Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Biochem Genet ; 62(1): 59-76, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37248373

RESUMEN

Hepatoblastoma (HB) is a common primary liver malignant tumor in children. Long non-coding RNAs (lncRNAs) are closely engaged in HB progression. The role and regulatory molecule mechanism of lncRNA small nucleolar RNA host gene 1 (SNHG1) in HB remain unclear. Through qRT-PCR or western blot, we found that SNHG1 and proviral integration site for moloney murine leukemia virus 3 (PIM3) were elevated but miR-6838-5p was decreased in HB cells. Cell biology experiments revealed that SNHG1 depletion or miR-6838-5p upregulation suppressed cell proliferation, migration, and invasion of HB cells. Mechanistically, luciferase activity assay validated that miR-6838-5p could interact with SNHG1 or PIM3. SNHG1 up-regulated PIM3 expression via sponging miR-6838-5p. Moreover, miR-6838-5p inhibitor abolished SNHG1 depletion-mediated suppression of malignant behaviors in HB cells. PIM3 overexpression neutralized miR-6838-5p mimics-mediated repression of malignant phenotypes in HB cells. Furthermore, miR-6838-5p overexpression suppressed RhoA activation, which was restored by PIM3 upregulation. What's more, the results at the cellular level were further verified by nude mice tumor formation experiment. In conclusion, SNHG1 regulated miR-6838-5p/PIM3/RhoA axis to promote malignant phenotypes of HB, which might provide novel therapeutic target for HB treatment.


Asunto(s)
Hepatoblastoma , MicroARNs , ARN Largo no Codificante , Animales , Ratones , Niño , Humanos , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Hepatoblastoma/genética , Ratones Desnudos , Proliferación Celular/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Movimiento Celular/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Serina-Treonina Quinasas/genética
2.
Am J Physiol Cell Physiol ; 325(3): C796-C806, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37575056

RESUMEN

We aimed to examine impacts and functional mechanism of circular RNA forkhead box N2 (FOXN2) in tacrolimus (TAC)- and dexamethasone (Dex)-induced lipid metabolism disorders. RNA level and protein contents in TAC, Dex, or combined TAC- plus Dex-treated patients and Huh-7 cells were measured utilizing quantitative real-time (qRT)-PCR and western blotting assays measured the formation of lipid droplet. Total cholesterol (TC) and triglyceride (TG) levels were determined using corresponding commercial kits and Oil red O staining. RNA immunoprecipitation and RNA pull-down verified the binding relationship among circFOXN2, polypyrimidine tract binding protein 1 (PTBP1) and fatty acid synthase (FASN). Male C57BL/6 mice were used to establish a dyslipidemia mouse model to validate the discoveries at the cellular level. Dex treatment significantly promoted TAC-mediated increase of TC and TG in serum samples and Huh-7 cells. Moreover, circFOXN2 was reduced but FASN was elevated in TAC-treated Huh-7 cells, and these expression trends were markedly enhanced by Dex cotreatment. Overexpression of circFOXN2 could reverse the accumulation of TC and TG and the upregulation of FASN and sterol regulatory element binding transcription factor 2 (SREBP2) mediated by Dex and TAC cotreatment. Mechanistically, circFOXN2 reduced FASN mRNA stability by recruiting PTBP1. The protective roles of circFOXN2 overexpression on lipid metabolism disorders were weakened by FASN overexpression. In vivo finding also disclosed that circFOXN2 greatly alleviated the dysregulation of lipid metabolism triggered by TAC plus Dex. CircFOXN2 alleviated the dysregulation of lipid metabolism induced by the combination of TAC and Dex by modulating the PTBP1/FASN axis.NEW & NOTEWORTHY Collectively, our experiments revealed for the first time that circFOXN2 alleviated the Dex- and TAC-induced dysregulation of lipid metabolism by regulating the PTBP1/FASN axis. These findings suggested that circFOXN2 and FASN might be candidate targets for the treatment of Dex- and TAC-induced metabolic disorders.


Asunto(s)
Dislipidemias , Trasplante de Hígado , Ratones , Animales , Masculino , Glucocorticoides , Tacrolimus/metabolismo , Ratones Endogámicos C57BL , Ácido Graso Sintasas , Dislipidemias/inducido químicamente , Dislipidemias/tratamiento farmacológico , Dislipidemias/genética , ARN/metabolismo , Estabilidad del ARN , Hígado/metabolismo
3.
Tissue Cell ; 81: 101989, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36642006

RESUMEN

This study is to explore the mechanism of KDM1A-regulated hepatoblastoma (HB) development. Cancerous and paracancer tissues of 30 HB patients were collected for detection of KDM1A and DKK3 expression. HuH-6 and HepG2 cells were subjected to assays of cellular activities after treatment with sh-KDM1A, sh-DKK3, and/or XAV-939 (an inhibitor of the Wnt/ß-catenin pathway). Chromatin immunoprecipitation was used to determine the interaction of KDM1A with DKK3. Nude mice were injected with HuH-6 cells in which KDM1A was knocked down. KDM1A was highly expressed and DKK3 was lowly expressed in HB patients. Knockdown of KDM1A reduced the proliferative and invasive capabilities of HepG2 and HuH-6 cells and accelerated the cell apoptosis; these influences were nullified by knockdown of DKK3. KDM1A inhibited DKK3 transcription by reducing H3 methylation. XAV-939 treatment inhibited the development of HepG2 and HuH-6 cells in which KDM1A and DKK3 were both knocked down. Knockdown of KDM1A reduced the tumor mass, inactivated the Wnt/ß-catenin signaling, and increased the expression of DKK3 in nude mice. KDM1A stimulates HB development by activating the Wnt/ß-catenin pathway through inhibition of DKK3 transcription.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Hepatoblastoma , Histona Demetilasas , Neoplasias Hepáticas , Vía de Señalización Wnt , Animales , Humanos , Ratones , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , beta Catenina/genética , beta Catenina/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Hepatoblastoma/genética , Hepatoblastoma/metabolismo , Hepatoblastoma/patología , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Neoplasias Hepáticas/patología , Ratones Desnudos , Vía de Señalización Wnt/genética
4.
Acta Neurobiol Exp (Wars) ; 82(3): 336-346, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36214716

RESUMEN

Hirschsprung's disease (HSCR) is an intestinal disease caused by defects in neural crest cell migration, proliferation, differentiation, and survival. Many reports have proposed that miRNA dysregulation is related to the occurrence of HSCR. However, the roles and mechanisms of miRNAs have not been thoroughly studied. The levels of miR­92a and KLF4 were examined using qRT­PCR and immunohistochemistry, respectively. Cell viability, migration and apoptosis were evaluated by MTT, Transwell and flow cytometry assays, respectively. A dual­luciferase reporter assay was employed to verify the binding relationship between miR­92a and KLF4. Levels of PI3K/AKT signals were further determined by western blot assay. Herein, elevated expression of miR­92a and reduced expression of KLF4 were found in HSCR tissues, and their expression patterns were negatively correlated. Overexpression of miR­92a inhibited cell viability and migration but enhanced cell apoptosis. However, overexpression of KLF4 had the opposite effects. Mechanistically, KLF4 was a target of miR­92a and it negatively affected biological functions by activating PI3K/AKT signaling. These results proved that miR­92a inhibited the proliferation and metastasis of nerve cells by regulating the KLF4/PI3K/AKT axis.


Asunto(s)
Enfermedad de Hirschsprung , MicroARNs , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Enfermedad de Hirschsprung/genética , Enfermedad de Hirschsprung/metabolismo , Enfermedad de Hirschsprung/patología , Humanos , MicroARNs/genética , Neuronas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
5.
Front Immunol ; 13: 891868, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35967364

RESUMEN

Liver ischemia-reperfusion injury (IRI) is a major complication of liver trauma, resection, and transplantation. IRI may lead to liver dysfunction and failure, but effective approach to address it is still lacking. To better understand the cellular and molecular mechanisms of liver IRI, functional roles of numerous cell types, including hepatocytes, Kupffer cells, neutrophils, and sinusoidal endothelial cells, have been intensively studied. In contrast, hepatic stellate cells (HSCs), which are well recognized by their essential functions in facilitating liver protection and repair, have gained less attention in their role in IRI. This review provides a comprehensive summary of the effects of HSCs on the injury stage of liver IRI and their associated molecular mechanisms. In addition, we discuss the regulation of liver repair and regeneration after IRI by HSCs. Finally, we highlight unanswered questions and future avenues of research regarding contributions of HSCs to IRI in the liver.


Asunto(s)
Células Estrelladas Hepáticas , Daño por Reperfusión , Células Endoteliales/metabolismo , Células Estrelladas Hepáticas/metabolismo , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Daño por Reperfusión/metabolismo
6.
Mol Med Rep ; 24(2)2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34080023

RESUMEN

Hepatoblastoma is the most common malignant hepatic tumour type with hypervascularity in early childhood. In recent decades, emerging evidence has proven that long non­coding RNAs (lncRNAs) serve an important oncogenic role in the pathogenesis of hepatoblastoma. However, the underlying mechanism of lncRNA taurine upregulated 1 (TUG1) in the angiogenesis of hepatoblastoma remains unknown. The expression patterns of TUG1 and microRNA (miR)­204­5p were detected in hepatoblastoma tissues and cell lines via reverse transcription­quantitative PCR and were analysed using a Pearson's correlation test. A tube formation assay was performed using human umbilical vein endothelial cells to assess the vasculogenic activity of treated HuH­6 cells. ELISA was used to detect the level of the secretory proangiogenic factor VEGFA in the culture media of HuH­6 cells. A dual luciferase reporter assay was performed to validate the binding relationships of TUG1/miR­204­5p and miR­204­5p/Janus kinase 2 (JAK2). Moreover, western blotting was conducted to measure the protein expression levels of VEGFA, phosphorylated (p)­JAK2, JAK2, p­STAT3 and STAT3. It was identified that TUG1 was upregulated, while miR­204­5p was downregulated in hepatoblastoma tissues and cells. TUG1 knockdown inhibited angiogenesis induced by hepatoblastoma cells. Furthermore, miR­204­5p was identified as a target of TUG1. The results demonstrated that TUG1 attenuated the inhibitory effect of miR­204­5p on the JAK2/STAT3 pathway and promoted angiogenesis in hepatoblastoma cells. In summary, TUG1 was upregulated in hepatoblastoma and suppressed miR­204­5p, thereby activating the downstream signalling pathway of JAK2/STAT3 to facilitate angiogenesis. The present findings will provide novel targets for the treatment of hepatoblastoma.


Asunto(s)
Hepatoblastoma/genética , Janus Quinasa 2/metabolismo , Neoplasias Hepáticas/genética , MicroARNs/metabolismo , Neovascularización Patológica/genética , ARN Largo no Codificante/genética , Factor de Transcripción STAT3/metabolismo , Línea Celular , Línea Celular Tumoral , Preescolar , Regulación hacia Abajo/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Hepatoblastoma/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Lactante , Neoplasias Hepáticas/metabolismo , Masculino , MicroARNs/genética , ARN Largo no Codificante/metabolismo , Transducción de Señal/genética , Regulación hacia Arriba/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
Saudi J Biol Sci ; 28(3): 1569-1575, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33732042

RESUMEN

BACKGROUND: Tacrolimus is a powerful immunosuppressant and has been widely used in organ transplantation. In order to further explore the role of tacrolimus in liver transplantation, we conducted network pharmacology analysis. METHODS: GSE100155 was obtained from the GEO database, and the DEGs of liver transplantation were analyzed. The 2D structure of tacrolimus was obtained from the National Library of Medicine, and the pharmacophore model of tacrolimus was predicted using the online tool pharmmapper. Then a network of tacrolimus and target genes was constructed through network pharmacology, and visualization and GO enrichment analysis was performed through Cytoscape. In addition, we also analyzed the correlation between key genes and immune infiltrating cells. The data of GSE84908 was used to verify the changes of key gene expression levels after tacrolimus treatment. RESULTS: The results of network pharmacological analysis showed that tacrolimus had 43 target genes, and the GO enrichment results showed many potential functions. Further analysis found that there were 5 key target genes in DEGs, and these 5 genes were significantly down-regulated in liver transplant patients. Another important finding was that 5 genes were significantly related to some immune infiltrating cells. The results of the GSE84908 data analysis showed that after tacrolimus treatment, the expression of DAAM1 was significantly increased (p = 0.015). CONCLUSION: Tacrolimus may inhibit the human immune response by affecting the expression of DAAM1 in liver transplant patients.

8.
Pathobiology ; 87(3): 161-170, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32182608

RESUMEN

OBJECTIVE: MiR-203 has been shown to participate in multiple malignancies, but the role of miR-203 in hepatoblastoma (HB) remains unclear. The aim of our study was to investigate the effects of miR-203 in HB. METHODS: A total of 15 pairs of HB tissues and para-tumour normal tissues were collected for the experiments. RT-qPCR and Western blotting were performed to detect the expression of CRNDE, miR-203, and VEGFA at the mRNA and/or protein levels, respectively. A dual luciferase assay verified the target relationship between miR-203 and the 3'UTR of VEGFA as well as miR-203 and CRNDE. In addition, MTT, wound healing, and tube formation assays were performed to assess the effects of miR-203, VEGFA, and CRNDE on cell proliferation, migration, and angiogenesis, respectively. RESULTS: Our data revealed that miR-203 expression was decreased in HB tissues, while long non-coding RNA (lncRNA) CRNDE expression was increased. The dysregulation of miR-203 and CRNDE was closely related to tumour size and stage. Moreover, overexpression of miR-203 inhibited angiogenesis. A dual luciferase assay verified that VEGFA is a direct target of miR-203 and that CRNDE binds to miR-203. Furthermore, our results showed that miR-203 suppressed cell viability, migration, and angiogenesis by regulating VEGFA expression. Additionally, it was confirmed that CRNDE promoted angiogenesis by negatively regulating miR-203 expression. CONCLUSION: lncRNA CRNDE targets the miR-203/VEGFA axis and promotes angiogenesis in HB. These results provide insight into the underlying mechanisms of HB and indicate that CRNDE and miR-203 might be potential targets for HB therapy.


Asunto(s)
Hepatoblastoma/genética , Neoplasias Hepáticas/genética , MicroARNs/genética , Neovascularización Patológica/genética , ARN Largo no Codificante/genética , Factor A de Crecimiento Endotelial Vascular/genética , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/metabolismo , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
IUBMB Life ; 72(2): 285-295, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31580537

RESUMEN

OBJECTIVE: This study is performed to explore the role of transcription factor FOXM1 in promoting the self-renewal and proliferation of liver cancer stem cells (LCSCs) by regulating the expression of acetaldehyde dehydrogenase-2 (ALDH2). METHODS: CD133+ CD24+ LCSCs were sorted and identified. A series of experiments were carried out to determine the proliferation, colony formation rate, migration, invasion, and apoptosis of LCSCs after interfering with FOXM1. Proliferation-, epithelial-mesenchymal transition (EMT)-, apoptosis-, and stemness-related factors were then detected by western blot analysis. Tumor xenograft in nude mice was used to figure out the role of FOXM1 in tumorigenesis in vivo by regulating ALDH2 expression. Luciferase activity assay was conducted to determine whether FOXM1 could target ALDH2 promoter region and thereby affecting ALDH2 expression. RESULTS: The sorted CD133+ CD24+ Huh-7 cells had the characteristic of stem cells. FOXM1 was highly expressed in CD133+ CD24+ Huh-7 cells. Silencing FOXM1 inhibited the proliferation and colony formation of LCSCs and decreased the expression of proliferating cell nuclear antigen and Ki-67 protein; inhibited the migration, invasion, and EMT of LCSCs while promoting the apoptosis of LCSCs, as well as promoted the expression of Bax and cleaved-caspase-3, and inhibited the expression of Bcl-2. Silencing FOXM1 inhibited the expression of Nanog, Oct4, and Sox2 in LCSCs by decreasing the expression of ALDH2. in vivo experiment, silencing FOXM1 suppressed tumorigenesis of LCSCs by decreasing the expression of ALDH2. CONCLUSION: Our study provides evidence that silencing FOXM1 inhibits stemness of LCSCs by decreasing the expression of ALDH2, and represses the proliferation, migration, invasion, and tumorigenesis while inducing the apoptosis of LCSCs.


Asunto(s)
Aldehído Deshidrogenasa Mitocondrial/metabolismo , Carcinoma Hepatocelular/patología , Movimiento Celular , Proliferación Celular , Proteína Forkhead Box M1/metabolismo , Regulación Neoplásica de la Expresión Génica , Células Madre Neoplásicas/patología , Aldehído Deshidrogenasa Mitocondrial/genética , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Transformación Celular Neoplásica , Transición Epitelial-Mesenquimal , Proteína Forkhead Box M1/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , Invasividad Neoplásica , Células Madre Neoplásicas/metabolismo , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA