOX-1 upregulation in macrophages, and that OA and LA inhibit LOX-1 induction through suppression of ER stress.


Endoplasmic Reticulum/drug effects , Fatty Acids, Unsaturated/pharmacology , Palmitic Acid/pharmacology , Receptors, Oxidized LDL/metabolism , Animals , Cell Line , Humans , Phenylbutyrates/pharmacology , RNA, Small Interfering/pharmacology , Stress, Physiological/drug effects , Thapsigargin/pharmacology , Up-Regulation
15.
Am J Chin Med ; 38(6): 1161-9, 2010.
Article En | MEDLINE | ID: mdl-21061468

This study investigates the effects of beriberine on the expression of lectin-like ox-LDL receptor-1 (LOX-1), scavenger receptor A (SR-A), SR class B type I (SR-BI) and ATP-binding cassette transporter A1 (ABCA1) in human macrophage-derived foam cells induced by ox-LDL. Different concentrations of Berberine were co-cultured with THP-1 derived foam cells. The mRNA and protein expressions of LOX-1, SR-A, SR-BI and ABCA1 were determined by RT-PCR and Western blot analysis, respectively. Ox-LDL significantly increased the expression of LOX-1 and inhibited the expression of SR-BI in a dose- and time-dependent manner. Berberine significantly inhibited the effects of ox-LDL in a dose- and time-dependent manner. Moreover, ox-LDL significantly promoted ABCA1 expression. However, berberine had no effect on SR-A or ABCA1 expression. Berberine can inhibit the expression of LOX-1 and promote the expression of SR-BI in macrophage-derived foam cells. Therefore, berberine could be used to treat atherosclerotic diseases.


Atherosclerosis/drug therapy , Berberine/pharmacology , Drugs, Chinese Herbal/pharmacology , Foam Cells/drug effects , Macrophages/drug effects , Receptors, Oxidized LDL/metabolism , Scavenger Receptors, Class B/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Berberine/therapeutic use , Blotting, Western , Cell Line , Coptis/chemistry , Dose-Response Relationship, Drug , Foam Cells/metabolism , Humans , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Macrophages/metabolism , Phytotherapy , RNA, Messenger/metabolism , Receptors, Oxidized LDL/genetics , Reverse Transcriptase Polymerase Chain Reaction , Rhizome , Scavenger Receptors, Class A/genetics , Scavenger Receptors, Class A/metabolism , Scavenger Receptors, Class B/genetics
16.
Circ Cardiovasc Imaging ; 3(4): 464-72, 2010 Jul.
Article En | MEDLINE | ID: mdl-20442371

BACKGROUND: The oxidized low-density lipoprotein receptor (LDLR) LOX-1 plays a crucial role in atherosclerosis. We sought to detect and assess atherosclerotic plaque in vivo by using single-photon emission computed tomography/computed tomography and magnetic resonance imaging and a molecular probe targeted at LOX-1. METHODS AND RESULTS: Apolipoprotein E(-/-) mice fed a Western diet and LDLR(-/-) and LDLR(-/-)/LOX-1(-/-) mice fed an atherogenic diet were used. Imaging probes consisted of liposomes decorated with anti-LOX-1 antibodies or nonspecific immunoglobulin G, (111)indium or gadolinium, and 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine fluorescence markers. In vivo imaging was performed 24 hours after intravenous injection (150 microL) of LOX-1 or nonspecific immunoglobulin G probes labeled with either (111)indium (600 muCi) or gadolinium (0.075 mmol/kg), followed by aortic excision for phosphor imaging and Sudan IV staining, or fluorescence imaging and hematoxylin/eosin staining. The LOX-1 probe also colocalized with specific cell types, apoptosis, and matrix metalloproteinase-9 expression in frozen aortic sections. Single-photon emission computed tomography/computed tomography imaging of the LOX-1 probe showed aortic arch "hot spots" in apolipoprotein E(-/-) mice (n=8), confirmed by phosphor imaging. Magnetic resonance imaging showed significant Gd enhancement in atherosclerotic plaques in LDLR(-/-) mice with the LOX-1 (n=7) but not with the nonspecific immunoglobulin G (n=5) probe. No signal enhancement was observed in LDLR(-/-)/LOX-1(-/-) mice injected with the LOX-1 probe (n=5). These results were confirmed by ex vivo fluorescence imaging. The LOX-1 probe bound preferentially to the plaque shoulder, a region with vulnerable plaque features, including extensive LOX-1 expression, macrophage accumulation, apoptosis, and matrix metalloproteinase-9 expression. CONCLUSIONS: LOX-1 can be used as a target for molecular imaging of atherosclerotic plaque in vivo. Furthermore, the LOX-1 imaging signal is associated with markers of rupture-prone atherosclerotic plaque.


Atherosclerosis/diagnosis , Atherosclerosis/metabolism , Magnetic Resonance Imaging , Molecular Imaging/methods , Receptors, Oxidized LDL/metabolism , Scavenger Receptors, Class E/metabolism , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed , Analysis of Variance , Animals , Atherosclerosis/diagnostic imaging , Gadolinium , Immunoblotting , Immunoglobulin G/chemistry , In Situ Nick-End Labeling , Indium Radioisotopes , Liposomes , Mice , Microscopy, Confocal , Radiopharmaceuticals/pharmacology , Staining and Labeling
17.
Biosci Biotechnol Biochem ; 74(2): 279-84, 2010.
Article En | MEDLINE | ID: mdl-20139626

In Leonurus sibiricus herb extract (LHE)-supplemented animals, plasma cholesterol decreased and high-density lipoprotein-cholesterol increased, resulting in a lowered atherogenic index. The plasma trolox equivalent antioxidant capacity, levels of hepatic thiobarbituric acid-reactive substances, and protein carbonyl values decreased significantly in LHE-supplemented mice (p<0.05), whereas the hepatic antioxidant indicators were all significantly elevated (p<0.05). In human umbilical vein endothelial cells stimulated with tumor necrosis factor alpha, LHE significantly suppressed intracellular reactive oxygen species, LOX-1, and adhesion molecules. LHE supplementation may modulate the lipoprotein composition and attenuate oxidative stress by elevated antioxidant processes, thus suppressing the activation of inflammatory mediators. This is a possible mechanism of the anti-atherogenic effect.


Cell Adhesion Molecules/metabolism , Endothelial Cells/drug effects , Hypercholesterolemia/metabolism , Leonurus/metabolism , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Receptors, Oxidized LDL/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cells, Cultured , Female , Humans , Lectins/metabolism , Mice , Umbilical Veins/cytology
18.
Clin Chem ; 56(3): 478-81, 2010 Mar.
Article En | MEDLINE | ID: mdl-20075180

BACKGROUND: C-reactive protein (CRP) increases in response to inflammation and is purported to be a risk factor for atherogenesis. We recently demonstrated that a scavenger receptor, lectin-like oxidized LDL receptor (LOX-1), is a receptor for CRP. In light of the overlapping ligand spectrum of scavenger receptors such as modified LDL, bacteria, and advanced glycation end products, we examined whether other scavenger receptors recognize CRP. METHODS: We analyzed the uptake of fluorescently labeled CRP in COS-7 cells expressing a series of scavenger receptors and in a monocytic cell line, THP-1, differentiated into macrophage with phorbol 12-myristate 13-acetate (PMA). We applied small interfering RNA (siRNA) against class-A scavenger receptor (SR-A) to THP-1 cells to suppress the expression of SR-A. We also analyzed the binding of nonlabeled CRP to immobilized recombinant LOX-1 and SR-A in vitro using anti-CRP antibody. RESULTS: COS-7 cells expressing LOX-1 and SR-A internalized fluorescently labeled CRP in a dose-dependent manner, but cells expressing CD36, SR-BI, or CD68 did not. The recombinant LOX-1 and SR-A proteins recognized nonlabeled purified CRP and native CRP in serum in vitro. THP-1 cells differentiated into macrophage-like cells by treatment with PMA-internalized fluorescently labeled CRP. siRNA against SR-A significantly and concomitantly inhibited the expression of SR-A (P < 0.01) and CRP uptake (P < 0.01), whereas control siRNA did not. CONCLUSIONS: CRP is recognized by SR-A as well as LOX-1 and taken up via SR-A in a macrophage-like cell line. This process might be of significance in the pathogenesis of atherosclerotic disease.


C-Reactive Protein/metabolism , Macrophages/metabolism , Protein Transport , Scavenger Receptors, Class A/metabolism , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Cricetinae , Down-Regulation , Humans , Protein Binding , RNA, Small Interfering/genetics , Receptors, Oxidized LDL/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Scavenger Receptors, Class A/genetics
19.
Arterioscler Thromb Vasc Biol ; 29(10): 1412-8, 2009 Oct.
Article En | MEDLINE | ID: mdl-19759373

Monocytes are central mediators in the advance of atherosclerotic plaque, making them a natural therapeutic target for reducing disease burden. Here, we highlight recent advances in our current understanding of monocyte heterogeneity and its relevance to regulation of monocyte accumulation and function within atherosclerotic plaques. Differences that distinguish monocyte subsets include differential expression of chemokine receptors, especially CCR2 and CX3CR1. Ablation of expression of these 2 receptors (or their ligands) in mice has an additive inhibition on monocyte recruitment to atherosclerotic plaques. Moreover, simultaneously interfering with 3 key pathways--CCR2, CX3CR1, and CCR5--essentially abolishes atherosclerosis in mice. Here, we discuss how these chemokine receptors act at multiple points on at least 1 monocyte subset, regulating their mobilization from bone marrow, survival, or recruitment to plaques. Finally, we discuss how this knowledge may be useful clinically, emphasizing that CX3CR1 may in particular be a viable target for therapeutic manipulation of monocyte-derived cell fate in cardiovascular disease.


Atherosclerosis/etiology , Monocytes/physiology , Receptors, Chemokine/physiology , Animals , CX3C Chemokine Receptor 1 , Cell Movement , Cell Survival , Chemokine CXCL16 , Chemokines, CXC/physiology , Humans , Monocytes/classification , Receptors, CCR2/physiology , Receptors, Oxidized LDL/metabolism , Receptors, Scavenger/physiology
...