Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 194
Filtrar
1.
Pharm Biol ; 60(1): 56-64, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34905418

RESUMEN

CONTEXT: Atherosclerosis (AS) is a chronic inflammatory disease. Human vascular smooth muscle cell (hVSMC) accumulation and human umbilical vein endothelial cell (HUVEC) dysfunction are associated with the pathogenesis of AS. This study explores whether myristicin plays a protective role in AS. MATERIALS AND METHODS: hVSMCs and HUVECs were stimulated with 100 µg/mL oxidized low-density lipoprotein (ox-LDL) to establish a cellular model of AS. Cell viability, lactate dehydrogenase (LDH) release and cell apoptosis were evaluated using MTT, LDH and flow cytometry assays, respectively. Cell migration and inflammatory cytokine release were assessed using Transwell assay and ELISA. RESULTS: Myristicin (5, 10, 25, and 50 µM) had no obvious effect on cell viability or the activity of LDH in hVSMCs, while 100 and 200 µM myristicin markedly suppressed hVSMCs viability and increased LDH release. Myristicin had no obvious effect on cell viability or the activity of LDH in HUVECs. Myristicin inhibited viability and increased apoptosis in ox-LDL-treated hVSMCs, but was associated with increased proliferation and inhibited apoptosis in HUVECs stimulated by ox-LDL. Additionally, myristicin markedly suppressed ox-LDL-induced hVSMCs migration and the release of inflammatory cytokines, including MCP-1, IL-6, VCAM-1 and ICAM-1, in HUVECs. Results also demonstrated that the promoting effects of ox-LDL on the PI3K/Akt and NF-κB signalling pathway in both hVSMCs and HUVECs were abolished by treatment with myristicin. DISCUSSION AND CONCLUSIONS: Myristicin regulated proliferation and apoptosis by regulating the PI3K/Akt/NF-κB signalling pathway in ox-LDL-stimulated hVSMCs and HUVECs. Thus, myristicin may be used as a new potential drug for AS treatment.


Asunto(s)
Derivados de Alilbenceno/farmacología , Apoptosis/efectos de los fármacos , Aterosclerosis/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Dioxolanos/farmacología , Derivados de Alilbenceno/administración & dosificación , Aterosclerosis/patología , Células Cultivadas , Dioxolanos/administración & dosificación , Relación Dosis-Respuesta a Droga , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Lipoproteínas LDL/administración & dosificación , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
2.
Pharm Biol ; 59(1): 1106-1116, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34403325

RESUMEN

CONTEXT: The effect of kaempferol, a regulator of oestrogen receptors, on atherosclerosis (AS) and the underlying mechanism is elusive. OBJECTIVE: To explore the effect and mechanism of kaempferol on AS. METHODS AND MATERIALS: In vivo, C57BL/6 and apolipoprotein E (APOE)-/- mice were randomly categorized into six groups (C57BL/6: control, ovariectomy (OVX), high-fat diet (HFD); APOE-/-: OVX-HFD, OVX-HFD + kaempferol (50 mg/kg) and OVX-HFD + kaempferol (100 mg/kg) and administered with kaempferol for 16 weeks, intragastrically. Oil-Red and haematoxylin-eosin (HE) staining were employed to examine the effect of kaempferol. In vitro, human aortic endothelial cells (HAECs) were pre-treated with or without kaempferol (5, 10 or 20 µM), followed by administration with kaempferol and oxidized low-density lipoprotein (ox-LDL) (200 µg/mL). The effect of kaempferol was evaluated using flow cytometry, and TdT-mediated dUTP Nick-End Labelling (TUNEL). RESULTS: In vivo, kaempferol (50 and 100 mg/kg) normalized the morphology of blood vessels and lipid levels and suppressed inflammation and apoptosis. It also activated the G protein-coupled oestrogen receptor (GPER) and PI3K/AKT/nuclear factor-erythroid 2-related factor 2 (Nrf2) pathways. In vitro, ox-LDL (200 µg/mL) reduced the cell viability to 50% (IC50). Kaempferol (5, 10 or 20 µM) induced-GPER activation increased cell viability to nearly 10%, 19.8%, 30%, and the decreased cellular reactive oxygen species (ROS) generation (16.7%, 25.6%, 31.1%), respectively, consequently attenuating postmenopausal AS. However, the protective effects of kaempferol were blocked through co-treatment with si-GPER. CONCLUSIONS: The beneficial effects of kaempferol against postmenopausal AS are associated with the PI3K/AKT/Nrf2 pathways, mediated by the activation of GPER.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Quempferoles/farmacología , Receptores de Estrógenos/genética , Receptores Acoplados a Proteínas G/genética , Animales , Aorta/citología , Apoptosis/efectos de los fármacos , Aterosclerosis/patología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Humanos , Quempferoles/administración & dosificación , Lipoproteínas LDL/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Factor 2 Relacionado con NF-E2/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba/efectos de los fármacos
3.
J Am Heart Assoc ; 10(13): e014572, 2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34121413

RESUMEN

Background MEDI6012 is recombinant human lecithin cholesterol acyltransferase, the rate-limiting enzyme in reverse cholesterol transport. Infusions of lecithin cholesterol acyltransferase have the potential to enhance reverse cholesterol transport and benefit patients with coronary heart disease. The purpose of this study was to test the safety, pharmacokinetic, and pharmacodynamic profile of MEDI6012. Methods and Results This phase 2a double-blind study randomized 48 subjects with stable coronary heart disease on a statin to a single dose of MEDI6012 or placebo (6:2) (NCT02601560) with ascending doses administered intravenously (24, 80, 240, and 800 mg) and subcutaneously (80 and 600 mg). MEDI6012 demonstrated rates of treatment-emergent adverse events that were similar to those of placebo. Dose-dependent increases in high-density lipoprotein cholesterol were observed with area under the concentration-time curves from 0 to 96 hours of 728, 1640, 3035, and 5318 should be: mg·h/mL in the intravenous dose groups and 422 and 2845 mg·h/mL in the subcutaneous dose groups. Peak mean high-density lipoprotein cholesterol percent change was 31.4%, 71.4%, 125%, and 177.8% in the intravenous dose groups and 18.3% and 111.2% in the subcutaneous dose groups, and was accompanied by increases in endogenous apoA1 (apolipoprotein A1) and non-ATP-binding cassette transporter A1 cholesterol efflux capacity. Decreases in apoB (apolipoprotein B) were observed across all dose levels and decreases in atherogenic small low-density lipoprotein particles by 41%, 88%, and 79% at the 80-, 240-, and 800-mg IV doses, respectively. Conclusions MEDI6012 demonstrated an acceptable safety profile and increased high-density lipoprotein cholesterol, endogenous apoA1, and non-ATP-binding cassette transporter A1 cholesterol efflux capacity while reducing the number of atherogenic low-density lipoprotein particles. These findings are supportive of enhanced reverse cholesterol transport and a functional high-density lipoprotein phenotype. Registration URL: https://www.clinicaltrials.gov; Unique identifier: NCT02601560.


Asunto(s)
Enfermedad Coronaria/tratamiento farmacológico , Lipoproteínas HDL/administración & dosificación , Lipoproteínas LDL/administración & dosificación , Fosfatidilcolina-Esterol O-Aciltransferasa/administración & dosificación , Proteínas Recombinantes/administración & dosificación , Administración Intravenosa , Adulto , Anciano , Apolipoproteína A-I/sangre , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Humanos , Inyecciones Subcutáneas , Lipoproteínas HDL/efectos adversos , Lipoproteínas HDL/sangre , Lipoproteínas LDL/efectos adversos , Lipoproteínas LDL/sangre , Masculino , Persona de Mediana Edad , Fosfatidilcolina-Esterol O-Aciltransferasa/efectos adversos , Fosfatidilcolina-Esterol O-Aciltransferasa/sangre , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/sangre , Resultado del Tratamiento
4.
J Ethnopharmacol ; 266: 113453, 2021 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-33039628

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Banxia Baizhu Tianma decoction (BBTD) is a classical representative prescription for expelling phlegm, extinguishing wind, strengthening the spleen and dissipating excessive fluid in traditional Chinese medicine (TCM). According to both TCM theory and about 300 years of clinical practice, BBTD is especially suitable for hypertensive patients of abdominal obesity and lacking physical activity. AIM OF THE STUDY: The present study tried to interpret the pharmacology of the ancient formula of BBTD. Herein, we focused on the plasma metabonomics of BBTD and evaluated the effect and targets of BBTD on endothelial protective effect. METHODS: Obesity-related hypertensive mice were induced by high-fat diet for 20 weeks. BBTD (17.8 g/kg) was administered intragastrically for 8 weeks, and telmisartan group (12.5 mg/kg) was used as positive drug. Body weight, blood pressure, triglyceride and cholesterol were recorded to evaluate the efficacy of BBTD in vivo. Lipid deposition in aortic roots was assessed by oil red O staining, while morphology of aortas was observed by HE staining. Ultra performance liquid chromatography/tandem mass spectrometry (UPLC-MS/MS) was performed to study the plasma non-targeted metabonomics. According to the data of metabonomics, human aortic endothelial cells (HAECs) were treated by oxidized low-density lipoprotein (ox-LDL, 50 µg/mL) with/without BBTD (2, 1 or 0.5 mg/mL). Apoptosis rate (Annexin V-FITC/PI), migration (Transwell), cytoskeleton (Phalloidin) and density of VE-cadherin (Immunofluorescence staining) were used to investigate the effect of BBTD in vitro. Transcriptome sequencing was performed (2 mg/mL BBTD vs ox-LDL) to screen the possible targets of BBTD in endothelial protection against ox-LDL. RESULTS: BBTD effectively reduced the body weight and total cholesterol, and decreased 12.1 mmHg in SBP and 10.5 mmHg in DBP of obesity-related hypertensive mice (P < 0.05). BBTD attenuated lipid deposition in arterial roots and improved the morphology of aortas in vivo. Plasma metabolite profiles identified 94 differential metabolites and suggested BBTD mainly affected glycerophospholipids and fatty acyls. Bioinformatics analysis indicated sphingolipid metabolism and fluid shear stress and atherosclerosis were main pathways. Therefore, we focused on endothelial protective effect of BBTD against ox-LDL. In vitro, BBTD demonstrated endothelial protective effects, decreasing apoptosis rate, improving cell migration in dose-dependent manner and maintaining cell morphology. Transcriptome sequencing identified 251 downregulated and 603 upregulated mRNAs after 24h-BBTD treatment, which reversed 51.8% change in mRNAs (393 DE mRNAs) induced by ox-LDL. Bioinformatics analysis supported the potential of BBTD in hypertension and suggested that BBTD improved endothelial cells by targeting mainly on p53 and PPAR signaling pathways. CONCLUSIONS: BBTD attenuates obesity-related hypertension by regulating metabolism of glycerophospholipids and endothelial protection.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Hipertensión/tratamiento farmacológico , Metabolómica , Obesidad/prevención & control , Animales , Aorta/citología , Aorta/efectos de los fármacos , Apoptosis/efectos de los fármacos , Células Cultivadas , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Medicamentos Herbarios Chinos/administración & dosificación , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Hipertensión/complicaciones , Hipertensión/metabolismo , Lipoproteínas LDL/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/metabolismo
5.
Physiol Int ; 107(2): 294-305, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32750030

RESUMEN

Although the use of aspirin has substantially reduced the risks of cardiovascular events and death, its potential mechanisms have not been fully elucidated. In a previous study, we found that aspirin triggers cellular autophagy. In the present study, we aimed to determine the protective effects of aspirin on human coronary artery endothelial cells (HCAECs) and explore its underlying mechanisms. HCAECs were treated with oxidized low-density lipoprotein (ox-LDL), angiotensin II (Ang-II), or high glucose (HG) with or without aspirin stimulation. The expression levels of endothelial nitric oxide (NO) synthase (eNOS), p-eNOS, LC3, p62, phosphor-nuclear factor kappa B (p-NF-κB), p-p38 mitogen-activated protein kinase (p-p38 MAPK), and Beclin-1 were detected via immunoblotting analysis. Concentrations of soluble intercellular adhesion molecule-1 (sICAM-1) and soluble vascular cell adhesion molecule-1 (sVCAM-1) were measured via ELISA. NO levels were determined using the Griess reagent. Autophagic flux was tracked by tandem mRFP-GFP-tagged LC3. Results showed that aspirin increased eNOS level and reduced injury to the endothelial cells (ECs) caused by ox-LDL, Ang-II, and HG treatment in a dose-dependent manner. Aspirin also increased the LC3II/LC3I ratio, decreased p62 expression, and enhanced autophagic flux (autophagosome and autolysosome puncta) in the HCAECs. p-NF-κB and p-p38 mitogen-activated protein kinase inhibition, sVCAM-1 and sICAM-1 secretion, and eNOS activity promotion by aspirin treatment were found to be dependent on Beclin-1. These results suggested that aspirin can protect ECs from ox-LDL-, Ang-II-, and HG-induced injury by activating autophagy in a Beclin-1-dependent manner.


Asunto(s)
Aspirina/farmacología , Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Vasos Coronarios/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Angiotensina II/administración & dosificación , Antiinflamatorios no Esteroideos/farmacología , Autofagia/efectos de los fármacos , Células Cultivadas , Enfermedad de la Arteria Coronaria/inducido químicamente , Enfermedad de la Arteria Coronaria/metabolismo , Enfermedad de la Arteria Coronaria/patología , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Lipoproteínas LDL/administración & dosificación , Transducción de Señal , Vasoconstrictores/administración & dosificación
6.
Lipids ; 55(6): 615-626, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32558932

RESUMEN

Cellular lipid metabolism, lipoprotein interactions, and liver X receptor (LXR) activation have been implicated in the pathophysiology and treatment of cancer, although findings vary across cancer models and by lipoprotein profiles. In this study, we investigated the effects of human-derived low-density lipoproteins (LDL), high-density lipoproteins (HDL), and HDL-associated proteins apolipoprotein A1 (apoA1) and serum amyloid A (SAA) on markers of viability, cholesterol flux, and differentiation in K562 cells-a bone marrow-derived, stem-like erythroleukemia cell model of chronic myelogenous leukemia (CML). We further evaluated whether lipoprotein-mediated effects were altered by concomitant LXR activation. We observed that LDL promoted higher K562 cell viability in a dose- and time-dependent manner and increased cellular cholesterol concentrations, while LXR activation by the agonist TO901317 ablated these effects. LXR activation in the presence of HDL, apoA1 and SAA-rich HDL suppressed K562 cell viability, while robustly inducing mRNA expression of ATP-binding cassette transporter A1 (ABCA1). HDL and its associated proteins additionally suppressed mRNA expression of anti-apoptotic B-cell lymphoma-extra large (BCL-xL), and the erythroid lineage marker 5'-aminolevulinate synthase 2 (ALAS2), while SAA-rich HDL induced mRNA expression of the megakaryocytic lineage marker integrin subunit alpha 2b (ITGA2B). Together, these findings suggest that lipoproteins and LXR may impact the viability and characteristics of CML cells.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Lipoproteínas HDL/farmacología , Lipoproteínas LDL/farmacología , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/metabolismo , Apolipoproteína A-I/metabolismo , Apolipoproteína A-I/farmacología , Diferenciación Celular/genética , Supervivencia Celular/efectos de los fármacos , Colesterol/metabolismo , Relación Dosis-Respuesta a Droga , Regulación Leucémica de la Expresión Génica , Humanos , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/sangre , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Lipoproteínas HDL/administración & dosificación , Lipoproteínas HDL/metabolismo , Lipoproteínas LDL/administración & dosificación , Lipoproteínas LDL/metabolismo , Receptores X del Hígado/metabolismo , Proteína Amiloide A Sérica/metabolismo , Proteína Amiloide A Sérica/farmacología , Factores de Tiempo , Proteína bcl-X/genética
7.
Toxicol Appl Pharmacol ; 400: 115037, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32417438

RESUMEN

BACKGROUND: In recent years, small animal arterial port-catheter systems have been implemented in rodents with reasonable success. The aim of the current study is to employ the small animal port-catheter system to evaluate the safety of multiple hepatic-artery infusions (HAI) of low-density lipoprotein-docosahexaenoic acid (LDL-DHA) nanoparticles to the rat liver. METHODS: Wistar rats underwent surgical placement of indwelling HAI ports. Repeated administrations of PBS or LDL-DHA nanoparticles were performed through the port at baseline and days 3 and 6. Rats were sacrificed on day 9 at which point blood and various organs were collected for histopathology and biochemical analyses. RESULTS: The port-catheter systems were implanted successfully and repeated infusions of PBS or LDL-DHA nanoparticles were tolerated well by all animals over the duration of the study. Measurements of serum liver/renal function tests, glucose and lipid levels did not differ between control and LDL-DHA treated rats. The liver histology was unremarkable in the LDL-DHA treated rats and the expression of hepatic inflammatory regulators (NF-κß, IL-6 and CRP) were similar to control rats. Repeated infusions of LDL-DHA nanoparticles did not alter liver glutathione content or the lipid profile in the treated rats. The DHA extracted by the liver was preferentially metabolized to the anti-inflammatory DHA-derived mediator, protectin DX. CONCLUSION: Our findings indicate that repeated HAI of LDL-DHA nanoparticles is not only well tolerated and safe in the rat, but may also be protective to the liver.


Asunto(s)
Catéteres de Permanencia/efectos adversos , Ácidos Docosahexaenoicos/administración & dosificación , Arteria Hepática , Infusiones Intraarteriales/efectos adversos , Lipoproteínas LDL/administración & dosificación , Hígado/metabolismo , Nanopartículas/administración & dosificación , Animales , Glucemia/análisis , Ácidos Docosahexaenoicos/farmacocinética , Infusiones Intraarteriales/métodos , Pruebas de Función Renal , Lípidos/sangre , Lipoproteínas LDL/farmacocinética , Hígado/irrigación sanguínea , Pruebas de Función Hepática , Masculino , Ratas Wistar , Distribución Tisular
8.
Life Sci ; 256: 117852, 2020 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-32470448

RESUMEN

AIMS: Atherosclerosis (AS) performs the important pathogenesis which refers to coronaryheart and vascular diseases. Long non-coding RNAs (lncRNAs) was reported to be related to the AS progression. We aimed to probe the role and potential mechanism of Myocardial Infarction Associated Transcript (MIAT) in AS. MATERIALS AND METHODS: Levels of MIAT, microRNA-148b (miR-148b) and pregnancy-associated plasma protein A (PAPPA) were detected by quantitative Real-time polymerase chain reaction (qRT-PCR) in oxidized low-density lipoprotein (ox-LDL)-induced human aorta vascular smooth muscle cells (HA-VSMCs). Proliferation and migration were examined by Cell counting kit-8 (CCK-8) and wound-healing assays, respectively. Protein levels of Ki-67, proliferating cell nuclear antigen (PCNA), matrix metalloproteinase (MMP)-2, MMP-9 and PAPPA were examined by western blot assay. Ki-67 and PCNA level was detected by flow cytometry. The interaction among MIAT, miR-148b and PAPPA was confirmed via dual-luciferase reporter and RNA immunoprecipitation (RIP). The biology role of MIAT was detected by an AS model in vivo. KEY FINDINGS: The levels of MIAT and PAPPA were augmented, whereas mature miR-148b level was repressed in ox-LDL-induced AS model. The inhibitory effects of knockdown of MIAT on proliferation and migration were relieved by miR-148b inhibitor. Additionally, miR-148b regulated proliferation and migration by targeting PAPPA. Mechanically, MIAT functioned as sponge of miR-148b to impact PAPPA expression. MIAT knockdown protected AS mice against lipid metabolic disorders in vivo. SIGNIFICANCE: Proliferation and migration were modified by MIAT/miR-148b/PAPPA axis in ox-LDL induced AS cell model, supplying a novel insight into the underlying application of MIAT in the clinical treatment of AS.


Asunto(s)
Aterosclerosis/patología , MicroARNs/genética , Proteína Plasmática A Asociada al Embarazo/metabolismo , ARN Largo no Codificante/genética , Animales , Aterosclerosis/genética , Movimiento Celular/genética , Proliferación Celular/genética , Progresión de la Enfermedad , Técnicas de Silenciamiento del Gen , Humanos , Lipoproteínas LDL/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
9.
Eur Rev Med Pharmacol Sci ; 24(6): 3282-3292, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32271446

RESUMEN

OBJECTIVE: Atherosclerosis (AS) is a representative inflammatory vascular disease. This study explored the molecular pathogenesis of AS based on circular RNA (circRNA), the checkpoint with forkhead-associated and ring-finger domains (circ_CHFR). PATIENTS AND METHODS: The cell model of AS in vitro was established by stimulating human vascular smooth muscle cells (VSMCs) with oxidized low-density lipoprotein (ox-LDL). The RNA expression was measured by quantitative Real-time polymerase chain reaction (qRT-PCR). Cell viability and colony formation ability were separately evaluated using 3-(4, 5-dimethylthiazol-2-y1)-2, 5-diphenyl tetrazolium bromide (MTT) and colony formation assay. Cell migration was assessed via the transwell assay. The inflammation injury was analyzed by enzyme-linked immunosorbent assay (ELISA). Associated proteins were determined through Western blot. The combination of hypothetic targets was ascertained using Dual-Luciferase reporter assay. RESULTS: Circ_CHFR was up-regulated in AS serums and ox-LDL-stimulated VSMCs. Circ_CHFR depletion weakened the ox-LDL-induced promotion of cell growth, migration and inflammation in VSMCs. Circ_CHFR positively regulated Wnt3 expression and the downregulation of Wnt3 abrogated the ox-LDL-triggered injuries in VSMCs. Circ_CHFR functioned as the sponge of microRNA-214-3p (miR-214-3p) and miR-214-3p targeted Wnt3. Circ_CHFR regulated cell growth, migration and inflammation via regulating the expression of Wnt3 as a competitive endogenous RNA (ceRNA) of miR-214 in ox-LDL-treated VSMCs. Circ_CHFR/miR-214-3p axis mediated the Wnt3/ß-catenin signal pathway. CONCLUSIONS: Circ_CHFR contributed to the progression of AS through the miR-214-3p/Wnt3/ß-catenin signals, which illuminated the molecular mechanism of AS and suggested circ_CHFR might be an index for AS treatment.


Asunto(s)
Aterosclerosis/patología , Proteínas de Ciclo Celular/genética , Inflamación/patología , Proteínas de Neoplasias/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Ubiquitina-Proteína Ligasas/genética , Aterosclerosis/genética , Movimiento Celular/genética , Proliferación Celular/genética , Células Cultivadas , Progresión de la Enfermedad , Humanos , Inflamación/genética , Lipoproteínas LDL/administración & dosificación , MicroARNs/genética , Músculo Liso Vascular/citología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/patología , ARN Circular/genética , Proteína Wnt3/genética , beta Catenina/metabolismo
10.
Life Sci ; 243: 117270, 2020 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-31923421

RESUMEN

AIMS: The purpose of this study is to investigate the effect of PP2A on the progression of AS and the special molecular mechanism. MAIN METHODS: The expression of PP2A in Human umbilical vein endothelial cells (HUVECs) induced by different concentrations of Ox-LDL was measured by RT-PCR and Western blot. The binding activity of PP2A and LOX-1 was determined by CoIP assay. Western blot was used to measure the protein expression of VCAM-1, ICAM-1 and MCP-1. KEY FINDING: The results revealed that the expression of PP2A was decreased with the increase of Ox-LDL concentration in HUVECs. Overexpression of PP2A alleviated Ox-LDL-induced dysfunction and inflammatory response in HUVECs. The results of Co-immunoprecipitation (CoIP) showed that PP2A had direct effect on LOX-1, and PP2A inhibited the expression of LOX-1. In addition, overexpression of LOX-1 reversed the inhibitory effect of PP2A on Ox-LDL-induced dysfunction and inflammatory response in HUVECs. What is more, PP2A inhibited LOX-1/ROS/MAPK axis. SIGNIFICANCE: it suggests that PP2A alleviates Ox-LDL-induced dysfunction and inflammatory response of HUVECs potentially by regulating the LOX-1/ROS/MAPK axis,which suggests that PP2A has anti-inflammatory effect during the formation of as, and the molecular therapy of PP2A provides a theoretical basis.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Lipoproteínas LDL/farmacología , Lipooxigenasa/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína Fosfatasa 2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Relación Dosis-Respuesta a Droga , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación/prevención & control , Lipoproteínas LDL/administración & dosificación
11.
DNA Cell Biol ; 38(12): 1499-1511, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31804889

RESUMEN

Although long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs) have been suggested to play important roles in the pathogenesis of diseases, atherosclerosis-related lncRNAs and circRNAs remain rarely reported. This study aimed to explore the underlying molecular mechanisms of atherosclerosis based on the competing endogenous RNA (ceRNA) regulatory hypothesis of lncRNAs and circRNAs. The expression profiles of circRNAs, lncRNAs, and mRNAs in human THP-1 macrophages treated with oxidized low-density lipoprotein (an in vitro atherosclerosis model), or not, were obtained from the Gene Expression Omnibus database under accession numbers GSE107522, GSE54666, and GSE54039, respectively. The present study identified 29 differentially expressed circRNAs in GSE107522, 544 differentially expressed genes (DEGs) in GSE54666, and 502 DEGs and 231 differentially expressed lncRNAs in GSE54039 datasets by using the Linear Models for Microarray Data method. Eight DEGs were found to be shared and expressed with the consistent trend in GSE54666 and GSE54039 datasets. Two of them (ASPH, aspartate beta-hydroxylase; and PDE3B, phosphodiesterase 3B) were suggested to be crucial based on functional enrichment, protein-protein interaction, and ceRNA network analyses. ASPH, through interaction with CACNA2D4 (calcium voltage-gated channel auxiliary subunit alpha2delta 4), may be associated with atherosclerosis by regulating the cellular response to calcium ion; and PDE3B may exert roles in negative regulation of angiogenesis through cross talk with ELMO1 (engulfment and cell motility 1). Furthermore, the expression of ASPH and PDE3B may be regulated by hsa_circ_0028198/hsa_circ_0092317/XIST-miR-543; PDE3B expression may be also modulated by hsa_circ_0092317/hsa_circ_0003546/H19/XIST-miR-326. In conclusion, our identified ceRNA interaction axes may possibly be important targets for treatment of atherosclerosis.


Asunto(s)
Células Espumosas/metabolismo , Lipoproteínas LDL/administración & dosificación , Macrófagos/metabolismo , MicroARNs/genética , ARN Circular/genética , ARN Largo no Codificante/genética , ARN Mensajero/genética , Células Cultivadas , Células Espumosas/citología , Células Espumosas/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes , Humanos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Oxidación-Reducción
12.
Arq. bras. med. vet. zootec. (Online) ; 71(6): 1889-1895, Nov.-Dec. 2019. tab, graf
Artículo en Portugués | LILACS, VETINDEX | ID: biblio-1055129

RESUMEN

A utilização da gema de ovo dificulta a padronização de meios diluidores e apresenta riscos biológicos. Assim, este estudo avaliou diferentes concentrações da lipoproteína de baixa densidade (LDL), em substituição à gema de ovo, para a confecção de diluentes para criopreservação espermática em ovinos. Foram utilizados um diluidor controle (CTR= 20% de gema de ovo) e cinco tratamentos, substituindo-se a gema pelas diferentes proporções de LDL (T1=6%; T2=8%; T3=12%; T4=16%; T5=20%), todos à base de TRIS-glicerol. Para o estudo, utilizaram-se dois ejaculados, de seis reprodutores da raça Santa Inês. Sessenta dias após a criopreservação, as amostras foram descongeladas e avaliadas subjetivamente quanto à motilidade total (MT, %) e progressiva (MP, %), ao vigor (1-5) e à integridade funcional (choque hisposmótico com água destilada, %) e estrutural (corante supravital eosina, %) das membranas espermáticas. As avaliações de vigor e funcionalidade de membrana não diferiram (P>0,05) entre os grupos. Entretanto, os grupos T4 (P<0,01) e T5 (P<0,05) foram superiores ao CTR para os parâmetros MT, MP e integridade estrutural de membrana, o que confirma que as LDLs podem ser alternativas eficientes para substituição da gema de ovo em diluidores para criopreservação de sêmen ovino.(AU)


The use of egg yolk makes it difficult to standardize extenders and presents biological hazards. Thus, this study evaluated different concentrations of low-density lipoprotein (LDL) to replace yolk extenders for production of sperm for cryopreservation in ovine. A control extender was used (CTR= 20% yolk) and five treatments, replacing the yolk by different ratios of LDL (T1= 6%; T2= 8%, T3= 12%; T4= 16%; T5= 20%) all based on TRIS-glycerol. For the study, two ejaculates from six Santa Ines breeding were used. Sixty days after cryopreservation, the samples were thawed and evaluated for total motility (MT, %) and progressive motility (MP, %), vigor (1-5) and the functional integrity (hyposmotic shock with distilled water, %) and structural (supravital dye eosin, %) of the sperm membranes. The evaluations of strength and membrane functionality didn't differ (P> 0.05) between groups. However, T4 (P< 0.01) and T5 (P< 0.05) groups were superior to the CTR for the MT, MP, and membrane structural integrity parameters, which confirms that LDLs can be efficient alternatives for yolk replacement in extenders for cryopreservation of ovine semen.(AU)


Asunto(s)
Animales , Masculino , Preservación de Semen/métodos , Ovinos , Criopreservación/veterinaria , Lipoproteínas LDL/administración & dosificación
13.
Biomed Pharmacother ; 115: 108840, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31048189

RESUMEN

BACKGROUND AND AIMS: Hirudo is an important Chinese medicine that has been widely used in patients with thrombosis-related diseases. We aimed to evaluate the protective effect and potential mechanism of Hirudo extract (HE) on the process of atherosclerosis (AS) as well as identify its active components in the lipopolysaccharide (LPS) - or oxidized low-density lipoprotein (ox-LDL)-induced cell models. METHODS: After treatment, adhesion molecules and pro-inflammatory cytokines induced by LPS were examined by qPCR and ELISA. ROS production, cell apoptosis, and lipid accumulation in ox-LDL-induced cells were analyzed by flow cytometry, qPCR, western blotting, and immunofluorescence staining. In addition, the main active components of HE were identified and analyzed for preventing the progression of AS. RESULTS: In this study, we found that HE pretreatment for 48 h significantly inhibited monocyte adhesion and reduced the levels of adhesion factors (ICAM-1 and VCAM-1) and pro-inflammatory factors (IL-6 and TNF-α) in LPS-induced endothelial cells. Moreover, HE attenuated ox-LDL-induced ROS accumulation and apoptosis in macrophage cells via mitochondrial apoptotic pathways. Additionally, HE pretreatment effectively inhibited cholesterol uptake and increased cholesterol efflux by regulating the LOX-1/LXR-α/ABCA1 pathway. Importantly, the polypeptides from HE (PP) with a molecular weight < 10,000 Da accounted for about 62.9% of the total amount of polypeptides, which in turn may be active components of HE that are responsible for inhibiting inflammation, foam cell formation and apoptosis. CONCLUSION: PP from HE potently inhibits endothelial cell inflammatory injury and macrophage foam cell formation and apoptosis by regulating the LOX-1/LXR-α/ABCA1 pathway, thereby providing additional support to the beneficial effects of HE in preventing AS.


Asunto(s)
Células Espumosas/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Sanguijuelas/química , Macrófagos/efectos de los fármacos , Péptidos/farmacología , Receptores Depuradores de Clase E/metabolismo , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Adhesión Celular/efectos de los fármacos , Colesterol/metabolismo , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/toxicidad , Lipoproteínas LDL/administración & dosificación , Lipoproteínas LDL/toxicidad , Receptores X del Hígado/genética , Receptores X del Hígado/metabolismo , Ratones , Péptidos/química , Células RAW 264.7 , Especies Reactivas de Oxígeno , Receptores Depuradores de Clase E/genética , Células THP-1
14.
Monaldi Arch Chest Dis ; 89(1)2019 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-30985095

RESUMEN

It is important, in our opinion, to provide physicians with a brief update of scientifically-sound evidence in preventive nutrition, to be employed in their everyday practice, since the latest scientific and clinical advances in this area are generally not well known. Here, we review the most recent evidence in support of an optimal cardio-protective diet, and we identify the need to focus mainly on protective food which should be part of such diet, rather than on nutrients with negative effects to be limited (salt, saturated fats, simple sugars). We conclude that, to favor patient compliance, it is also necessary to underscore indications on the topics for which there is convincing and coherent literature, leaving other less-explored aspects to individual preferences.


Asunto(s)
Enfermedades Cardiovasculares/dietoterapia , Dieta/métodos , Cooperación del Paciente/psicología , Consumo de Bebidas Alcohólicas/efectos adversos , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/prevención & control , Dieta/normas , Dieta Hiposódica/efectos adversos , Dieta Hiposódica/estadística & datos numéricos , Carbohidratos de la Dieta/administración & dosificación , Carbohidratos de la Dieta/efectos adversos , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/efectos adversos , Fibras de la Dieta/administración & dosificación , Fibras de la Dieta/efectos adversos , Fibras de la Dieta/estadística & datos numéricos , Suplementos Dietéticos/efectos adversos , Suplementos Dietéticos/estadística & datos numéricos , Medicina Basada en la Evidencia/educación , Medicina Basada en la Evidencia/métodos , Femenino , Humanos , Lipoproteínas LDL/administración & dosificación , Lipoproteínas LDL/efectos adversos , Masculino , Médicos , Guías de Práctica Clínica como Asunto , Ensayos Clínicos Controlados Aleatorios como Asunto/métodos , Conducta de Reducción del Riesgo
16.
Trends Cardiovasc Med ; 29(1): 22-26, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29934015

RESUMEN

We provide an up-to-date overview of current topics surrounding oxidized low-density lipoprotein (oxLDL) and its related antibodies in the quest to better identify the individuals at risk of cardiovascular disease and atherosclerotic plaques with unfavorable characteristics. We discuss the potential of oxLDL and anti-oxLDL antibodies as serum biomarkers of cardiovascular disease and emerging studies examining the targeting of arterial oxLDL for imaging and therapeutic delivery.


Asunto(s)
Anticuerpos/metabolismo , Arterias/metabolismo , Aterosclerosis/metabolismo , Lipoproteínas LDL/metabolismo , Animales , Anticuerpos/administración & dosificación , Anticuerpos/inmunología , Arterias/diagnóstico por imagen , Arterias/inmunología , Arterias/patología , Aterosclerosis/diagnóstico por imagen , Aterosclerosis/inmunología , Aterosclerosis/terapia , Biomarcadores/metabolismo , Humanos , Inmunización/métodos , Lipoproteínas LDL/administración & dosificación , Lipoproteínas LDL/inmunología , Imagen Molecular/métodos , Placa Aterosclerótica , Valor Predictivo de las Pruebas , Pronóstico , Vacunas/administración & dosificación , Vacunas/inmunología
17.
Life Sci ; 216: 67-74, 2019 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-30218721

RESUMEN

AIMS: The accumulation of lipids in macrophages contributes to the development of atherosclerosis. Cholesterol efflux of lipid-loaded macrophages mediated by ATP binding cassette (ABC) cholesterol transporters, on the other hand, has been shown to attenuate atherosclerosis progression in patients with unknown mechanism. We therefore sought to test the effect of metformin that reduced cardiovascular risk in diabetic patients independent of its hypoglycemia effect on cholesterol transport in murine raw264.7 macrophages. MATERIALS AND METHODS: Mouse raw264.7 macrophages were loaded with Ox-LDL (50 µg/ml) for 24 h before incubated with metformin (15 µM) for 24 h. Foam cell formation was assessed by Oil red staining and BIODIPY fluorescent staining as well as cholesterol-ester quantification by commercial kit. Cholesterol uptake and expression of scavenger receptors were detected by flow-cytometry. Cholesterol efflux capacity was measured by fluorescent plate-reader and ABC transporters were detected by Western Blots. Cytokines were detected by ELISA in supernatants and normalized by cellular lysates. KEY FINDINGS: Our results showed that metformin decreased oxidized low-density lipoprotein (Ox-LDL)-induced cholesterol accumulation and foam cell formation by increasing cholesterol efflux to HDL, which was associated with an upregulation of ABC transporter ABCG-1. Moreover, metformin increased Ox-LDL-impaired IL-10 secretion, an important anti-foam cell cytokine in atherosclerosis. SIGNIFICANCE: Our data highlighted the therapeutic potential of targeting macrophage cholesterol efflux with new or existing drugs for the possible reduction of foam cell formation in the prevention and treatment of diabetes-accelerated atherosclerosis.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/metabolismo , Aterosclerosis/prevención & control , Hipoglucemiantes/farmacología , Lipoproteínas LDL/administración & dosificación , Metformina/farmacología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/genética , Animales , Colesterol/metabolismo , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Células Espumosas/efectos de los fármacos , Células Espumosas/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Células RAW 264.7 , Regulación hacia Arriba/efectos de los fármacos
18.
Med Sci Monit ; 24: 5509-5516, 2018 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-30088495

RESUMEN

BACKGROUND ox-LDL-induced injury of brain microvascular endothelial cells (BMECs) is strongly associated with cerebral vascular diseases such as cerebral arterial atherosclerosis. ROCK inhibitor was proved to be anti-apoptotic and has been used in treating cerebral vascular diseases. Research on the neuroprotective effects of a novel ROCK inhibitor, FSD-C10, is still limited. The present study investigated the anti-apoptotic effect and underlying molecular mechanism of FSD-C10 in ox-LDL-mediated apoptosis of BMECs. MATERIAL AND METHODS ox-LDL and/or FSD-C10 were used to incubate immortalized human BMECs. MTT assay was used to assess cell viability. Cell apoptosis was evaluated by TUNEL assay. A colorimetric method was used to assess ROCK activity. Western blot analysis was used to examine the expression and phosphorylation levels of proteins. RESULTS ox-LDL incubation reduced the viability of BMECs by inducing cell apoptosis in a concentration-dependent manner. ROCK activity was also elevated by ox-LDL incubation in BMECs in a concentration-dependent manner. Expression level of Bcl2 was reduced while expression levels of Bax and active caspase3 were increased by ox-LDL treatment in a concentration-dependent manner. ox-LDL also increased the phosphorylation levels of p38, JNK, and ERK1/2 in a concentration-dependent manner. FSD-C10 treatment increased the cell viability by reducing apoptosis of BMECs exposed to ox-LDL. Moreover, FSD-C10 was found to suppress the phosphorylation levels of p38, JNK, and ERK1/2 and the expression levels of Bax and active caspase3 in ox-LDL treated BMECs. CONCLUSIONS FSD-C10 increases cell viability in ox-LDL-treated BMECs by reducing cell apoptosis. ROCK/MAPKs-mediated apoptosis appears to be the underlying molecular mechanism.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Células Endoteliales/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Apoptosis/efectos de los fármacos , Encéfalo/irrigación sanguínea , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/patología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Lipoproteínas LDL/administración & dosificación , Lipoproteínas LDL/metabolismo , Microvasos/efectos de los fármacos , Microvasos/enzimología , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
19.
Circulation ; 138(22): 2513-2526, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-29997115

RESUMEN

BACKGROUND: Atherosclerotic cardiovascular disease is a chronic inflammatory process initiated when cholesterol-carrying low-density lipoprotein (LDL) is retained in the arterial wall. CD4+ T cells, some of which recognize peptide components of LDL as antigen, are recruited to the forming lesion, resulting in T-cell activation. Although these T cells are thought to be proatherogenic, LDL immunization reduces disease in experimental animals. These seemingly contradictory findings have hampered the development of immune-based cardiovascular therapy. The present study was designed to clarify how activation of LDL-reactive T cells impacts on metabolism and vascular pathobiology. METHODS: We have developed a T-cell receptor-transgenic mouse model to characterize the effects of immune reactions against LDL. Through adoptive cell transfers and cross-breeding to hypercholesterolemic mice expressing the antigenic human LDL protein apolipoprotein B-100, we evaluate the effects on atherosclerosis. RESULTS: A subpopulation of LDL-reactive T cells survived clonal selection in the thymus, developed into T follicular helper cells in lymphoid tissues on antigen recognition, and promoted B-cell activation. This led to production of anti-LDL immunoglobulin G antibodies that enhanced LDL clearance through immune complex formation. Furthermore, the cellular immune response to LDL was associated with increased cholesterol excretion in feces and with reduced vascular inflammation. CONCLUSIONS: These data show that anti-LDL immunoreactivity evokes 3 atheroprotective mechanisms: antibody-dependent LDL clearance, increased cholesterol excretion, and reduced vascular inflammation.


Asunto(s)
Aterosclerosis/prevención & control , Linfocitos T CD4-Positivos/inmunología , Colesterol/sangre , Lipoproteínas LDL/inmunología , Animales , Anticuerpos/inmunología , Apolipoproteína B-100/sangre , Apolipoproteínas E , Aterosclerosis/patología , Linfocitos B/citología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Modelos Animales de Enfermedad , Lipoproteínas LDL/administración & dosificación , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo
20.
Pharmazie ; 73(7): 408-412, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30001776

RESUMEN

This present investigation examined the mitigating impact of Ginkgolic acid in the organization on oxidized low-density lipoproteinox-LDL (ox- LDL) animated in HUVECs, and to clear up its fundamental molecular components. The levels of nitric oxide (NO), prostaglandin E2 (PGE2), and pro-inflammatory cytokines were measured by Griess examine and catalyst connected immunosorbent test. The declarations of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), mitogen-initiated protein kinases (MAPKs), and Akt were measured utilizing Western smearing. ox-LDL-instigated was utilized as the HUVECs cell model of inflammation. Ginkgolic acid significantly inhibited the production of NO, PGE2, and pro-inflammatory cytokines in a dose-dependent manner and suppressed the expression of iNOS and COX-2 in ox-LDL-stimulated HUVECs cells. Ginkgolic acid strongly suppressed NF-κB by preventing degradation of inhibitor of κB-α as well as by inhibiting phosphorylation of Akt and MAPKs. Ginkgolic acid reduced LDL-stimulated inflammation in endothelial cells. These outcomes suggest that the anti-inflammatory properties of Ginkgolic acid are related to a down-control of iNOS, COX-2, and master provocative cytokines through the restraint of NF-κB pathway in ox- LDL-animated endothelial cells.


Asunto(s)
Antiinflamatorios/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Inflamación/tratamiento farmacológico , Salicilatos/farmacología , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Dinoprostona/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Inflamación/patología , Lipoproteínas LDL/administración & dosificación , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA