Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 214
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell Biol Int ; 47(5): 954-968, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36740226

RESUMEN

Irreversible destruction of joints is the hallmark of rheumatoid arthritis (RA). Osteoclasts are the only bone-resorbing cells and play an important role in joint rebuilding. BML-111 (5(S),6(R),7-trihydroxyheptanoic acid methyl ester, C8 H16 O5 ) is a synthetic lipoxin A4 agonist with antioxidant and anti-inflammatory properties. The present study aimed to investigate the effect of BML-111 on osteoclasts in vivo and in vitro, to investigate its therapeutic effect on joint destruction in RA. Cell Counting Kit-8 assay and flow cytometry were used to exclude cytotoxic effects of BML-111 to bone marrow-derived macrophages (BMMs). Then, osteoclasts were differentiated in vitro from BMMs by used macrophage colony-stimulating factor and receptor activator of nuclear factor-κB ligand, and osteoclasts were observed following tartrate-resistant acid phosphatase staining with or without BML-111 treatment. Meanwhile, absorption pit assay and immunofluorescence staining of the fibrous actin ring were used to observe osteoclast function. Moreover, we examined mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) activation. We established collagen-induced arthritis in a rat model and, after treatment with BML-111, joint swelling was measured and the knee joints were processed for histology. We also examined serum and tissue for osteoclastogenesis-related markers. BML-111 inhibited osteoclast formation and differentiation in a time- and concentration-dependent manner, and downregulated the expression levels of MAPK and NF-κB in vitro. Meanwhile, BML-111 effectively alleviated joint structural damage and inhibited osteoclast formation in vivo. BML-111 inhibited osteoclast formation and differentiation in vitro and in vivo, and delayed the progression of joint destruction.


Asunto(s)
Resorción Ósea , Osteoclastos , Ratas , Animales , FN-kappa B/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Resorción Ósea/metabolismo , Resorción Ósea/patología , Diferenciación Celular , Articulación de la Rodilla/metabolismo , Articulación de la Rodilla/patología , Ligando RANK/metabolismo
3.
J Proteome Res ; 19(6): 2471-2482, 2020 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-32283030

RESUMEN

Severe acute pancreatitis (SAP) is caused by complicated biological factors, and revealing its complex pathogenesis by single-target analysis is difficult. Systematic studies have developed slowly because extraction of degradable pancreatic proteins exposed to multiple proteases is challenging. We present integrated whole proteomic and phosphoproteomic profiles of SAP rats based on a modified protein extraction strategy with less protein degradation. Data-dependent acquisition (DDA) and data-independent acquisition (DIA) strategies were applied to select an appropriate method. Total 275 differentially expressed proteins and 757 differentially expressed phosphorylated proteins were identified by DIA-based quantitative proteomics. Several signal transduction pathways, including the AMPK, MAPK, and PI3K-Akt pathways, were enriched in SAP. Up-regulation of phosphorylated proteins involved in the process of TNFA signaling and inflammatory response was also detected in SAP. Our results improve the understanding of SAP development and progression.


Asunto(s)
Pancreatitis , Enfermedad Aguda , Animales , Páncreas , Fosfatidilinositol 3-Quinasas , Proteómica , Ratas
4.
J Cell Mol Med ; 24(2): 1700-1712, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31782248

RESUMEN

Circular RNAs are a class of widespread and diverse endogenous RNAs that may regulate gene expression in various diseases, but their regulation and function in hypertensive renal injury remain unclear. In this study, we generated ribosomal-depleted RNA sequencing data from normal mouse kidneys and from injured mouse kidneys induced by deoxycorticosterone acetate-salt hypertension and identified at least 4900 circRNA candidates. A total of 124 of these circRNAs were differentially expressed between the normal and injured kidneys. Furthermore, we characterized one abundant circRNA, termed circNr1h4, which is derived from the Nr1h4 gene and significantly down-regulated in the injured kidneys. RNA sequencing data and qPCR analysis also showed many microRNAs and mRNAs, including miR-155-5p and fatty acid reductase 1 (Far1), were differentially expressed between the normal and injured kidney and related to circNr1h4. In vitro, the silencing of circNr1h4 or overexpression of miR-155-5p significantly decreased Far1 levels and increased reactive oxygen species. Mechanistic investigations indicated that circNr1h4 acts as a competing endogenous RNA for miR-155-5p, leading to regulation of its target gene Far1. Our study provides novel insight into the molecular mechanisms underlying kidney injury in hypertension, which will be required to develop therapeutic strategies of targeting circRNAs for hypertensive kidney injury.


Asunto(s)
Hipertensión/genética , Riñón/patología , MicroARNs/metabolismo , ARN Circular/metabolismo , Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/metabolismo , Animales , Secuencia de Bases , Acetato de Desoxicorticosterona , Regulación de la Expresión Génica , Masculino , Ratones Endogámicos C57BL , MicroARNs/genética , ARN Circular/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados
5.
J Clin Lab Anal ; 34(3): e23091, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31709646

RESUMEN

BACKGROUND: Reactive oxygen species modulator 1 (ROMO1) is recognized to be involved in cell proliferation and is elevated in serum of various cancer patients. However, ROMO1 had little research in distinguishing between malignant pleural effusions (MPEs) and benign pleural effusions (BPEs). METHODS: Malignant pleural effusion samples from patients with non-small-cell lung cancer (NSCLC) and benign pleural effusion (BPE) samples containing tuberculous and inflammatory pleural effusions were collected. The samples were tested for ROMO1, pleural effusion adenosine deaminase (pADA), pleural effusion carbohydrate antigen (pCA125, pCA153, pCA199), pleural effusion ferritin (pFER), and pleural effusion lactate dehydrogenase (pLDH) levels, and the other relevant partial clinical data that were gathered were used to conduct statistical analysis. RESULTS: The ROMO1, pCA125, pCA199, pCA153, pADA + ROMO1, pCA153 + ROMO1, pCA125 + ROMO1, and pCA199 + ROMO1 levels in MPE were appreciably higher in comparison with BPE group (all P = .000). The concentration of pADA in MPE was markedly lower than BPE (P = .000). When the cutoff = 0.38, the sensitivity of combined detection of ROMO1 + pADA is 98.67% and the specificity is 70.73%, respectively, and the AUC (0.941) is the highest among other parameters. CONCLUSION: The combined detection of ROMO1 + ADA in pleural effusion is an effective biomarker for identifying MPE caused by NSCLC.


Asunto(s)
Adenosina Desaminasa/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Derrame Pleural Maligno/metabolismo , Área Bajo la Curva , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Femenino , Humanos , Neoplasias Pulmonares/diagnóstico , Masculino , Persona de Mediana Edad , Derrame Pleural Maligno/diagnóstico , Curva ROC , Especies Reactivas de Oxígeno , Sensibilidad y Especificidad
6.
J Cell Mol Med ; 23(3): 2020-2031, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30585412

RESUMEN

Peroxisomes play a central role in lipid metabolism. We previously demonstrated that Pex11a deficiency impairs peroxisome abundance and fatty acid ß-oxidation and results in hepatic triglyceride accumulation. The role of Pex11a in dyslipidaemia and obesity is investigated here with Pex11a knockout mice (Pex11a-/- ). Metabolic phenotypes including tissue weight, glucose tolerance, insulin sensitivity, cholesterol levels, fatty acid profile, oxygen consumption, physical activity were assessed in wild-type (WT) and Pex11a-/- fed with a high-fat diet. Molecular changes and peroxisome abundance in adipose tissue were evaluated through qRT-PCR, Western blotting, and Immunofluorescence. Pex11a-/- showed increased fat mass, decreased skeletal muscle, higher cholesterol levels, and more severely impaired glucose and insulin tolerance. Pex11a-/- consumed less oxygen, indicating a decrease in fatty acid oxidation, which is consistent with the accumulation of very long- and long-chain fatty acids. Adipose palmitic acid (C16:0) levels were elevated in Pex11a-/- , which may be because of dramatically increased fatty acid synthase mRNA and protein levels. Furthermore, Pex11a deficiency increased ventricle size and macrophage infiltration, which are related to the reduced physical activity. These data demonstrate that Pex11a deficiency impairs physical activity and energy expenditure, decreases fatty acid ß-oxidation, increases de novo lipogenesis and results in dyslipidaemia and obesity.


Asunto(s)
Dislipidemias/metabolismo , Proteínas de la Membrana/metabolismo , Obesidad/metabolismo , Tejido Adiposo/metabolismo , Animales , Colesterol/metabolismo , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético/fisiología , Ácidos Grasos/metabolismo , Resistencia a la Insulina/fisiología , Metabolismo de los Lípidos/fisiología , Lipogénesis/fisiología , Lipólisis/fisiología , Hígado/metabolismo , Ratones , Ratones Noqueados , Peroxisomas/metabolismo , Triglicéridos/metabolismo
7.
Cancer Cell Int ; 19: 107, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31049029

RESUMEN

BACKGROUND: Compelling lines of evidence indicate that DNA methylation of non-coding RNAs (ncRNAs) plays critical roles in various tumour progression. In addition, the differential methylation of ncRNAs can predict prognosis of patients. However, little is known about the clear relationship between DNA methylation profile of ncRNAs and the prognosis of pancreatic adenocarcinoma (PAC) patients. METHODS: The data of DNA methylation, RNA-seq, miRNA-seq and clinical features of PAC patients were collected from TCGA database. The DNA methylation profile was obtained using the Infinium HumanMethylation450 BeadChip array. LASSO regression was performed to construct two methylation-based classifiers. The risk score of methylation-based classifiers was calculated for each patient, and the accuracy of the classifiers in predicting overall survival (OS) was examined by ROC curve analysis. In addition, Cox regression models were utilized to assess whether clinical variables and the classifiers were independent prognostic factors for OS. The targets of miRNA and the genes co-expressed with lncRNA were identified with DIANA microT-CDS and the Multi-Experiment Matrix (MEM), respectively. Moreover, DAVID Bioinformatics Resources were applied to analyse the functional enrichment of these targets and co-expressed genes. RESULTS: A total of 4004 CpG sites of miRNA and 11,259 CpG sites of lncRNA were screened. Among these CpG sites, 8 CpG sites of miRNA and 7 CpG sites of lncRNA were found with regression coefficients. By multiplying the sum of methylation degrees of the selected CpGs with these coefficients, two methylation-based classifiers were constructed. The classifiers have shown good performance in predicting the survival rate of PAC patients at varying follow-up times. Interestingly, both of these two classifiers were predominant and independent factors for OS. Furthermore, functional enrichment analysis demonstrated that aberrantly methylated miRNAs and lncRNAs are related to calcium ion transmembrane transport and MAPK, Ras and calcium signalling pathways. CONCLUSION: In the present study, we identified two methylation-based classifiers of ncRNA associated with OS in PAC patients through a comprehensive analysis of miRNA and lncRNA profiles. We are the first group to demonstrate a relationship between the aberrant DNA methylation of ncRNAs and the prognosis of PAC, and this relationship would contribute to individualized PAC therapy.

8.
Cell Commun Signal ; 17(1): 145, 2019 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-31718671

RESUMEN

BACKGROUND: Notch1 signalling is a stem-cell-related pathway that is essential for embryonic development, tissue regeneration and organogenesis. However, the role of Notch1 in the formation of myofibroblasts and fibrosis in kidneys following injury remains unknown. METHODS: The activity of Notch1 signalling was evaluated in fibrotic kidneys in CKD patients and in ureteral obstructive models in vivo and in cultured fibroblasts and TECs in vitro. In addition, the crosstalk of Notch1 with TGF-ß1/Smad2/3 signalling was also investigated. RESULTS: Notch1 activity was elevated in fibrotic kidneys of rat models and patients with chronic kidney disease (CKD). Further study revealed that epithelial and interstitial Notch1 activity correlated with an α-SMA-positive myofibroblastic phenotype. In vitro, injury stimulated epithelial Notch1 activation and epithelial-mesenchymal transition (EMT), resulting in matrix deposition in tubular epithelial cells (TECs). Additionally, interstitial Notch1 activation in association with fibroblast-myofibroblast differentiation (FMD) in fibroblasts mediated a myofibroblastic phenotype. These TGF-ß1/Smad2/3-dependent phenotypic transitions were abolished by Notch1 knockdown or a specific antagonist, DAPT, and were exacerbated by Notch1 overexpression or an activator Jagged-1-Fc chimaera protein. Interestingly, as a major driving force behind the EMT and FMD, TGF-ß1, also induced epithelial and interstitial Notch1 activity, indicating that TGF-ß1 may engage in crosstalk with Notch1 signalling to trigger fibrogenesis. CONCLUSION: These findings suggest that epithelial and interstitial Notch1 activation in kidneys following injury contributes to the myofibroblastic phenotype and fibrosis through the EMT in TECs and to the FMD in fibroblasts by targeting downstream TGF-ß1/Smad2/3 signalling.


Asunto(s)
Diaminas/farmacología , Células Epiteliales/efectos de los fármacos , Fibrosis/tratamiento farmacológico , Miofibroblastos/efectos de los fármacos , Receptor Notch1/antagonistas & inhibidores , Tiazoles/farmacología , Animales , Línea Celular , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Masculino , Miofibroblastos/metabolismo , Fenotipo , Ratas , Ratas Sprague-Dawley , Receptor Notch1/metabolismo , Transducción de Señal/efectos de los fármacos
9.
Exp Cell Res ; 367(2): 274-281, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29627320

RESUMEN

Hepatocellular carcinoma (HCC) is one of the most common cancers diagnosed worldwide. However, the mechanism underlying HCC pathogenesis remains unknown. In the present study, TRIM24 was found increased in human HCC clinical samples and positively correlated with HCC tumor grade. Furthermore, TRIM24 knockdown inhibits proliferation and migration in a human HCC cell line in vitro while also inhibiting tumor growth in vivo. Mechanistically, TRIM24 appears to promote liver tumor development via AMPK signaling as AMPK knockdown alleviated the in vitro and in vivo effects of TRIM24 knockdown in a human HCC cell line. Taken together, these data enhance our understanding of HCC development in addition to highlighting TRIM24-regulated AMPK signaling as a potential therapeutic target for HCC treatment.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Carcinoma Hepatocelular/metabolismo , Proteínas Portadoras/metabolismo , Neoplasias Hepáticas/metabolismo , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Transducción de Señal
10.
Mediators Inflamm ; 2019: 9019404, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31097921

RESUMEN

Our previous studies verified the potent anti-inflammatory effects against severe acute pancreatitis (SAP) of AT-Lipoxin A4 and their analogues. However, the anti-inflammatory effects of AT-Lipoxin A4 on SAP-associated lung injury are not thoroughly known. We used western blot, polymerase chain reaction (PCR), and immunofluorescence to investigate the downregulation of TNF-α signals in cellular and animal models of SAP-associated lung injury following AT-Lipoxin A4 intervention. In vitro, we found that AT-Lipoxin A4 markedly suppressed protein expression in TNF-α signals in human pulmonary microvascular endothelial cell, such as tumor necrosis factor receptor-associated factor 2 (TRAF2), TNF-R1-associated death domain (TRADD), receptor-interacting protein (RIP), vascular cell adhesion molecule-1 (VCAM-1), and E-selectin. Moreover, AT-Lipoxin A4 inhibited downstream signals activated by TNF-α, including NF-κB/p65, JNK/MAPK, and ERK/MAPK. In vivo, AT-Lipoxin A4 significantly decreased pathological scores of the pancreas and lungs and the serum levels of IL-6 and TNF-α. Immunofluorescence, western blotting, and real-time PCR assay showed that AT-Lipoxin A4 significantly attenuated the expression of TNF-R1, TRADD, TRAF2, and RIP in the lungs of SAP rats. In addition, the activation of NF-κB was also downregulated by AT-Lipoxin A4 administration as compared with SAP rats. AT-Lipoxin A4 could inhibit the production of proinflammatory mediators and activation of TNF-α downstream signals such as NF-κB and MAPK. Downregulation of TNF-α signals by AT-Lipoxin A4 may be a significant mechanism in the attenuation of SAP-associated lung injury.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Lipoxinas/metabolismo , FN-kappa B/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Lesión Pulmonar Aguda/genética , Animales , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Humanos , Interleucina-6/metabolismo , Lipoxinas/genética , Masculino , FN-kappa B/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
11.
J Cell Physiol ; 233(5): 4044-4055, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28657147

RESUMEN

The identification and characterization of long non-coding RNAs (lncRNAs) in diverse biological process has currently developed rapidly. LncRNA-PVT1, located adjacent to the MYC locus on chromosomal region 8q24, has been reported to be associated with many biological processes. However, the function and mechanism of PVT1 in pancreatic carcinoma (PC) is poorly understood. In this present study, we first measured the level of PVT1 in the PC cell lines and tissues by quantitative real-time PCR (qRT-PCR), and then employed loss-of-function and gain-of-function approaches to explore the association between PVT1 expression levels and PC cell proliferation/migration ability. Furthermore, bioinformatics analysis was utilized to show that PVT1 contains binding site for miR-448 and an inverse correlation between PVT1 and miR-448 was obtained in PC specimens. Additionally, dual luciferase reporter assay, RNA-binding protein immunoprecipitation (RIP) and applied biotin-avidin pulldown system were applied to further confirm that PVT1 directly bind with microRNA binding site harboring in the PVT1 sequence. Then, SERBP1 was identified as a target of miR-448 according to the gene expression array analysis of PC clinical samples. Together, we revealed that PVT1 functions as an endogenous "sponge" by competing for miR-448 binding to regulate the miRNA target SERBP1 and, therefore, promotes the proliferation and migration of PC cells.


Asunto(s)
MicroARNs/genética , Neoplasias Pancreáticas/genética , ARN Largo no Codificante/genética , Sitios de Unión , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pancreáticas/patología , Proteínas de Unión al ARN
12.
Cell Physiol Biochem ; 46(1): 82-92, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29587268

RESUMEN

BACKGROUND/AIMS: The activation of hepatic stellate cells (HSCs) is considered as a pivotal event in liver fibrosis and epithelial-mesenchymal transition (EMT) process has been reported to be involved in HSC activation. It is known that microRNAs (miRNAs) play a pro-fibrotic or anti-fibrotic role in HSC activation. Recently, emerging studies show that miR-30a is down-regulated in human cancers and over-expression of miR-30a inhibits tumor growth and invasion via suppressing EMT process. However, whether miR-30a could regulate EMT process in HSC activation is still unclear. METHODS: miR-30a expression was quantified using real-time PCR in carbon tetrachloride (CCl4)-induced rat liver fibrosis, activated HSCs and patients with cirrhosis. Roles of miR-30a in liver fibrosis in vivo and in vitro were also analyzed. Luciferase activity assays were performed to examine the binding of miR-30a to the 3'-untranslated region of snail family transcriptional repressor 1 (Snai1). RESULTS: miR-30a was down-regulated in human cirrhotic tissues. In CCl4 rats, reduced miR-30a was found in fibrotic liver tissues as well as isolated HSCs. There was a significant reduction in miR-30a in primary HSCs during culture days. miR-30a over-expression resulted in the suppression of CCl4-induced liver fibrosis. Restoration of miR-30a led to the inhibition of HSC activation including cell proliferation, α-SMA and collagen expression. Notably, miR-30a inhibited EMT process, with a reduction in TGF-ß1 and Vimentin as well as an increase in GFAP and E-cadherin. miR-30a induced a significant reduction in Snai1 protein expression when compared with the control. Interestingly, Snail protein expression was increased during liver fibrosis, indicating that there may be a negative correlation between miR-30a level and Snai1 protein expression. Further studies demonstrated that Snai1 was a target of miR-30a. CONCLUSION: Our results suggest that miR-30a inhibits EMT process, at least in part, via reduction of Snai1, leading to the suppression of HSC activation in liver fibrosis.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Cirrosis Hepática/patología , MicroARNs/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Regiones no Traducidas 3' , Actinas/metabolismo , Animales , Antagomirs/metabolismo , Secuencia de Bases , Cadherinas/metabolismo , Tetracloruro de Carbono/toxicidad , Proliferación Celular , Células Cultivadas , Regulación hacia Abajo , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Células Estrelladas Hepáticas/citología , Células Estrelladas Hepáticas/metabolismo , Humanos , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/genética , Masculino , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Ratas , Ratas Sprague-Dawley , Alineación de Secuencia , Factores de Transcripción de la Familia Snail/antagonistas & inhibidores , Factores de Transcripción de la Familia Snail/genética , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Vimentina/metabolismo
13.
BMC Endocr Disord ; 18(1): 49, 2018 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-30053902

RESUMEN

BACKGROUND: Glycaemic control is one of the most effective strategies for the treatment of diabetes-related erectile dysfunction (DMED). Compared to conventional anti-diabetic drugs and insulin, islet transplantation is more effective in the treatment of diabetic complications. The aim of this study was to investigate the efficacy of islet transplantation for reversing advanced-stage DMED in rats and to observe its influence on corpus cavernosum fibrosis. METHODS: Wistar rats were intraperitoneally injected with streptozotocin to establish a diabetes model. After 12 weeks, the rats were divided into 4 groups: diabetic, insulin, islet transplantation, and normal control. Following supplementation, the changes in blood glucose and weight were determined sequentially. Penile erectile function was evaluated by apomorphine experiments in the fourth week, and the penile corpus cavernosum was also collected for assessment by Masson staining, immunohistochemistry and Western blot to observe the spongy tissue and the related cellular changes at the molecular level. RESULTS: Islet transplantation significantly ameliorated penile erectile function in advanced-stage diabetic rats. The ratio of corpus cavernosum smooth muscle cells to fibroblasts and the expression level of α-SMA in the islet transplantation group were significantly higher than those in the diabetic and insulin groups. In addition, the expression levels of TGF-ß1, p-Samd2, and connective tissue growth factor (CTGF) in the islet transplantation and insulin groups were much lower than those in the diabetic group, while those in the islet transplantation group were significantly lower than those in the insulin group. CONCLUSIONS: Our findings strongly suggest that islet transplantation can promote the regeneration of smooth muscle cells and ameliorate corpus cavernosum fibrosis to restore its normal structure in advanced-stage diabetic rats. The possible mechanism of ameliorating corpus cavernosum fibrosis by islet transplantation may be associated with improvement of the hyperglycaemic status in diabetic rats, thereby inhibiting the TGF-ß1/Samd2/CTGF pathway.


Asunto(s)
Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/terapia , Trasplante de Islotes Pancreáticos , Enfermedades del Pene/terapia , Pene/patología , Animales , Complicaciones de la Diabetes/terapia , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Tipo 1/inducido químicamente , Diabetes Mellitus Tipo 1/complicaciones , Disfunción Eréctil/etiología , Disfunción Eréctil/terapia , Fibrosis/etiología , Fibrosis/terapia , Masculino , Enfermedades del Pene/etiología , Ratas , Ratas Wistar , Estreptozocina
14.
Mol Ther ; 25(1): 205-217, 2017 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-28129115

RESUMEN

Homeobox transcript antisense RNA (HOTAIR), as a long intergenic non-coding RNA (lincRNA), is upregulated in various cancers and involved in diverse cellular functions. However, its role in liver fibrosis is unclear. In this study, HOTAIR expression was upregulated in hepatic stellate cells (HSCs) in vivo and in vitro during liver fibrosis. HOTAIR knockdown suppressed HSC activation including α-smooth muscle actin (α-SMA) and typeIcollagen in vitro and in vivo. Both HSC proliferation and cell cycle were inhibited by HOTAIR knockdown. Notably, inhibition of HOTAIR led to an increase in PTEN, associated with the loss of DNA methylation. miR-29b-mediated control of PTEN methylation was involved in the effects of HOTAIR knockdown. HOTAIR was confirmed a target of miR-29b and lack of the miR-29b binding site in HOTAIR prevented the suppression of miR-29b, suggesting HOTAIR contributes to PTEN expression downregulation via sponging miR-29b. Interestingly, increased HOTAIR was also observed in hepatocytes during liver fibrosis. Loss of HOTAIR additionally led to the increase in PTEN and the reduction in typeIcollagen in hepatocytes. Collectively, we demonstrate that HOTAIR downregulates miR-29b expression and attenuates its control on epigenetic regulation, leading to enhanced PTEN methylation, which contributes to the progression of liver fibrosis.


Asunto(s)
Epigénesis Genética , Regulación de la Expresión Génica , Cirrosis Hepática/genética , MicroARNs/genética , Fosfohidrolasa PTEN/genética , ARN Largo no Codificante/genética , Regiones no Traducidas 3' , Animales , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Técnicas de Silenciamiento del Gen , Células Estrelladas Hepáticas/metabolismo , Humanos , Cirrosis Hepática/patología , Ratones , Modelos Biológicos , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-akt/metabolismo , ADN Metiltransferasa 3B
15.
Int J Med Sci ; 15(13): 1433-1442, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30443162

RESUMEN

Renal tubule cell apoptosis plays a pivotal role in the progression of chronic renal diseases. The previous study indicates that Sirolimus is effective on unilateral ureteral obstruction (UUO)-induced renal fibrosis. However, the role of Sirolimus in renal tubular apoptosis induced by UUO has not yet been addressed. The aim of this study was to determine the role of Sirolimus in renal tubular apoptosis induced by UUO. Male Sprague-Dawley rats were divided into three groups, sham-operated rats, and after which unilateral ureteral obstruction (UUO) was performed: non-treated and sirolimus-treated (1mg/kg). After 4, 7 and 14 d, animals were sacrificed and blood, kidney tissue samples were collected for analyses. Histologic changes and interstitial collagen were determined microscopically following HE and Masson's trichrome staining. The expression of PCNA was investigated using immunohistochemistry and the expression of Bcl-2, Bax, caspase-9, and caspase-3 were investigated using Western blot in each group. Tubular apoptotic cell deaths were assessed by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) assay. Sirolimus administration resulted in a significant reduction in tubulointerstitial fibrosis scores. After UUO, there was an increase in tubular and interstitial apoptosis in untreated controls as compared to Sirolimus treatment rats (P<0.05). In addition, the expression of PCNA, Bcl-2, Bax, caspase-9, and caspase-3 in obstructed kidney was characterized by immunohistochemistry and Western blot analyses demonstrating that sirolimus treatment significantly reduced PCNA, Bax, caspase-9 and cleaved caspase-3 expression compared to those observed in controls (P<0.05), whereas, Bcl-2 in the obstructed kidney were decreased in untreated controls compared to Sirolimus treatment rats subjected to the same time course of obstruction (P<0.05). We demonstrated a marked renoprotective effect of sirolimus by inhibition of UUO-induced renal tubular apoptosis in vivo.


Asunto(s)
Sirolimus/uso terapéutico , Obstrucción Ureteral/tratamiento farmacológico , Obstrucción Ureteral/metabolismo , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Riñón/efectos de los fármacos , Riñón/metabolismo , Masculino , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas
16.
Clin Lab ; 64(5): 749-757, 2018 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-29739052

RESUMEN

BACKGROUND: Pancreatic cancer is one of the most lethal cancers with limited treatment options. Gemcitabine has been the standard drug for patients with advanced pancreatic cancer. Dasatinib is a competitive inhibitor of Src kinase, which has shown promise in treatment of pancreatic cancer. Several studies have revealed the drug resistant mechanism of gemcitabine or dasatinib in human cancers; however, few reports focused on the different mechanisms of gemcitabine and dasatinib resistance in pancreatic cancer. Here, we integrate mRNA and miRNA expression profiles to achieve it. METHODS: Two mRNA expression profiles were downloaded from GEO database. The differentially expressed genes (DEGs) were identified with |fold change| ≥ 2 and p-value ≤ 0.05. Further function of the DEGs were annotated with GO and KEGG pathway enrichment. Finally, the mRNA-miRNA interaction networks were constructed to explore the molecular mechanism. RESULTS: Results showed that 116 and 238 DEGs were detected in gemcitabine-resistant cell lines and dasatinib-resistant cell lines respectively. Meanwhile, 4 common DEGs were identified in both resistant cell lines, which can clearly divide all cell lines into different sub-groups. KEGG pathway enrichment analysis displayed that the DEGs of both gemcitabine-resistant cell lines and dasatinib-resistant cell lines can map to drug metabolism-cytochrome P450 and metabolism of xenobiotics by cytochrome P450, while DEGs of gemcitabine-resistant cell lines can also map to several metabolism related pathways and dasatinib-resistant cell lines for several cancer related pathways. GO annotation analysis showed that the DEGs of gemcitabine-resistant cell lines and dasatinib-resistant cell lines can also be categorized into drug metabolism. Additionally, the miRNA-mRNA regulation network of gemcitabine-resistant cell lines revealed 16 DEGs were regulated by 6 miRNAs, indicating that these miRNAs may play a key role in gemcitabine treatment of pancreatic cancer. CONCLUSIONS: The difference of gemcitabine resistance in pancreatic cancer were explored by mechanism comparison via the mRNA and miRNA expression profile. These findings support strategies to target molecules and relevant pathways for improving the efficacy of chemotherapy in pancreatic cancer patients.


Asunto(s)
Dasatinib/farmacología , Desoxicitidina/análogos & derivados , Resistencia a Antineoplásicos/genética , MicroARNs/genética , Neoplasias Pancreáticas/genética , ARN Mensajero/genética , Antimetabolitos Antineoplásicos/farmacología , Antineoplásicos/farmacología , Línea Celular Tumoral , Desoxicitidina/farmacología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Ontología de Genes , Redes Reguladoras de Genes , Humanos , Neoplasias Pancreáticas/patología , Gemcitabina
17.
J BUON ; 23(1): 200-204, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29552784

RESUMEN

Prostate cancer is the most frequently diagnosed malignancy in men and the second major reason of cancer death in males. Currently, there are no viable options available for the treatment of advanced-stage prostate cancer. Against this backdrop, the present study aimed to study the anticancer effect of panaxatriol against prostate DU-15 cancer cells. METHODS: MTT cell viability assay evaluated the effects of the drug on cell cytotoxicity, while clonogenic assay was used to assess the effects on colony formation in DU-15 cells. Apoptotic effects were evaluated by DAPI staining using fluorescence microscopy. Effects on reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) were evaluated by flow cytometry using DCFH-DA and DiOC6. Effects on cell cycle were measured by flow cytometry, while cell migration tendency of the cells was evaluated by in vitro wound healing assay. RESULTS: The results indicated that panaxatriol exerts dosedependent cytotoxic effects on prostate DU-15 cancer cells. The IC50 of panaxatriol was 30 µM. Panaxatriol was found to exert its anticancer activity through induction of apoptosis. The apoptosis of DU-15 cancer cells was prompted by ROSmediated alterations in mitochondrial membrane potential. Additionally, panaxatriol induced sub-G1 cell cycle arrest and suppressed the DU-15 cell migration ability in a concentration-dependent manner. CONCLUSION: Taken together, we strongly believe that panaxatriol may prove handy in the treatment and management of prostate cancer and deserves further research.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Ginsenósidos/farmacología , Mitocondrias/efectos de los fármacos , Panax/química , Neoplasias de la Próstata/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Humanos , Masculino , Metaloproteinasas de la Matriz/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Especies Reactivas de Oxígeno/metabolismo
18.
Cancer Sci ; 108(11): 2130-2141, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28795470

RESUMEN

Dermokine (DMKN) was first identified in relation to skin lesion healing and skin carcinoma. Recently, its expression was associated with pancreatic cancer tumorigenesis, although its involvement remains poorly understood. Herein, we showed that DMKN loss of function in Patu-8988 and PANC-1 pancreatic cancer cell lines resulted in reduced phosphorylation of signal transducer and activator of transcription 3, and increased activation of ERK1/2 and AKT serine/threonine kinase. This decreased the proliferation ability of pancreatic ductal adenocarcinoma (PDAC) cells. In addition, DMKN knockdown decreased the invasion and migration of PDAC cells, partially reversed the epithelial-mesenchymal transition, retarded tumor growth in a xenograft animal model by decreasing the density of microvessels, and attenuated the distant metastasis of human PDAC in a mouse model. Taken together, these data suggested that DMKN could be a potential prognostic biomarker and therapeutic target in pancreatic cancer.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Pancreáticas/genética , Proteínas/genética , Factor de Transcripción STAT3/genética , Animales , Carcinogénesis/genética , Movimiento Celular/genética , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Técnicas de Silenciamiento del Gen , Humanos , Péptidos y Proteínas de Señalización Intercelular , Ratones , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Metástasis de la Neoplasia , Neoplasias Pancreáticas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Cell Physiol Biochem ; 42(6): 2145-2158, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28813705

RESUMEN

BACKGROUND/AIMS: Pancreatic carcinoma (PC) is the one of the most common and malignant cancers worldwide. LncRNA taurine upregulated gene 1 (TUG1) was initially identified as a transcript upregulated by taurine, and the abnormal expression of TUG1 has been reported in many cancers. However, the biological role and molecular mechanism of TUG1 in PC still needs further investigation. METHODS: Quantitative real-time PCR (qRT-PCR) was performed to measure the expression of TUG1 in PC cell lines and tissues. MTT and colony formation assays were used to measure the effect of TUG1 on cell proliferation. A wound healing assay, transwell assay and western blot assay were employed to determine the effect of TUG1 on cell migration and the epithelial mesenchymal transition (EMT) phenotype. RNA-binding protein immunoprecipitation (RIP) and a biotin-avidin pulldown system were performed to confirm the interaction between miR-328 and TUG1. A gene expression array analysis using clinical samples and RT-qPCR suggested that enhancer of zeste homolog 2 (EZH2) was a target of miR-382 in PC. RESULTS: In this study, we reported that TUG1 was overexpressed in PC tissues and cell lines, and high expression of TUG1 predicted poor prognosis. Further experiments revealed that overexpressed TUG1 promoted cell proliferation, migration and contributed to EMT formation, whereas silenced TUG1 led to opposing results. Additionally, luciferase reporter assays, an RIP assay and an RNA-pulldown assay demonstrated that TUG1 could competitively sponge miR-382 and thereby regulate EZH2. CONCLUSION: Collectively, these findings revealed that TUG1 functions as an oncogenic lncRNA that promotes tumor progression, at least partially, by functioning as an endogenous 'sponge' and competing for miR-382 binding to the miRNA target EZH2.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/metabolismo , MicroARNs/metabolismo , Neoplasias Pancreáticas/patología , ARN Largo no Codificante/metabolismo , Anciano , Antagomirs/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Proteína Potenciadora del Homólogo Zeste 2/genética , Transición Epitelial-Mesenquimal , Femenino , Células HEK293 , Humanos , Inmunoprecipitación , Masculino , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Persona de Mediana Edad , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/mortalidad , Pronóstico , Interferencia de ARN , ARN Largo no Codificante/genética , Alineación de Secuencia , Regulación hacia Arriba
20.
Cell Physiol Biochem ; 43(6): 2242-2252, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29073595

RESUMEN

BACKGROUND/AIMS: Recently, microRNAs (miRNAs) have been demonstrated to act as regulators of activation of hepatic stellate cells (HSCs). It is well known that the main profibrogenic inducer transforming growth factor-ß1 (TGF-ß1) contributes to HSC activation, which is a key event in liver fibrosis. Increasing studies show that miR-9-5p is down-regulated in liver fibrosis and restoration of miR-9-5p limits HSC activation. However, the role of miR-9-5p in TGF-ß1-induced HSC activation is still not clear. METHODS: miR-9-5p expression was quantified using real-time PCR in chronic hepatitis B (CHB) patients and TGF-ß1-treated LX-2 cells. In CHB patients, histological activity index (HAI) and fibrosis stages were assessed using the Ishak scoring system. Effects of miR-9-5p on liver fibrosis in vivo and in vitro were analyzed. Luciferase activity assays were performed to examine the binding of miR-9-5p to the 3'-untranslated region of type I TGF-ß receptor (TGFBR1) as well as TGFBR2. RESULTS: Compared with healthy controls, miR-9-5p was reduced in CHB patients. There was a lower miR-9-5p expression in CHB patients with higher fibrosis scores or HAI scores. miR-9-5p was down-regulated by TGF-ß1 in a dose-dependent manner. TGF-ß1-induced HSC activation including cell proliferation, α-SMA and collagen expression was blocked down by miR-9-5p. Notably, miR-9-5p ameliorates carbon tetrachloride-induced liver fibrosis. As determined by luciferase activity assays, TGFBR1 and TGFBR2 were targets of miR-9-5p. Further studies demonstrated that miR-9-5p inhibited TGF-ß1/Smads pathway via TGFBR1 and TGFBR2. Interestingly, promoter methylation was responsible for miR-9-5p down-regulation in liver fibrosis. The relationship between miR-9-5p expression and methylation was confirmed in CHB patients and TGF-ß1-treated cells. CONCLUSION: Our results demonstrate that miR-9-5p could inhibit TGF-ß1-induced HSC activation through TGFBR1 and TGFBR2. Loss of miR-9-5p is associated with its methylation status in liver fibrosis.


Asunto(s)
MicroARNs/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Regiones no Traducidas 3' , Actinas/genética , Actinas/metabolismo , Adulto , Animales , Antagomirs/metabolismo , Secuencia de Bases , Tetracloruro de Carbono/toxicidad , Línea Celular , Proliferación Celular/efectos de los fármacos , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Femenino , Células Estrelladas Hepáticas/citología , Células Estrelladas Hepáticas/metabolismo , Hepatitis B Crónica/genética , Hepatitis B Crónica/metabolismo , Hepatitis B Crónica/patología , Humanos , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Persona de Mediana Edad , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/genética , Alineación de Secuencia , Factor de Crecimiento Transformador beta1/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA