Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Bioorg Med Chem Lett ; 106: 129762, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38649117

ABSTRACT

Lipids play an important role in varying vital cellular processes including cell growth and division. Elevated levels of low-density lipoprotein (LDL) and oxidized-LDL (ox-LDL), and overexpression of the corresponding receptors including LDL receptor (LDLR), lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1), and cluster of differentiation 36 (CD36), have shown strong correlations with different facets of carcinogenesis including proliferation, invasion, and angiogenesis. Furthermore, a high serum level of LOX-1 is considered as a poor prognostic factor in many types of cancer including colorectal cancer. Ox-LDL could contribute to cancer progression and metastasis through endothelial-to-mesenchymal transition (EMT) and autophagy. Thus, many studies have shed light on the significant role of ox-LDL as a potential therapeutic target for cancer therapy. In various repurposing approaches, anti-dyslipidemia agents, phytochemicals, autophagy modulators as well as recently developed ldl-like nanoparticles have been investigated as potential tumor therapeutic agents by targeting oxidized-LDL/LOX-1 pathways. Herein, we reviewed the role of oxidized-LDL and LOX-1 in cancer progression, invasion, metastasis, and also cancer-associated angiogenesis. Moreover, we addressed therapeutic utility of several compounds that proved to be capable of targeting the metabolic moieties in cancer. This review provides insights on the potential impact of targeting LDL and ox-LDL in cancer therapy and their future biomedical implementations.


Subject(s)
Lipoproteins, LDL , Neoplasms , Humans , Lipoproteins, LDL/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Scavenger Receptors, Class E/metabolism , Scavenger Receptors, Class E/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Animals
2.
Cell Rep ; 36(12): 109750, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34551309

ABSTRACT

The inappropriate use of antibiotics is a severe public health problem worldwide, contributing to the emergence of multidrug-resistant (MDR) bacteria. To explore the possible impacts of the inappropriate use of antibiotics on the immune system, we use Klebsiella pneumoniae (K. pneumoniae) infection as an example and show that imipenem increases the mortality of mice infected by MDR K. pneumoniae. Further studies demonstrate that imipenem enhances the secretion of outer membrane vesicles (OMVs) with significantly elevated presentation of GroEL, which promotes the phagocytosis of OMVs by macrophages that depends on the interaction between GroEL and its receptor, lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1). OMVs cause the pyroptosis of macrophages and the release of proinflammatory cytokines, which contribute to exacerbated inflammatory responses. We propose that the inappropriate use of antibiotics in the cases of infection by MDR bacteria such as K. pneumoniae might cause damaging inflammatory responses, which underlines the pernicious effects of inappropriate use of antibiotics.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Multiple, Bacterial/drug effects , Extracellular Vesicles/metabolism , Klebsiella pneumoniae/pathogenicity , Pyroptosis , Animals , Bacterial Proteins/metabolism , Carbapenems/pharmacology , Chaperonin 60/metabolism , Cytokines/metabolism , Inflammation/metabolism , Inflammation/pathology , Inflammation/veterinary , Klebsiella Infections/mortality , Klebsiella Infections/pathology , Klebsiella Infections/veterinary , Klebsiella pneumoniae/metabolism , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Phagocytosis , RNA Interference , RNA, Small Interfering/metabolism , Scavenger Receptors, Class E/antagonists & inhibitors , Scavenger Receptors, Class E/genetics , Scavenger Receptors, Class E/metabolism , Survival Rate
3.
Arterioscler Thromb Vasc Biol ; 41(1): 153-166, 2021 01.
Article in English | MEDLINE | ID: mdl-33176449

ABSTRACT

Atherosclerosis, the underlying cause of cardiovascular disease (CVD), is a worldwide cause of morbidity and mortality. Reducing ApoB-containing lipoproteins-chiefly, LDL (low-density lipoprotein)-has been the main strategy for reducing CVD risk. Although supported by large randomized clinical trials, the persistence of residual cardiovascular risk after effective LDL reduction has sparked an intense search for other novel CVD biomarkers and therapeutic targets. Recently, Lox-1 (lectin-type oxidized LDL receptor 1), an innate immune scavenger receptor, has emerged as a promising target for early diagnosis and cardiovascular risk prediction and is also being considered as a treatment target. Lox-1 was first described as a 50 kDa transmembrane protein in endothelial cells responsible for oxLDL (oxidized LDL) recognition, triggering downstream pathways that intensify atherosclerosis via endothelial dysfunction, oxLDL uptake, and apoptosis. Lox-1 is also expressed in platelets, where it enhances platelet activation, adhesion to endothelial cells, and ADP-mediated aggregation, thereby favoring thrombus formation. Lox-1 was also identified in cardiomyocytes, where it was implicated in the development of cardiac fibrosis and myocyte apoptosis, the main determinants of cardiac recovery following an ischemic insult. Together, these findings have revealed that Lox-1 is implicated in all the main steps of atherosclerosis and has encouraged the development of immunoassays for measurement of sLox-1 (serum levels of soluble Lox-1) to be used as a potential CVD biomarker. Finally, the recent development of synthetic Lox-1 inhibitors and neutralizing antibodies with promising results in animal models has made Lox-1 a target for drug development. In this review, we discuss the main findings regarding the role of Lox-1 in the development, diagnosis, and therapeutic strategies for CVD prevention and treatment.


Subject(s)
Cardiovascular Diseases/blood , Scavenger Receptors, Class E/blood , Animals , Antibodies, Neutralizing/therapeutic use , Biomarkers/blood , Cardiovascular Agents/therapeutic use , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/epidemiology , Heart Disease Risk Factors , Humans , Predictive Value of Tests , Prognosis , Risk Assessment , Scavenger Receptors, Class E/antagonists & inhibitors
4.
Int J Med Sci ; 17(8): 1056-1061, 2020.
Article in English | MEDLINE | ID: mdl-32410835

ABSTRACT

The aim of this study was to determine whether interleukin-1ß (IL-1ß) promotes oxidised low-density lipoprotein (Ox-LDL) uptake by human glomerular mesangial cells (HMCs) and its effect on the expression of lectin-like Ox-LDL receptor 1 (LOX-1) and to identify pathways through which IL-1ß affects lipid uptake. Confocal laser scanning microscopy and flow cytometry were used to observe the effect of IL-1ß on lipid uptake by HMCs and the pathway by which IL-1ß may mediate lipid uptake. Real-time polymerase chain reaction (PCR) and western blotting were used to evaluate the effect of IL-1ß on LOX-1 expression. Confocal laser scanning microscopy and flow cytometry revealed that IL-1ß promoted uptake of fluorescent Dil-labelled Ox-LDL(Dil-Ox-LDL) by HMCs and the enhanced uptake of Dil-Ox-LDL was partially inhibited by an anti-LOX-1 antibody evaluated by flow cytometry. Further, IL-1ß promoted LOX-1 mRNA and protein expression of HMCs in a dose- and time-dependent manner. Thus, Ox-LDL is ingested by HMCs under basic conditions. Inflammatory cytokine IL-1ß promotes Ox-LDL uptake by HMCs. Furthermore, IL-1ß promotes the mRNA and protein expression of LOX-1, a specific receptor of Ox-LDL, suggesting that the enhancement of Ox-LDL uptake may be mediated by LOX-1 pathway. Anti-LOX-1 therapy may be a promising option for treatment of glomerulosclerosis.


Subject(s)
Interleukin-1beta/metabolism , Lipoproteins, LDL/metabolism , Mesangial Cells/metabolism , Scavenger Receptors, Class E/metabolism , Cell Line , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/immunology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Flow Cytometry , Humans , Lipid Metabolism/drug effects , Lipid Metabolism/immunology , Mesangial Cells/immunology , Mesangial Cells/ultrastructure , Microscopy, Confocal , Scavenger Receptors, Class E/antagonists & inhibitors
5.
Int Immunopharmacol ; 80: 106190, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31945611

ABSTRACT

PURPOSE: To explore the expression and role of lectin-like oxidized low-density lipoprotein receptor 1 (LOX-1) in retinal degeneration. METHODS: The retinal degeneration of BALB/c mice was induced by light exposure. BV2 cells were activated by LPS stimulation. Retinas or BV2 cells were pretreated with LOX-1 neutralizing antibody or Polyinosinic acid (PolyI) (the inhibitor of LOX-1) before light damage (LD) or LPS stimulation. LOX-1, TNF-α, IL-1ß, CCL2 and NF-κB expression were detected in retinas or BV2 cells by real-time RT-PCR, western blot or ELISA. Histological analyses of retinas were performed. Photoreceptor cell death was assessed by TUNEL assay in retinas or by flow cytometry in 661W cells cultured in microglia-conditioned medium. RESULTS: Photoreceptor cell death and elevated expression of LOX-1 were induced by LD in retinas of BALB/c mice. LOX-1 neutralizing antibody or PolyI pretreatment significantly reduced the elevated expression of LOX-1, TNF-α, IL-1ß, CCL2 and p-NF-κB caused by LD in retinas. Inhibition of LOX-1 by LOX-1 neutralizing antibody or PolyI significantly reduced photoreceptor cell death induced by LD in retinas. Elevated levels of TNF-α, IL-1ß and CCL2 caused by LPS were down-regulated by inhibition of LOX-1 in BV2 cells. Inhibition of LOX-1 reduces microglial neurotoxicity on photoreceptors. CONCLUSIONS: LOX-1 expression is increased in light induced retinal degeneration, what's more, inhibition of LOX-1 prevents inflammation and photoreceptor cell death in retinal degeneration and reduces microglial neurotoxicity on photoreceptors. Therefore, LOX-1 can be used as a potential therapeutic target for such retinal degeneration diseases.


Subject(s)
Antibodies, Neutralizing/pharmacology , Photoreceptor Cells/drug effects , Retinal Degeneration/drug therapy , Scavenger Receptors, Class E/antagonists & inhibitors , Animals , Antibodies, Neutralizing/therapeutic use , Apoptosis/drug effects , Apoptosis/immunology , Cell Line , Disease Models, Animal , Female , Humans , Mice , Microglia/drug effects , Microglia/immunology , Photoreceptor Cells/immunology , Photoreceptor Cells/pathology , Retinal Degeneration/immunology , Retinal Degeneration/pathology , Scavenger Receptors, Class E/metabolism
6.
Med Hypotheses ; 136: 109513, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31812013

ABSTRACT

PCSK9 (Proprotein convertase Subtilisin/Kexin Type 9), an important regulator of lipid metabolism, has been shown to play a role in hepatocellular carcinoma by promoting metastasis. PCSK9 interferes with LDL metabolism and causes dyslipidemias in hematological malignancies particularly acute lymphoblastic leukemia. Nutraceuticals like berberine, curcumin and polydatin have been found effective in modulating PCSK9 expression by lowering LDL levels. Eugenol, a nutraceutical has shown a promising role in cancer due to its antioxidant and antihypercholesterolemic effects. In the present study, PCSK9 expression was measured in acute lymphoblastic leukemia (ALL) patients and was found to be significantly induced. Based on the results of expression analysis, a plausible hypothesis was made. Eugenol being an antioxidant will prevent oxidation of LDL. In the absence of ox-LDL, LOX1 scavenger receptor, which regulates PCSK9 expression, will not be activated. As the circulating LDL is reduced, it will no longer be able to support leukemia cell growth. The hypothesis was validated by an in silico and in vitro study. Molecular docking revealed hydrophobic interactions between ligand eugenol and macromolecules PCSK9 and LOX1. Expression of both PCSK9 and LOX1 were significantly reduced by eugenol in Jurkat cells. To conclude, PCSK9 could therapeutically be targeted by eugenol in leukemia cells.


Subject(s)
Eugenol/pharmacology , PCSK9 Inhibitors , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Antioxidants/pharmacology , Dietary Supplements , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Humans , Jurkat Cells , Ligands , Lipid Metabolism , Lipoproteins, LDL/metabolism , Molecular Docking Simulation , Neoplasm Metastasis , Proprotein Convertase 9/metabolism , Scavenger Receptors, Class E/antagonists & inhibitors , Scavenger Receptors, Class E/metabolism
7.
Lipids ; 54(11-12): 697-705, 2019 11.
Article in English | MEDLINE | ID: mdl-31574567

ABSTRACT

Oxidized low-density lipoprotein (Ox-LDL) may induce apoptosis and dysfunction of vascular endothelial cells, contributing to the initiation and development of atherosclerosis and lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) plays a central role in Ox-LDL uptake in the course of atherogenesis. Humanin (HN), a mitochondrial-derived peptide, was recently demonstrated to exert a protective role against endothelial dysfunction and Ox-LDL-induced progression of atherosclerosis. The HN analog HNGF6A (HNG) modulates cholesterol metabolism in macrophage RAW 264.7 cells. However, whether HNG affects Ox-LDL metabolism in endothelial cells is unknown. In this study, we investigated the effect of HNG on Ox-LDL accumulation in human umbilical vein endothelial cell (HUVEC) and its underlying mechanisms. HUVEC were preincubated with HNG for 1 h before addition of Ox-LDL. Total cholesterol content was measured by using a tissue total cholesterol assay kit and flow cytometry. Cell viability was measured by CCK8 assay. Protein content was examined by Western blot assays. Flow cytometry was used to identify apoptotic cells. Flow cytometry and tissue total cholesterol assays showed that HNG reduced Ox-LDL accumulation in HUVEC. In addition, HNG inhibited Ox-LDL-induced apoptosis of HUVEC. Western blot results showed that HNG reduced LOX-1 protein content. However, when LOX-1 was knocked down or inhibited, the effect of HNG in reducing Ox-LDL aggregation and apoptosis in HUVEC disappeared. Our study demonstrated that HNG reduces lipid aggregation and apoptosis in HUVEC in a LOX-1-dependent manner.


Subject(s)
Apoptosis/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Lipid Metabolism/drug effects , Peptide Fragments/pharmacology , Scavenger Receptors, Class E/antagonists & inhibitors , Cell Survival/drug effects , Cells, Cultured , Humans , Protein Aggregates/drug effects , Scavenger Receptors, Class E/metabolism
8.
Drug Des Devel Ther ; 13: 2579-2589, 2019.
Article in English | MEDLINE | ID: mdl-31440038

ABSTRACT

Purpose: Functional impairment of endothelial progenitor cells (EPCs) is frequently observed in patients with diabetic vascular complications. Astragaloside IV (ASV) has a significant protective effect against vascular endothelial dysfunction. Thus, this study aimed to investigate the role of ASV on oxidized low-density lipoprotein (ox-LDL)-induced EPCs dysfunction and its potential mechanisms. Methods: EPCs were isolated from the peripheral blood of mice and treated with different concentration of ASV (10, 20, 40, 60, 80, 100 and 200 µM). ox-LDL was served as a stimulus for cell model. The proliferation and migration, and improved tube formation ability of EPCs were determined. Reactive oxygen species (ROS) production and the levels of inflammatory cytokines, including interleukin 1ß (IL-1ß), IL-6, IL-10 and tumor necrosis factor (TNF-α) were measured. The expression oflectin-like oxidized LDL receptor (LOX-1) andNod-like receptor nucleotide-binding domain leucine rich repeat containing protein 3 (NLRP3) inflammasome were detected by Western blot analysis. Results: We found ASV treatment alleviated ox-LDL-induced cellular dysfunction, as evidenced by promoted proliferation and migration, and improved tube formation ability. Besides, ASV treatment significantly suppressed ox-LDL-induced ROS production and the levels of inflammatory cytokines. ASV inhibited ox-LDL-induced expression of LOX-1 in a concentration-dependent manner. Overexpression of LOX-1 in EPCs triggered NLRP3inflammasome activation, while inhibition of LOX-1 or treatment with ASV suppressed ox-LDL-induced NLRP3 inflammasome activation. Furthermore, overexpression of LOX-1 in ox-LDL-induced EPCs furtherly impaired cellular function, which could be ameliorated by ASV treatment. Conclusion: Our study showed that ASV may protect EPCs against ox-LDL-induced dysfunction via LOX-1/NLRP3 pathway.


Subject(s)
Endothelial Progenitor Cells/drug effects , Inflammasomes/drug effects , Lipoproteins, LDL/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Saponins/pharmacology , Scavenger Receptors, Class E/antagonists & inhibitors , Triterpenes/pharmacology , Animals , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Endothelial Progenitor Cells/metabolism , Inflammasomes/metabolism , Lipoproteins, LDL/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress/drug effects , Rats , Scavenger Receptors, Class E/metabolism , Structure-Activity Relationship
9.
Peptides ; 121: 170122, 2019 11.
Article in English | MEDLINE | ID: mdl-31386895

ABSTRACT

Growth hormone secretagogues (GHS) have been proved to exert protective effects on the cardiovascular system, while their potential beneficial effects on macrophages in atherosclerosis (AS) are rarely been clarified. This study aimed to demonstrate whether hexarelin, a synthetic peptidyl GHS, can suppress AS progression via regulating the function of macrophages. AS was induced by chronic (3 months) feeding with high lipid diet in ApoE-/- mice. Mice were treated either with hexarelin (100 µg/kg s.c., q.d. for 3 months) (AS + Hex group) or saline (AS group). Age-matched C57BL/6 J mice were used as normal controls. AS and related signaling molecules in aortic tissues and RAW264.7 macrophages were identified with variant methods including histological staining, ELISA, western blotting, confocal microscopy and flow cytometry. AS significantly developed in ApoE-/- mice fed with high lipids diet. Hexarelin decreased serum TC, TG and LDL-c, increased serum HDL-c and attenuated the formation of atherosclerotic plaques and neointima compared with the AS group. Hexarelin decreased the aortic expressions of CD68 and LOX-1 which were elevated in the AS group. Hexarelin increased GHSR expression, suppressed ox-LDL uptake and LOX-1 expression and inhibited nuclear factor-kappa B (NF-κB) activation both in the aorta of ApoE-/- mice and in RAW264.7 macrophages. We conclude that hexarelin effectively attenuates AS progression in ApoE-/- mice by modulating circulatory lipids profile and inhibiting macrophage ox-LDL uptake via suppressing the LOX-1-NF-κB signaling pathway. The study supports the perspective of hexarelin as an anti-AS drug.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/drug therapy , NF-kappa B/genetics , Oligopeptides/pharmacology , Plaque, Atherosclerotic/drug therapy , Scavenger Receptors, Class E/genetics , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/deficiency , Atherosclerosis/etiology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Biological Transport/drug effects , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Diet, High-Fat/adverse effects , Disease Models, Animal , Gene Expression Regulation , Lipoproteins, LDL/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Plaque, Atherosclerotic/etiology , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/metabolism , RAW 264.7 Cells , Receptors, Ghrelin/genetics , Receptors, Ghrelin/metabolism , Scavenger Receptors, Class E/antagonists & inhibitors , Scavenger Receptors, Class E/metabolism , Signal Transduction , Triglycerides/blood
10.
Curr Atheroscler Rep ; 21(10): 38, 2019 07 27.
Article in English | MEDLINE | ID: mdl-31350594

ABSTRACT

PURPOSE OF REVIEW: In any case, in proatherogenic conditions, LOX-1 is uniquely upregulated in vascular cells and mediates the entire atherogenic process from LDL oxidation to plague arrangement. As evidence supporting the crucial role of LOX-1 in atherogenesis keeps accumulating, there is developing an enthusiasm for LOX-1 as a potential remedial target. RECENT FINDINGS: Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is the major receptor for binding and uptake of oxidized low-density lipoprotein (oxLDL) in endothelial cells. Following internalization of oxLDL, LOX-1 starts a vicious cycle from activation of proinflammatory signaling pathways, subsequently advancing an expanded responsive oxygen species arrangement and secretion of proinflammatory cytokines. In healthy arteries, expression of lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is practically undetectable. This review portrays existing evidence supporting the role of LOX-1 in mediating of proatherosclerotic impacts of oxLDL which result in endothelial dysfunction, proinflammatory recruitment of monocytes into the arterial intima, arrangement of foam cells, endothelial cell dysfunction and vascular smooth muscle cell proliferation, and platelet enactment, angiogenesis just as in plaque development. Likewise, abridges LOX-1 modulatory compounds and in vivo and in vitro examinations toward the improvement of small molecules and biologics that could be of therapeutic use.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/pathology , Scavenger Receptors, Class E/metabolism , Up-Regulation , Endothelial Cells/metabolism , Foam Cells/metabolism , Humans , Inflammation Mediators/metabolism , Lipoproteins, LDL/metabolism , Muscle, Smooth, Vascular/physiopathology , Plaque, Atherosclerotic/metabolism , Platelet Activation , Reactive Oxygen Species/metabolism , Scavenger Receptors, Class E/antagonists & inhibitors , Signal Transduction
11.
Biosci Biotechnol Biochem ; 83(6): 1094-1101, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30871430

ABSTRACT

Oxidized low-density lipoprotein (ox-LDL) leads to atherosclerosis via lectin-like oxidized lipoprotein receptor-1 (LOX-1), one of the major receptor for ox-LDL. Inhibition of the binding of ox-LDL to LOX-1 decreases the proinflammatory and atherosclerotic events. The aim of the present study was to investigate whether protamine, a polybasic nuclear protein, interferes the binding of ox-LDL to LOX-1. Using sandwich ELISA with newly generated antibody, we measured the blocking effect of protamine on the binding of ox-LDL to LOX-1. Protamine dose-dependently inhibited the binding of ox-LDL to LOX-1. DiI-labeled ox-LDL uptake assay in two types of cultured human endothelial cells was performed with fluorescence microplate reader. Activation of extracellular-signal-regulated kinase (ERK)1/2 by ox-LDL was analyzed by immunoblotting. We found that protamine suppressed uptake of ox-LDL in endothelial cells and inhibited ERK1/2 activation by ox-LDL. These results suggest that protamine may possess anti-atherogenic potential by inhibiting ox-LDL binding to LOX-1 through electrostatic interactions.


Subject(s)
Atherosclerosis/prevention & control , Lipoproteins, LDL/metabolism , Protamines/pharmacology , Scavenger Receptors, Class E/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Fluorescence , Humans , Lipoproteins, LDL/antagonists & inhibitors , Protamines/administration & dosage , Protein Binding , Scavenger Receptors, Class E/antagonists & inhibitors
12.
Biomacromolecules ; 20(3): 1157-1166, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30642176

ABSTRACT

Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is a cell surface scavenger receptor. The protein is involved in binding and internalization of oxidized low-density lipoprotein (oxLDL), which leads under pathophysiological circumstances to plaque formation in arteries and initiation of atherosclerosis. A structural feature of LOX-1 relevant to oxLDL binding is the "basic spine" motif consisting of linearly aligned arginine residues stretched over the dimer surface. Inhibition of LOX-1 can be done by blocking these positively charged motifs. Here we report on the design, synthesis, and evaluation of a series of novel LOX-1 inhibitors having different numbers of sulfates and polyethylene glycerol (PEG) spacer. Two molecules, compounds 6b and 6d, showed binding affinity in the low nM range, i.e. 45.8 and 47.4 nM, respectively. The in vitro biological studies reveal that these molecules were also able to block the interaction of LOX-1 with its cognate ligands oxLDL, aged RBC, and bacteria.


Subject(s)
Drug Design , Glycerol/chemistry , Polyethylene Glycols/chemistry , Scavenger Receptors, Class E/antagonists & inhibitors , Sulfates/chemistry , Binding Sites , HEK293 Cells , Humans , Ligands
13.
Molecules ; 23(5)2018 05 07.
Article in English | MEDLINE | ID: mdl-29735887

ABSTRACT

Atherosclerosis, a chronic inflammatory disease, is a major contributor to cardiovascular diseases. Ursolic acid (UA) is a phytonutrient with widely biological effects including anti-oxidative, anti-inflammatory, and so on. At present, the effect of UA on atherosclerosis and the mechanism of action are still obscure. This study focused on investigating the effects of UA on atherosclerosis both in vivo and in vitro. We first selected LOX-1 as our target, which was reckoned as a new promising receptor for treating atherosclerosis. The evaluation in vitro suggested that UA significantly decreased endothelial LOX-1 expression induced by LPS both in mRNA and protein levels. Pre-treatment of UA also inhibited TLR4/MyD88 signaling activated by LPS. Moreover, UA reduced ROS production and suppressed the activation of NF-κB stimulated by LPS. Particularly, the evaluation in vivo further verified the conclusion obtained in vitro. In ApoE−/− mice fed with an atherogenic diet, both UA (100 mg/kg/day) and simvastatin significantly attenuated atherosclerotic plaque formation and shrunk necrotic core areas. The enhanced expression of LOX-1 in atherosclerotic aorta was also dramatically decreased by administration of UA. Taken together, these results suggested that UA, with anti-atherosclerotic activity through inhibition of LOX-1 mediated by ROS/NF-κB signaling pathways, may become a valuable vascular protective candidate for the treatment of atherosclerosis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Aorta/drug effects , Apolipoproteins E/genetics , Atherosclerosis/drug therapy , NF-kappa B/genetics , Scavenger Receptors, Class E/genetics , Triterpenes/pharmacology , Animals , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/deficiency , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Disease Models, Animal , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells , Humans , Inflammation , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Scavenger Receptors, Class E/antagonists & inhibitors , Scavenger Receptors, Class E/metabolism , Signal Transduction , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Ursolic Acid
14.
Bioorg Med Chem Lett ; 27(22): 5010-5013, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29054360

ABSTRACT

Proanthocyanidins are oligomers of catechins that exhibit potent antioxidative activity and inhibit binding of oxidized low-density lipoprotein (OxLDL) to the lectin-like oxidized LDL receptor (LOX-1), which is involved in the onset and development of arteriosclerosis. Previous attempts aimed at developing proanthocyanidin derivatives with more potent antioxidative activity and stronger inhibition for LOX-1 demonstrated the synthesis of a novel proanthocyanidin derivative (1), in which the geometry of one catechin molecule in procyanidin B3 was constrained to a planar orientation. The radical scavenging activity of 1 was 1.9-fold higher than that of procyanidin B3. Herein, we synthesized another procyanidin B3 analogue (2), in which the geometries of both catechin molecules in the dimer were constrained to planar orientations. The radical scavenging activity of 2 was 1.5-fold higher than that of 1, suggesting that 2 may be a more effective candidate than 1 as a therapeutic agent to reduce oxidative stress induced in arteriosclerosis or related cerebrovascular disease.


Subject(s)
Biflavonoids/chemistry , Catechin/chemistry , Free Radical Scavengers/chemistry , Proanthocyanidins/chemistry , Biflavonoids/chemical synthesis , Biflavonoids/metabolism , Catechin/chemical synthesis , Catechin/metabolism , Dimerization , Free Radical Scavengers/chemical synthesis , Free Radical Scavengers/metabolism , Humans , Kinetics , Lipoproteins, LDL/antagonists & inhibitors , Lipoproteins, LDL/metabolism , Proanthocyanidins/chemical synthesis , Proanthocyanidins/metabolism , Scavenger Receptors, Class E/antagonists & inhibitors , Scavenger Receptors, Class E/metabolism
15.
Cancer Lett ; 408: 60-70, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28844714

ABSTRACT

Metastatic spread of cancer cells is the most life-threatening aspect of breast cancer and involves multiple steps including cell migration. We recently found that the TBC1D3 oncogene promotes the migration of breast cancer cells, and its interaction with CaM enhances the effects of TBC1D3. However, little is known regarding the mechanism by which TBC1D3 induces the migration of cancer cells. Here, we demonstrated that TBC1D3 stimulated the expression of oxidized low density lipoprotein receptor 1 (OLR1), a stimulator of cell migration, in breast cancer cells at the transcriptional level. Depletion of OLR1 by siRNAs or down-regulation of OLR1 expression using pomalidomide, a TNFα inhibitor, significantly decreased TBC1D3-induced migration of these cells. Notably, TBC1D3 overexpression activated NF-κB, a major effector of TNFα signaling, while inhibition of TNFα signaling suppressed the effects of TBC1D3. Consistent with this, NF-κB inhibition using its specific inhibitor caffeic acid phenethyl ester decreased both TBC1D3-induced OLR1 expression and cell migration, suggesting a critical role for TNFα/NF-κB signaling in TBC1D3-induced migration of breast cancer cells. Mechanistically, TBC1D3 induced activation of this signaling pathway on multiple levels, including by increasing the release of TNFα, elevating the transcription of TNFR1, TRAF1, TRAF5 and TRAF6, and decreasing the degradation of TNFR1. In summary, these studies identify the TBC1D3 oncogene as a novel regulator of TNFα/NF-κB signaling that mediates this oncogene-induced migration of human breast cancer cells by up-regulating OLR1.


Subject(s)
Breast Neoplasms/drug therapy , Cell Movement , GTPase-Activating Proteins/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins/metabolism , Scavenger Receptors, Class E/metabolism , Tumor Necrosis Factor-alpha/metabolism , Apoptosis , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Proliferation , Female , GTPase-Activating Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , NF-kappa B/genetics , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , Scavenger Receptors, Class E/antagonists & inhibitors , Scavenger Receptors, Class E/genetics , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/genetics
16.
Lipids Health Dis ; 16(1): 77, 2017 Apr 13.
Article in English | MEDLINE | ID: mdl-28407763

ABSTRACT

BACKGROUND: Atherosclerosis is a common cardiovascular disease that causes myocardial infarction, heart failure, and stroke. Increased oxidized low density lipoprotein (ox-LDL) in the sub-endothelium is the characteristic origin of atherogenesis. Klotho, an anti-aging protein, has been reported to protect against atherosclerosis and ameliorate endothelial dysfunction in vivo. The aim of this study is to investigatethe anti-oxidative activity of Klothoin ox-LDL-treated human umbilical vein endothelial cells (HUVECs). METHODS: After pre-treatment with 200 pMKlotho for 1 h, HUVECs were stimulated with 50 µg/ml ox-LDL for 24 h. Reactive oxygen species (ROS) and superoxide dismutase (SOD) levels were analyzed in the cells. Nitric oxide (NO) concertation was measured in the medium supernatant. Related proteins or genes were detected with Western blot or real time PCR, respectively, in the cell lysates. RESULTS: Initially, oxidative damage in HUVECs was established by adding 50 µg/mL ox-LDL, which resulted in decreased cellular viability, SOD/Cu/Zn-SOD and endothelial NO synthase (eNOS) expression and NO production, as well as increased malondialdehyde (MDA) levels, ROS production, inducible NO synthase (iNOS), phosphatidyl inositol-3 kinase (PI3K), protein kinase B (Akt), gp91 phox, and lectin-like ox-LDL receptor (LOX-1) expression in HUVECs. Pre-incubation with recombinant Klotho (200 pM) significantly prevented all of these alterations. These results suggest that Klotho can attenuate ox-LDL-induced oxidative stress in HUVECs through upregulating oxidative scavengers (SOD and NO) viaactivating the PI3K/Akt/eNOS pathway and depressing LOX-1expression. CONCLUSIONS: These results suggest that Klotho has a potential therapeutic effect on attenuating endothelial dysfunction and ameliorating atherosclerosis.


Subject(s)
Endothelium, Vascular/metabolism , Glucuronidase/metabolism , Lipoproteins, LDL/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress , Scavenger Receptors, Class E/antagonists & inhibitors , Second Messenger Systems , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Survival , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/pathology , Glucuronidase/genetics , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Klotho Proteins , Lipid Peroxidation , Lipoproteins, LDL/metabolism , Malondialdehyde/agonists , Malondialdehyde/antagonists & inhibitors , Malondialdehyde/metabolism , Nitric Oxide/agonists , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/chemistry , Nitric Oxide Synthase Type III/genetics , Phosphatidylinositol 3-Kinase/chemistry , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/agonists , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/agonists , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Recombinant Proteins/metabolism , Scavenger Receptors, Class E/agonists , Scavenger Receptors, Class E/metabolism , Superoxide Dismutase/antagonists & inhibitors , Superoxide Dismutase/chemistry , Superoxide Dismutase/metabolism
17.
Sci Rep ; 5: 16740, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26578342

ABSTRACT

Atherosclerosis related cardiovascular diseases continue to be the primary cause of mortality in developed countries. The elevated level of low density lipoprotein (LDL) is generally considered to be the driver of atherosclerosis, but recent years have seen a shift in this perception in that the vascular plaque buildup is mainly caused by oxidized LDL (ox-LDL) rather than native-LDL. The scavenger receptor LOX-1 found in endothelial cells binds and internalizes ox-LDL which leads to the initiation of plaque formation in arteries. Using virtual screening techniques, we identified a few potential small molecule inhibitors of LOX-1 and tested their inhibitory potential using differential scanning fluorimetry and various cellular assays. Two of these molecules significantly reduced the uptake of ox-LDL by human endothelial cells, LOX-1 transcription and the activation of ERK1/2 and p38 MAPKs in human endothelial cells. In addition, these molecules suppressed ox-LDL-induced VCAM-1 expression and monocyte adhesion onto human endothelial cells demonstrating their therapeutic potential.


Subject(s)
Drug Design , Ligands , Lipoproteins, LDL/chemistry , Scavenger Receptors, Class E/chemistry , Cell Adhesion/genetics , Computer Simulation , Drug Discovery , Gene Expression , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mitogen-Activated Protein Kinases/metabolism , Models, Molecular , Molecular Conformation , Protein Binding , Protein Interaction Domains and Motifs , Protein Stability , Scavenger Receptors, Class E/antagonists & inhibitors , Scavenger Receptors, Class E/genetics , Small Molecule Libraries , Structure-Activity Relationship , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
18.
J Cardiovasc Transl Res ; 8(8): 458-65, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26385009

ABSTRACT

Lectin-like oxidized low-density lipoprotein receptor-1 (SR-E1, LOX-1, OLR1) was first discovered as a vascular receptor for modified lipoprotein particles nearly 20 years ago. Since then, in vitro and in vivo studies have demonstrated an association between LOX-1, a soluble form (sLOX-1) and a number of diseases including atherosclerosis, arthritis, hypertension and pre-eclampsia. However, converting such discoveries into tools and drugs for routine clinical use is dependent on translational preclinical and clinical studies but such studies have only begun to emerge in the past decade. In this review, we identify the key clinical applications and corresponding criteria that need to be addressed for the effective use of LOX-1-related probes and molecules for patient benefit in different disease states.


Subject(s)
Antibodies/therapeutic use , Cardiovascular Agents/therapeutic use , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/drug therapy , Immunologic Techniques , Scavenger Receptors, Class E/antagonists & inhibitors , Translational Research, Biomedical/methods , Animals , Antibodies/adverse effects , Antibodies/immunology , Antibody Specificity , Cardiovascular Agents/immunology , Cardiovascular Diseases/immunology , Cardiovascular Diseases/metabolism , Drug Design , Epitopes , Humans , Molecular Targeted Therapy , Predictive Value of Tests , Scavenger Receptors, Class E/immunology , Scavenger Receptors, Class E/metabolism , Signal Transduction
19.
Cell Cycle ; 14(10): 1583-95, 2015.
Article in English | MEDLINE | ID: mdl-25950192

ABSTRACT

Statins are largely used in clinics in the treatment of patients with cardiovascular diseases for their effect on lowering circulating cholesterol. Lectin-like oxidized low-density lipoprotein (LOX-1), the primary receptor for ox-LDL, plays a central role in the pathogenesis of atherosclerosis and cardiovascular disorders. We have recently shown that chronic exposure of cells to lovastatin disrupts LOX-1 receptor cluster distribution in plasma membranes, leading to a marked loss of LOX-1 function. Here we investigated the molecular mechanism of statin-mediated LOX-1 inhibition and we demonstrate that all tested statins are able to displace the binding of fluorescent ox-LDL to LOX-1 by a direct interaction with LOX-1 receptors in a cell-based binding assay. Molecular docking simulations confirm the interaction and indicate that statins completely fill the hydrophobic tunnel that crosses the C-type lectin-like (CTLD) recognition domain of LOX-1. Classical molecular dynamics simulation technique applied to the LOX-1 CTLD, considered in the entire receptor structure with or without a statin ligand inside the tunnel, indicates that the presence of a ligand largely increases the dimer stability. Electrophoretic separation and western blot confirm that different statins binding stabilize the dimer assembly of LOX-1 receptors in vivo. The simulative and experimental results allow us to propose a CTLD clamp motion, which enables the receptor-substrate coupling. These findings reveal a novel and significant functional effect of statins.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Scavenger Receptors, Class E/metabolism , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Dimerization , Fluorescent Dyes/chemistry , HEK293 Cells , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lipoproteins, LDL/chemistry , Lipoproteins, LDL/metabolism , Lovastatin/chemistry , Lovastatin/metabolism , Lovastatin/pharmacology , Microscopy, Fluorescence , Molecular Docking Simulation , Protein Binding , Protein Stability/drug effects , Protein Structure, Tertiary , Scavenger Receptors, Class E/antagonists & inhibitors , Scavenger Receptors, Class E/genetics
20.
Acta Biochim Biophys Sin (Shanghai) ; 47(3): 174-82, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25630653

ABSTRACT

Coronary artery disease (CAD) is a major health problem worldwide. The most severe form of CAD is acute coronary syndrome (ACS). Recent studies have demonstrated the beneficial role of atorvastatin in ACS; however, the mechanisms underlying this effect have not been fully clarified. Growing evidence indicates that activation of the lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) plays an important role in oxidative stress-induced cardiomyocyte apoptosis during ACS. In this study, we examined whether atorvastatin inhibits H2O2-induced LOX-1 expression and H9c2 cardiomyocyte apoptosis, and investigated the underlying signaling pathway. Treatment of H9c2 cardiomyocytes with H2O2 resulted in elevated expression of LOX-1 mRNA and protein, as well as increased caspase-3 and -9 protein expression and cell apoptosis. H2O2-induced LOX-1 expression, caspase protein expression, and cardiomyocyte apoptosis were attenuated by pretreatment with atorvastatin. Atorvastatin activated H2O2-inhibited phosphorylation of Akt in a concentration-dependent manner. The Akt inhibitor, LY294002, inhibited the effect of atorvastatin on inducing Akt phosphorylation and on suppressing H2O2-mediated caspase up-regulation and cell apoptosis. These findings indicate that atorvastatin protects cardiomyocyte from oxidative stress via inhibition of LOX-1 expression and apoptosis, and that activation of H2O2-inhibited phosphorylation of Akt may play an important role in the protective function of atorvastatin.


Subject(s)
Atorvastatin/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line , Cell Survival/drug effects , Chromones/pharmacology , Hydrogen Peroxide/toxicity , Morpholines/pharmacology , Myocytes, Cardiac/cytology , Oxidative Stress/drug effects , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Scavenger Receptors, Class E/antagonists & inhibitors , Scavenger Receptors, Class E/genetics , Scavenger Receptors, Class E/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL