Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Environ Sci Pollut Res Int ; 31(24): 35688-35704, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38740681

RESUMEN

In this work, iron-phosphorus based composite biochar (FPBC) was prepared by modification with potassium phosphate and iron oxides for the removal of heavy metal ions from single and mixed heavy metal (Pb and Cd) solutions. FTIR and XPS characterization experiments showed that the novel modified biochar had a greater number of surface functional groups compared to the pristine biochar. The maximum adsorption capacities of FPBC for Pb(II) and Cd(II) were 211.66 mg·g-1 and 94.08 mg·g-1 at 293 K. The adsorption of Pb(II) and Cd(II) by FPBC followed the proposed two-step adsorption kinetic model and the Freundlich isothermal adsorption model, suggesting that the mechanism of adsorption of Pb(II) and Cd(II) by FPBC involved chemical adsorption of multiple layers. Mechanistic studies showed that the introduction of -PO4 and -PO3 chemisorbed with Pb(II) and Cd(II), and the introduction of -Fe-O increased the ion exchange with Pb(II) and Cd(II) during the adsorption process and produced precipitates such as Pb3Fe(PO4)3 and Cd5Fe2(P2O7)4. Additionally, the abundant -OH and -COOH groups also participated in the removal of Pb(II) and Cd(II). In addition, FPBC demonstrated strong selective adsorption of Pb(II) in mixed heavy metal solutions. The Response Surface Methodology(RSM) analysis determined the optimal adsorption conditions for FPBC as pH 5.31, temperature 26.01 °C, and Pb(II) concentration 306.30 mg·L-1 for Pb(II). Similarly, the optimal adsorption conditions for Cd(II) were found to be pH 5.66, temperature 39.34 °C, and Cd(II) concentration 267.68 mg·L-1. Therefore, FPBC has the potential for application as a composite-modified adsorbent for the adsorption of multiple heavy metal ions.


Asunto(s)
Cadmio , Carbón Orgánico , Plomo , Fósforo , Contaminantes Químicos del Agua , Adsorción , Carbón Orgánico/química , Cadmio/química , Plomo/química , Contaminantes Químicos del Agua/química , Fósforo/química , Hierro/química , Cinética , Purificación del Agua/métodos , Metales Pesados/química
2.
Shock ; 60(2): 206-213, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37548713

RESUMEN

ABSTRACT: Background: The dysregulation of circular RNAs (circRNAs) is involved in various human diseases, including sepsis-induced acute lung injury (ALI). We aimed to investigate the role of circTDRD9 in the development of sepsis-induced ALI. Methods: Cell models of sepsis-induced ALI were established by treating A549 cells with LPS. The expression of circTDRD9, miR-223-3p, and RAB10 mRNA was measured by quantitative real-time PCR (qPCR). The levels of inflammatory factors were measured by ELISA. Oxidative stress-related indicators were monitored by using commercial detection kits. The expression of fibrosis-related proteins was detected by Western blot assay. Cell proliferation was assessed by EdU assay. The predicted binding relationship between miR-223-3p and circTDRD9 or RAB10 was verified by dual-luciferase reporter assay, RIP assay or pull-down assay. Results: CircTDRD9 was highly expressed in LPS-treated A549 cells. CircTDRD9 downregulation prevented LPS-induced inflammation, oxidative stress, cell proliferation inhibition, and cell fibrosis in A549 cells, whereas these effects were reversed by the inhibition of miR-223-3p, a target of circTDRD9. In addition, RAB10 was verified as a target of miR-223-3p, and RAB10 overexpression recovered LPS-induced inflammation, oxidative stress, cell proliferation inhibition, and cell fibrosis in A549 cells that were ameliorated by miR-223-3p restoration. Importantly, circTDRD9 positively regulated RAB10 expression by binding to miR-223-3p. Conclusion: CircTDRD9 overexpression was closely associated with LPS-induced ALI. CircTDRD9 contributed to LPS-induced ALI partly by upregulating RAB10 via binding to miR-223-3p.


Asunto(s)
Lesión Pulmonar Aguda , MicroARNs , Sepsis , Humanos , Apoptosis , Inflamación , Lipopolisacáridos/toxicidad , MicroARNs/genética , Sepsis/complicaciones , Sepsis/genética
4.
J Cell Mol Med ; 26(13): 3648-3658, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35678255

RESUMEN

Myocardial injury is a frequently occurring complication of sepsis. This study aims to investigate the molecular mechanism of long noncoding RNA (lncRNA) small nucleolar RNA host gene 1 (SNHG1)-mediated DNA methyltransferase 1/B-cell lymphoma-2 (DNMT1/Bcl-2) axis in sepsis-induced myocardial injury. Mice and HL-1 cells were treated with lipopolysaccharide (LPS) to establish animal and cellular models simulating sepsis and inflammation. LncRNA SNHG1 was screened out as a differentially expressed lncRNA in sepsis samples through microarray profiling, and the upregulated expression of lncRNA SNHG1 was confirmed in myocardial tissues of LPS-induced septic mice and HL-1 cells. Further experiments suggested that silencing of lncRNA SNHG1 reduced the inflammation and apoptotic rate of LPS-induced HL-1 cells. LncRNA SNHG1 inhibited Bcl-2 expression by recruiting DNMT1 to Bcl-2 promoter region to cause methylation. Inhibition of Bcl-2 promoter methylation reduced the inflammation and apoptotic rate of LPS-induced HL-1 cells. In vivo experiments substantiated that lncRNA SNHG1 silencing alleviated sepsis-induced myocardial injury in mice. Taken together, lncRNA SNHG1 promotes LPS-induced myocardial injury in septic mice by downregulating Bcl-2 through DNMT1-mediated Bcl-2 methylation.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasa 1 , MicroARNs , Proteínas Proto-Oncogénicas c-bcl-2 , ARN Largo no Codificante , Sepsis , Animales , Apoptosis/fisiología , Proliferación Celular/fisiología , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Lipopolisacáridos/farmacología , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Largo no Codificante/metabolismo , Sepsis/genética , Sepsis/metabolismo
5.
Cell Signal ; 78: 109840, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33221374

RESUMEN

Atherosclerosis (AS) is one of the significant chronic inflammatory pathology considering public health impact. Up-regulation of HDAC1 has been proved to be related with endothelial dysfunction which is correlated intimately with AS. Our research aims to investigate how histone deacetylase 1 (HDAC1)/miR-182-5p/vav guanine nucleotide exchange factor 3 (VAV3)/AKT axis participates in AS in terms of molecular mechanism. We detected miR-181-5p in human umbilical vein endothelial cells after treatment with aorta and ox-LDL in AS model mice. Dual luciferase reporter assay was employed to verify interaction of miR-182-5p and VAV3. ChIP was performed to determine the relationship between HDAC1 and promoter of miR-182-5p. Protein levels of HADC1, VAV3, AKT, p-AKT, vascular cell adhesion molecule-1 (VCAM-1), intercellular cell adhesion molecule-1 (ICAM-1), and monocyte chemotactic protein 1 (MCP-1) were detected by western blot analysis. CCK8 and flow cytometry were used to detect cell viability and apoptosis, respectively. After different treatments, the ability of cells to form monoclonal cells was detected, and AS was evaluated by detecting arterial injury and inflammation-related factors. Overexpression of HDAC1 could inhibit HUVECs proliferation and promote AS in mouse model. It was verified by dual luciferase assay that miR-182-5p could bind to VAV3 3'UTR mRNA. Meanwhile, HDAC1 repressed miR-182-5p expression through binding to miR-182-5p promoter and then inhibit VAV3 expression further. In summary, HDAC1 promoted AS through AKT pathway, which was improved by VAV3 activation mediated by miR-182-5p. Our results demonstrated that HDAC1 repressed miR-182-5p and activating AKT pathway via improving VAV3 to promote AS progression.


Asunto(s)
Aorta , Aterosclerosis/metabolismo , Histona Desacetilasa 1/metabolismo , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-vav/metabolismo , Transducción de Señal , Animales , Aorta/lesiones , Aorta/metabolismo , Aterosclerosis/genética , Modelos Animales de Enfermedad , Histona Desacetilasa 1/genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados para ApoE , MicroARNs/genética , Proteínas Proto-Oncogénicas c-vav/genética
6.
Cell Transplant ; 29: 963689720967672, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33172292

RESUMEN

Myocardial infarction (MI) is one of the most serious cardiovascular diseases associated with myocardial ischemia/reperfusion (I/R) injury. Glaucocalyxin A (GLA) is a biologically active ent-kauranoid diterpenoid that has been found to ameliorate myocardial I/R injury in mice. However, the mechanism has not been fully investigated. In the present study, we aimed to investigate the effect of GLA on rat cardiomyocytes H9c2 cells exposed to hypoxia/reoxygenation (H/R). The results showed that GLA treatment improved cell viability of H/R-stimulated H9c2 cells. Administration with GLA suppressed the H/R-stimulated reactive oxygen species (ROS) production in H9c2 cells. GLA also elevated the activities of antioxidant enzymes, including superoxide dismutase and glutathione peroxidase in H/R-stimulated H9c2 cells. Moreover, GLA prevented H/R-stimulated cell apoptosis in H9c2 cells, as evidenced by increased bcl-2 expression, decreased bax expression, as well as reduced caspase-3 activity. Furthermore, GLA enhanced the activation of protein kinase B (Akt)/nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling pathway in H9c2 cells exposed to H/R. Additionally, treatment with LY294002 reserved the protective effects of GLA on H/R-stimulated oxidative injury in H9c2 cells. In conclusion, these findings suggested that GLA protected H9c2 cells from H/R-stimulated oxidative damage, which was mediated by the Akt/Nrf2/HO-1 signaling pathway. Thus, GLA might be a promising therapeutic agent for the prevention and treatment of myocardial I/R.


Asunto(s)
Diterpenos de Tipo Kaurano/farmacología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Western Blotting , Hipoxia de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Hemo Oxigenasa (Desciclizante)/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
7.
Cell Transplant ; 29: 963689720949247, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32841049

RESUMEN

Tripartite motif 8 (TRIM8) is a member of the TRIM protein family that has been found to be implicated in cardiovascular disease. However, the role of TRIM8 in myocardial ischemia/reperfusion (I/R) has not been investigated. We aimed to explore the effect of TRIM8 on cardiomyocyte H9c2 cells exposed to hypoxia/reoxygenation (H/R). We found that TRIM8 expression was markedly upregulated in H9c2 cells after stimulation with H/R. Gain- and loss-of-function assays proved that TRIM8 knockdown improved cell viability of H/R-stimulated H9c2 cells. In addition, TRIM8 knockdown suppressed reactive oxygen species production and elevated the levels of superoxide dismutase and glutathione peroxidase. Knockdown of TRIM8 suppressed the caspase-3 activity, as well as caused significant increase in bcl-2 expression and decrease in bax expression. Furthermore, TRIM8 overexpression exhibited apposite effects with knockdown of TRIM8. Finally, knockdown of TRIM8 enhanced the activation of PI3K/Akt signaling pathway in H/R-stimulated H9c2 cells. Inhibition of PI3K/Akt by LY294002 reversed the effects of TRIM8 knockdown on cell viability, oxidative stress, and apoptosis of H9c2 cells. These present findings defined TRIM8 as a therapeutic target for attenuating and preventing myocardial I/R injury.


Asunto(s)
Citoprotección , Técnicas de Silenciamiento del Gen , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteínas del Tejido Nervioso/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Animales , Apoptosis/genética , Hipoxia de la Célula , Línea Celular , Supervivencia Celular/genética , Regulación hacia Abajo/genética , Proteínas del Tejido Nervioso/genética , Estrés Oxidativo , Oxígeno , Ratas , Regulación hacia Arriba/genética
8.
Mar Environ Res ; 157: 104934, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32275514

RESUMEN

Phytoplankton response to interannual climate variability has an important regulatory effect on the regional marine ecological environment and carbon cycle. In this study, we focused on the phytoplankton response in the upwelling region of the Sulu Ridge to the El Niño-Southern Oscillation (ENSO) based on monthly remote sensing chlorophyll-a concentration (Chl-a) and physical parameters from various sources from September 1997 to December 2017. We selected two El Niño events in 1997/1998 and 2015/2016 and two La Niña events in 1998/1999 and 2010/2011 to examine the response of Chl-a to ENSO events in this region. Results showed that El Niño and La Niña could enhance and inhibit the growth of phytoplankton in the Sulu Ridge in winter, respectively. For other seasons, the influence of ENSO on the Chl-a was inconsistent. Specifically, during El Niño events, the largest Chl-a increases occurred in winter, and the low sea surface temperature (SST) center appeared northwest of Sulu Ridge. The significant decrease of SST (~1.5 °C) during El Niño events in winter in the northeastern Sulu Ridge was mainly caused by the increase in Ekman transport (ET) and Ekman pumping velocity (EPV), which brought nutrient-rich subsurface water to the surface layer through the thin barrier layer and enhanced Chl-a. During La Niña events, the SST was higher (~0.8 °C) than the average and the high SST center generally appeared in the middle of the Sulu Ridge with the east-west direction in winter, which was resulted from the intensification of barrier layer thickness (BLT) and the decrease of ET, thus reducing the Chl-a. The different responses to El Niño and La Niña events indicate the high sensitivity of Chl-a in this region to the ENSO.


Asunto(s)
El Niño Oscilación del Sur , Fitoplancton/crecimiento & desarrollo , Clorofila A/análisis , Filipinas , Estaciones del Año
9.
Chem Biol Interact ; 316: 108922, 2020 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-31837296

RESUMEN

Homeodomain interacting protein kinase-2 (HIPK2) has emerged as a crucial stress-responsive kinase that plays a critical role in regulating cell survival and apoptosis. However, whether HIPK2 participates in regulating cardiomyocyte survival during myocardial ischemia/reperfusion injury remains unclear. Here, we investigated the regulatory effect of HIPK2 on hypoxia/reoxygenation (H/R)-induced cardiomyocyte injury and its potential underlying molecular mechanism. We found that HIPK2 expression was induced in response to H/R exposure. HIPK2 depletion by small interfering RNA (siRNA)-mediated gene silencing significantly decreased the viability and exacerbated H/R-induced apoptosis and reactive oxygen species (ROS) production in cardiomyocytes. Comparatively, HIPK2 overexpression effectively rescued H/R-impaired viability and repressed H/R-induced apoptosis and ROS production in cardiomyocytes. HIPK2 overexpression significantly increased the nuclear expression of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and enhanced Nrf2-mediated transcriptional activity. Moreover, HIPK2 overexpression significantly increased the transcription of Nrf2/ARE target genes. Additionally, Nrf2 inhibition partially reversed the HIPK2-mediated protective effect. Overall, these results demonstrate that HIPK2 overexpression protects cardiomyocytes from H/R-induced injury by enhancing Nrf2/ARE antioxidant signaling, data that suggest HIPK2 is a potential target for cardioprotection.


Asunto(s)
Apoptosis/efectos de los fármacos , Hipoxia de la Célula , Estrés Oxidativo/efectos de los fármacos , Oxígeno/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Elementos de Respuesta Antioxidante/genética , Células Cultivadas , Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo
10.
Oncotarget ; 8(38): 64143-64156, 2017 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-28969058

RESUMEN

Lung adenocarcinoma (LUAD) is the most common histological subtype of non-small cell lung cancer, but novel biomarkers for early diagnosis are lacking. Extensive effort has been exerted to identify miRNA biomarkers in LUAD. Unfortunately, high inter-lab variability and small sample sizes have produced inconsistent conclusions in this field. To resolve the above-mentioned limitations, we performed a comprehensive analysis based on LUAD miRNome profiling studies using the robust rank aggregation (RRA) method. Moreover, miRNA-gene interaction network, pathway enrichment analysis and Kaplan-Meier survival curves were used to investigate the clinical values and biological functions of the identified miRNAs. A total of six common differentially expressed miRNAs (DEMs) were identified in LUAD. An independent cohort further confirmed that four miRNAs (miR-21-5p, miR-210-3p, miR-182-5p and miR-183-5p) were up-regulated and two miRNAs (miR-126-3p and miR-218-5p) were down-regulated in LUAD tissues. Pathway enrichment analysis also suggested that the above-listed six DEMs may affect LUAD progression via the estrogen signaling pathway. Survival analysis based on the TCGA dataset revealed the potential prognostic values of six DEMs in patients with LUAD (P-value<0.01). In conclusion, we identified a panel of six miRNAs from LUAD using miRNome profiling studies. Our results provide evidence for the use of these six DEMs as novel diagnostic and prognostic biomarkers for LUAD patients.

11.
Int J Mol Med ; 36(1): 316-22, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26017061

RESUMEN

Pulmonary arterial hypertension (PAH) is a progressive pulmonary vascular disorder with high morbidity and mortality, and is characterized by excessive growth of endothelial cells. Recently, the mammalian target of rapamycin (mTOR) has attracted increasing attention due to its potential as a therapeutic target against certain diseases associated with proliferative and metabolic abnormalities. However, the effect on mTOR on PAH has not yet been elucidated. In the present study, a marked downregulation of mTOR was observed in PAH patients. Following construction of a mouse model of PAH by chronic exposure to hypoxia, adenovirus-mediated upregulation of mTOR significantly attenuated right ventricular systolic pressure, right ventricular hypertrophy and wall thickness of pulmonary arterioles, indicating a protective effect of mTOR on PAH. Further analysis confirmed that mTOR overexpression inhibited autophagy triggered by hypoxia through blocking light chain 3 II expression and increasing p62 levels. In vitro, hypoxia enhanced the proliferation of human pulmonary artery endothelial cells (PAECs), which was markedly abrogated by mTOR overexpression. Of note, upregulation of mTOR inhibited the hypoxia-induced autophagy pathway, which contributed to cell proliferation, while silencing of autophagy by RNA interference with ATG5 significantly inhibited cell proliferation. In conclusion, the results of the present study suggested a potential protective effect of mTOR on the progression of PAH by suppressing PAEC proliferation through blocking the autophagic pathway. Therefore, the present study suggested that mTOR is a promising therapeutic agent against PAH.


Asunto(s)
Autofagia/fisiología , Hipoxia de la Célula/fisiología , Hipertensión Pulmonar/patología , Proteínas Asociadas a Microtúbulos/genética , Serina-Treonina Quinasas TOR/biosíntesis , Adulto , Animales , Proteína 5 Relacionada con la Autofagia , Línea Celular , Proliferación Celular , Regulación hacia Abajo , Células Endoteliales , Femenino , Humanos , Hipertrofia Ventricular Derecha , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/biosíntesis , Persona de Mediana Edad , Arteria Pulmonar/citología , Interferencia de ARN , ARN Interferente Pequeño , Proteínas de Unión al ARN/biosíntesis , Función Ventricular Derecha
12.
Mol Med Rep ; 10(5): 2563-7, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25190613

RESUMEN

We previously demonstrated that resveratrol (Res) regulates the expression of PKC α and δ, to eventually inhibit growth and induce apoptosis in human gastric cancer cells. In the present study, the effect of Res on the growth of human pancreatic cancer cells was investigated, to further unveil the underlying mechanism. The human pancreatic cancer cell line MIA PaCa-2 was treated with three different concentrations of Res. Cell proliferation was assessed by the MTT assay, and apoptosis was detected by flow cytometry. Reverse transcription-quantitative PCR (RT-qPCR) and western blot analysis were used to measure the mRNA and protein levels of the Hedgehog (Hh) signaling proteins Ihh, Ptch and Smo. Our results revealed that Res can inhibit the cell proliferative ability in a time- and dose-dependent manner. The number of formed colonies in the Res- and 5-Fu (positive control)-treated groups was decreased as compared to the negative control group. Res further induced apoptosis of MIA PaCa-2 cells in a dose-dependent manner. In addition, the levels of Ihh, Ptch and Smo were decreased by Res treatment. Our findings suggest that Res inhibits proliferation and induces apoptosis of MIA PaCa-2 pancreatic cancer cells in vitro, which may be related to its inhibitory effect on the Hh signaling pathway.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Estilbenos/farmacología , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Fluorouracilo/farmacología , Proteínas Hedgehog/metabolismo , Humanos , Neoplasias Pancreáticas , Receptores Patched , Receptor Patched-1 , Receptores de Superficie Celular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Resveratrol , Receptor Smoothened
13.
DNA Cell Biol ; 33(4): 198-204, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24512183

RESUMEN

Recently, autophagy has drawn more attention in cardiovascular disease as it has important roles in lipid metabolism. Mammalian target of rapamycin (mTOR) is a key regulator of autophagy; however, its effect on atherosclerosis and the underlying mechanism remains undefined. In this study, an obvious upregulation of mTOR and p-mTOR protein was observed in macrophage-derived foam cells. Blocking mTOR expression with specific small interference RNA (siRNA) dramatically suppressed foam cell formation, accompanied by a decrease of lipid deposition. Further mechanistic analysis indicated that suppressing mTOR expression significantly upregulated autophagic marker LC3 expression and downregulated autophagy substrate p62 levels, indicating that mTOR silencing triggered autophagosome formation. Moreover, blocking mTOR expression obviously accelerated neutral lipid delivery to lysosome and cholesterol efflux from foam cells, implying that mTOR could induce macrophage foam cell formation by suppressing autophagic pathway. Further, mTOR silencing significantly upregulated ULK1 expression, which was accounted for mTOR-induced foam cell formation via autophagic pathway as treatment with ULK1 siRNA dampened LC3-II levels and increased p62 expression, concomitant with lipid accumulation and decreased cholesterol efflux from foam cells. Together, our data provide an insight into how mTOR accelerates the pathological process of atherosclerosis. Accordingly, blocking mTOR levels may be a promising therapeutic agent against atherosclerotic complications.


Asunto(s)
Autofagia/fisiología , Células Espumosas/citología , Regulación de la Expresión Génica/fisiología , Metabolismo de los Lípidos/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia , Compuestos Azo , Células Espumosas/metabolismo , Regulación de la Expresión Génica/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lipoproteínas LDL , Ratones , Microscopía Fluorescente , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Serina-Treonina Quinasas TOR/genética
14.
Int J Mol Med ; 32(5): 1215-21, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24043133

RESUMEN

Atherosclerotic plaque destabilization and rupture leads to acute coronary syndromes which cause serious damage to human health worldwide. However, there is currently a lack of efficient therapeutic methods. Mammalian target of rapamycin (mTOR) has been suggested to be involved in the development of atherosclerotic plaques and serves as a therapeutic target. The present study was performed to determine whether RNA interference (RNAi) of mTOR in vivo by LV­mediated small hairpin RNA (shRNA) was capable of inhibiting the progression of atherosclerotic plaques. LV­mediated shRNA against mTOR (LV­shmTOR) was designed and obtained. Male apolipoprotein E­deficient mice were fed a high­fat diet and a constrictive collar was placed around the right carotid arteries of these mice to induce plaque formation. Eight weeks after surgery, mice were randomly divided into the mTOR RNA interference (LV­shmTOR) group, receiving treatment with LV­mTOR­shRNA; the LV­shCON group, receiving treatment with LV­non­specific­shRNA; and the control group, receiving treatment with phosphate­buffered saline. Following transfection, the mice were sacrificed to evaluate the effects of mTOR expression silencing on atherosclerosis. Transfection of LV­mTOR­shRNA markedly inhibited the mRNA and protein expression levels. Knockdown of mTOR ameliorated dysregulated blood lipid metabolism and stabilized aortic atherosclerotic plaques by decreasing the plaque area and increasing the fibrous cap and cap­to­core ratio. Furthermore, macrophages were decreased by silencing mTOR in atherosclerotic plaques. In addition, western blot analysis revealed that the knockdown of mTOR increased autophagy­related protein 13 (Atg13) dephosphorylation and light chain 3­I/light chain 3­II (LC3­I/LC3­II) ratios, both of which were associated with a high activity of autophagy, suggesting an increase of autophagy in atherosclerotic plaques. Moreover, genes including matrix metalloproteinase 2, monocyte chemoattractant protein 1 and tissue factor, which promote plaque instability, were downregulated by silencing mTOR. These results demonstrate that LV­mediated mTOR silencing by RNAi treatment induces macrophage autophagy and is a potential strategy for the treatment of atherosclerotic plaques.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Aterosclerosis/terapia , Autofagia/fisiología , Lentivirus/genética , Interferencia de ARN/fisiología , Serina-Treonina Quinasas TOR/genética , Animales , Apolipoproteínas E/genética , Aterosclerosis/genética , Autofagia/genética , Macrófagos/metabolismo , Masculino , Ratones , Ratones Mutantes , Serina-Treonina Quinasas TOR/metabolismo
15.
Inflamm Res ; 62(6): 581-7, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23483217

RESUMEN

OBJECTIVE AND DESIGN: This study was aimed at investigating the effect of chlorogenic acid (CGA) on lipopolysaccharide (LPS)-induced proinflammatory signaling in hepatic stellate cells (HSCs). METHODS: An immortalized rat HSC line was cultured in vitro and treated with LPS in the absence or presence of CGA. Reactive oxygen species (ROS) production in the HSCs was monitored by flow cytometer using DCFH-DA. The protein expression levels of toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), nuclear factor-κB (NF-κB), and p-IκB-α were determined by Western blot. The mRNA expression levels of TLR4, MyD88, monocyte chemotactic protein 1(MCP-1), and interleukin 6 (IL-6) were detected by RT-PCR. The levels of MCP-1 and IL-6 in the culture supernatant of HSCs were measured by ELISA. RESULTS: CGA had no effect on expression of TLR4 and MyD88. However, the treatment of CGA can inhibit LPS-induced production of ROS in HSCs. Meanwhile, CGA can inhibit LPS-induced nuclear translocation of NF-κB and IκB-α phosphorylation in HSCs, as well as NAC (a ROS scavenger). The mRNA expression and the levels of MCP-1 and IL-6 in the culture supernatant of the HSCs in this study were elevated by LPS stimulation and inhibited by CGA treatment, as well as NAC and PDTC (a NF-κB inhibitor). CONCLUSION: Our results indicate that CGA can efficiently inhibit LPS-induced proinflammatory responses in HSCs and the anti-inflammatory effect may be due to the inhibition of LPS/ROS/NF-κB signaling pathway.


Asunto(s)
Antiinflamatorios/farmacología , Ácido Clorogénico/farmacología , Células Estrelladas Hepáticas/efectos de los fármacos , Animales , Línea Celular , Quimiocina CCL2/genética , Quimiocina CCL2/inmunología , Células Estrelladas Hepáticas/inmunología , Proteínas I-kappa B/inmunología , Inflamación/inducido químicamente , Inflamación/metabolismo , Interleucina-6/genética , Interleucina-6/inmunología , Lipopolisacáridos , Inhibidor NF-kappaB alfa , FN-kappa B/inmunología , Ratas , Especies Reactivas de Oxígeno/inmunología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
16.
Oncol Res ; 21(6): 353-63, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-25198665

RESUMEN

MicroRNA-133a has been proven downregulated in many human malignancies and correlated with tumor progression. However, the roles of miR-133a and its related molecular mechanisms in pancreatic cancer are still not clear. Here we found that miR-133a expression was significantly downregulated in pancreatic cancer tissue samples and cell lines by using quantitative real-time RT-PCR. Decreased miR-133a expression was significantly correlated with aggressive clinicopathological features and poor survival. In addition, miR-133a was identified to be a tumor suppressor, as transfection of miR-133a mimics in PANC-1 cells was able to reduce cell proliferation, invasion, and migration and promote cell apoptosis in vitro and suppress tumorigenicity in vivo. Further, we observed an obvious inverse correlation between FSCN1 and miR-133a levels in tumor samples, and FSCN1 was confirmed as a direct target of miR-133a by using Luciferase Reporter Assay. These findings suggest an important role of miR-133a in the molecular etiology of cancer and implicate its potential application in gene therapy of pancreatic cancer.


Asunto(s)
Proteínas Portadoras/genética , MicroARNs/genética , Proteínas de Microfilamentos/genética , Neoplasias Pancreáticas/genética , Interferencia de ARN , Regiones no Traducidas 3' , Adulto , Anciano , Animales , Apoptosis/genética , Secuencia de Bases , Sitios de Unión , Proteínas Portadoras/química , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Xenoinjertos , Humanos , Masculino , MicroARNs/química , Proteínas de Microfilamentos/química , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Neovascularización Patológica , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Pronóstico , Carga Tumoral
17.
Chin J Integr Med ; 19(2): 119-26, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23001460

RESUMEN

OBJECTIVE: To investigate the effects of serum containing Chinese medicine (CM) Sanpi Pingwei (, SPPW) formula on the proliferation and apoptosis of human SGC-7901 cells and the possible mechanism. METHODS: Serum containing CM SPPW formula (SPPW serum) was prepared by a serum pharmacology method. Human SGC-7901 cells were incubated with SPPW serum at three different concentrations and with the anticancer drug 5-fluorouracil (5-FU), respectively. Cell proliferation was assessed by MTT assay, and cell apoptosis was detected by flow cytometry assay. Real-time quantitative polymerase chain reaction (RT-PCR) and Western blot assay were employed to confirm the expressions of Bcl-2, Bax and p53 in SGC-7901 cells at mRNA and protein levels, respectively. RESULTS: SPPW serum suppressed the proliferation of SGC-7901 cells in a time- and dose-dependent manner. The colony forming rate of negative control was 48.2%, while those in the three SPPW serum groups and the 5-FU group decreased significantly (P<0.01). The number of colony forming units in the SPPW high dosage group was significantly smaller than that in the 5-FU group (P<0.01). MTT assay showed that SPPW serum restrained the proliferation of SGC-7901 cells, and the inhibition rate increased significantly in a dose-dependent manner. Annexin V/PI Assay suggested that SPPW serum induced the apoptosis of SGC-7901 cells significantly. RT-PCR and western blot assay indicated that SPPW serum upregulated the protein and mRNA expression levels of Bax and p53 in SGC-7901 cells, but downregulated the protein and mRNA expressions of Bcl-2. CONCLUSIONS: SPPW formula inhibits the proliferation of SGC-7901 cells in vitro and induces the cell apoptosis. It plays an anticancer role by regulating the expressions of Bax, p53 and Bcl-2 in SGC-7901 cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Suero/química , Animales , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Fluorouracilo/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Ensayo de Tumor de Célula Madre , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
18.
Toxicology ; 303: 107-14, 2013 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-23146752

RESUMEN

Chlorogenic acid (CGA) is a type of polyphenol with anti-inflammatory, antioxidant activities. Our previous studies showed CGA could efficiently inhibit carbon tetrachloride (CCl(4))-induced liver fibrosis in rats. However, the specific underlying mechanism remains unclear. The aim of this study is to investigate the effects of CGA on liver inflammation and fibrosis induced by CCl(4) and whether they are related to inhibition of toll-like receptor 4 (TLR4) signaling pathway. Male Sprague-Dawley (SD) rats were administrated CCl(4) together with or without CGA for 8 weeks. Histopathological and biochemical analyses were carried out. The mRNA and protein expression levels of proinflammatory and profibrotic mediators were detected by RT-PCR and Western blot, respectively. The levels of serum proinflammatory cytokines were detected by ELISA. CGA significantly attenuated CCl(4)-induced liver damage and symptoms of liver fibrosis, accompanied by reduced serum transaminase levels, collagen I and α-smooth muscle actin (α-SMA) expression. As compared with the CCl(4)-treated group, the expression levels of TLR4, myeloid differentiation factor 88 (MyD88), inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) were reduced in the treatment group of CCl(4) and CGA, whereas bone morphogenetic protein and activin membrane-bound inhibitor (Bambi) expression was increased. CGA also suppressed CCl(4) induced nuclear factor-κB (NF-κB) activation. Moreover, the hepatic mRNA expression and serum levels of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and interleukin-1ß (IL-1ß) were significantly increased in CCl(4)-treated rats and attenuated by co-treatment with CGA. Our data indicate that CGA can efficiently inhibit CCl(4)-induced liver fibrosis in rats and the protective effect may be due to the inhibition of TLR4/MyD88/NF-κB signaling pathway.


Asunto(s)
Ácido Clorogénico/farmacología , Inflamación/prevención & control , Cirrosis Hepática/prevención & control , Hígado/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Animales , Tetracloruro de Carbono/toxicidad , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/inmunología , Hígado/patología , Cirrosis Hepática/patología , Masculino , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos
19.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 31(7): 921-5, 2011 Jul.
Artículo en Chino | MEDLINE | ID: mdl-21866662

RESUMEN

OBJECTIVE: To clarify the action and possible mechanisms of SPPW, a Chinese herbal preparation consisting of Herba Scutellariae Barbatae, Radix Astragalus, Radix Glycyrrhizae, etc., in suppressing the metastasis of human gastric cancer, by way of observing its effect on the invasive and metastatic capacities of gastric cancer cells. METHODS: In vitro serial sub-cultured human gastric cancer cell line SGC-7901 at the logarithmic growth phase were randomly divided into 5 groups, i.e. the negative control group, the 4 treatment groups intervened respectively with SPPW at three different doses (high, middle, and low), and 5-FU. The adhesion capacities of gastric cancer cells to matrigel were detected by MTT assay 48 h after intervention. The invasive and migratory capacities of gastric cancer cells were determined by Transwell assay. The mRNA and protein expressions of metalloproteinase-9 (MMP-9) and vascular endothelial growth factor (VEGF) in gastric cancer cells were detected by Western blot and Real-time polymerase chain reaction (RT-PCR) respectively. RESULTS: Compared with the negative control group, the adhesive, invasive and migratory capacities of gastric cancer cells were all significantly inhibited in the four treatment groups (P<0.05, P<0.01). In addition, the protein and mRNA expressions of MMP-9 and VEGF were down-regulated (P<0.01). Significant dose-dependent relation existed in the three SPPW treatment groups (P<0.01). Compared with the 5-FU treatment group, the high dose SPPW treatment group showed significant difference in inhibiting the adhesive and metastatic capacities of gastric cancer cells, lowering VEGF protein expression, and mRNA expressions of MMP-9 and VEGF (P<0.01). CONCLUSIONS: SPPW could lower the adhesion of gastric cancer cells to matrigel, and lower the invasion and migration of gastric cancer cells. Meanwhile, it could down-regulate the mRNA and protein expressions of MMP-9 and VEGF, which may possibly be one of its mechanisms for influencing the invasion and metastasis of gastric cancer cells.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Neoplasias Gástricas/patología , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , ARN Mensajero/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA