Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
1.
Int J Mol Sci ; 22(23)2021 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-34884763

RESUMEN

The adipokine Neuregulin 4 (Nrg4) protects against obesity-induced insulin resistance. Here, we analyze how the downregulation of Nrg4 influences insulin action and the underlying mechanisms in adipocytes. Validated shRNA lentiviral vectors were used to generate scramble (Scr) and Nrg4 knockdown (KD) 3T3-L1 adipocytes. Adipogenesis was unaffected in Nrg4 KD adipocytes, but there was a complete impairment of the insulin-induced 2-deoxyglucose uptake, which was likely the result of reduced insulin receptor and Glut4 protein. Downregulation of Nrg4 enhanced the expression of proinflammatory cytokines. Anti-inflammatory agents recovered the insulin receptor, but not Glut4, content. Proteins enriched in Glut4 storage vesicles such as the insulin-responsive aminopeptidase (IRAP) and Syntaxin-6 as well as TBC1D4, a protein involved in the intracellular retention of Glut4 vesicles, also decreased by Nrg4 KD. Insulin failed to reduce autophagy in Nrg4 KD adipocytes, observed by a minor effect on mTOR phosphorylation, at the time that proteins involved in autophagy such as LC3-II, Rab11, and Clathrin were markedly upregulated. The lysosomal activity inhibitor bafilomycin A1 restored Glut4, IRAP, Syntaxin-6, and TBC1D4 content to those found in control adipocytes. Our study reveals that Nrg4 preserves the insulin responsiveness by preventing inflammation and, in turn, benefits the insulin regulation of autophagy.


Asunto(s)
Autofagia/fisiología , Transportador de Glucosa de Tipo 4/metabolismo , Resistencia a la Insulina/fisiología , Neurregulinas/metabolismo , Receptor de Insulina/biosíntesis , Células 3T3 , Adipocitos/metabolismo , Animales , Línea Celular , Cistinil Aminopeptidasa/biosíntesis , Citocinas/biosíntesis , Desoxiglucosa/metabolismo , Regulación hacia Abajo , Proteínas Activadoras de GTPasa/biosíntesis , Inflamación/patología , Insulina/metabolismo , Ratones , Neurregulinas/biosíntesis , Neurregulinas/genética , Proteínas Qa-SNARE/biosíntesis , Interferencia de ARN , ARN Interferente Pequeño/genética
2.
Biochem Pharmacol ; 192: 114736, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34411567

RESUMEN

Reprogramming of energy metabolism is a hallmarkofcancer, and the pentose phosphate pathway (PPP) is a major glucose metabolic pathway important for meeting the cellular demands of biosynthesis and anti-oxidant defense. Our previous study showed that phosphoinositide 3-kinase enhancer-activating Akt (PIKE-A) plays an important role in glioblastoma cell survival and growth under cellular energy stress condition. However, the crucial functions of PIKE-A in cancer energy metabolism are poorly understood.In the present study, we show that PIKE-A promotes DNA biosynthesis, NADPH production and inhibits reactive oxygen species (ROS) production, leading to increasing proliferation and growth of glioblastoma cell and suppressing cellular senescence. Mechanistically, PIKE-A binds to STAT3 and stimulates its phosphorylation mediated by tyrosine kinase Fyn, which enhances transcription of the rate-limitting enzyme glucose-6-phosphate dehydrogenase (G6PD) in the PPP. Finally, targeting PIKE-A-G6PD axis sensitizes glioblastoma to temozolomide (TMZ)treatment. This study reveals that STAT3 is a novel binding partner of PIKE-A which recruits Fyn to phosphorylate STAT3, contributing to the expression of G6PD, leading to promoting tumor growth and suppressing cellular senescence. Thus, the PIKE-A/STAT3/G6PD axis strongly links the PPP to carcinogenesis and may become a promising cancer therapeutic target.


Asunto(s)
Proliferación Celular/fisiología , Proteínas de Unión al GTP/biosíntesis , Proteínas Activadoras de GTPasa/biosíntesis , Glioblastoma/metabolismo , Glucosafosfato Deshidrogenasa/biosíntesis , Vía de Pentosa Fosfato/fisiología , Factor de Transcripción STAT3/biosíntesis , Animales , Línea Celular Tumoral , Regulación Enzimológica de la Expresión Génica , Técnicas de Inactivación de Genes/métodos , Glioblastoma/patología , Glucosafosfato Deshidrogenasa/genética , Células HEK293 , Humanos , Masculino , Ratones , Ratones Desnudos , Fosforilación/fisiología
3.
J Surg Res ; 267: 397-403, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34225052

RESUMEN

BACKGROUND: Diverticular disease is a common but poorly understood disease of the gastrointestinal tract. Recent studies have identified several single nucleotide polymorphisms (SNPs) that are associated with diverticular disease. MATERIALS AND METHODS: The genotypes of three SNPs (rs4662344 in ARHGAP15, rs7609897 in COLQ, and rs67153654 in FAM155A) were identified by Taqman assay in 204 patients with diverticular disease. Clinical characteristics were obtained from the medical record to study association with genotype. To evaluate gene expression in colon tissue, qPCR was performed on 24 patients with diverticulitis, and COLQ was localized using immunohistochemistry. RESULTS: The ARHGAP15 and COLQ SNPs were significantly associated with both diverticular disease and specifically diverticulitis, while the FAM155A was not associated with either. No association was found with clinical disease characteristics. Heterozygous genotypes at the ARHGAP15 SNP was associated with lower ARHGAP15 expression in colon tissues. COLQ protein localized to the myenteric plexus in the colon. CONCLUSIONS: This study confirmed association of the ARHGAP15 and COLQ SNPs with diverticular disease in our patients but could not confirm FAM155A SNP association. Neither of these SNPs appeared to associate with more severe disease, but genotype at the ARHGAP15 SNP did impact expression of ARHGAP15 in the colon. Additionally, this study is the first to localize COLQ in the colon. Its presence in the myenteric nervous system suggests COLQ SNP variants may contribute to diverticular disease by altering motility.


Asunto(s)
Acetilcolinesterasa , Enfermedades Diverticulares , Diverticulitis , Proteínas Activadoras de GTPasa , Proteínas Musculares , Acetilcolinesterasa/biosíntesis , Acetilcolinesterasa/genética , Colágeno , Colon/metabolismo , Colon/patología , Enfermedades Diverticulares/genética , Enfermedades Diverticulares/metabolismo , Enfermedades Diverticulares/patología , Diverticulitis/genética , Diverticulitis/metabolismo , Diverticulitis/patología , Proteínas Activadoras de GTPasa/biosíntesis , Proteínas Activadoras de GTPasa/genética , Humanos , Proteínas Musculares/biosíntesis , Proteínas Musculares/genética , Plexo Mientérico/metabolismo , Plexo Mientérico/patología , Polimorfismo de Nucleótido Simple
4.
Respir Res ; 22(1): 192, 2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-34210319

RESUMEN

BACKGROUND: To explore the role of family with sequence similarity 13 member A (FAM13A) in TGF-ß1-induced EMT in the small airway epithelium of patients with chronic obstructive pulmonary disease (COPD). METHODS: Small airway wall thickness and protein levels of airway remodeling markers, EMT markers, TGF-ß1, and FAM13A were measured in lung tissue samples from COPD and non-COPD patients. The correlations of FAM13A expression with COPD severity and EMT marker expression were evaluated. Gain- and loss-of-function assays were performed to explore the functions of FAM13A in cell proliferation, motility, and TGF-ß1-induced EMT marker alterations in human bronchial epithelial cell line BEAS-2B. RESULTS: Independent of smoking status, lung tissue samples from COPD patients exhibited significantly increased small airway thickness and collagen fiber deposition, along with enhanced protein levels of remodeling markers (collagen I, fibronectin, and MMP-9), mesenchymal markers (α-SMA, vimentin, and N-cadherin), TGF-ß1, and FAM13A, compared with those from non-COPD patients. FAM13A expression negatively correlated with FEV1% and PO2 in COPD patients. In small airway epithelium, FAM13A expression negatively correlated with E-cadherin protein levels and positively correlated with vimentin protein levels. In BEAS-2B cells, TGF-ß1 dose-dependently upregulated FAM13A protein levels. FAM13A overexpression significantly promoted cell proliferation and motility in BEAS-2B cells, whereas FAM13A silencing showed contrasting results. Furthermore, FAM13A knockdown partially reversed TGF-ß1-induced EMT marker protein alterations in BEAS-2B cells. CONCLUSIONS: FAM13A upregulation is associated with TGF-ß1-induced EMT in the small airway epithelium of COPD patients independent of smoking status, serving as a potential therapeutic target for anti-EMT therapy in COPD.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/fisiología , Transición Epitelial-Mesenquimal/fisiología , Proteínas Activadoras de GTPasa/biosíntesis , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Mucosa Respiratoria/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Adulto , Anciano , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Células Cultivadas , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/patología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/patología
5.
Zhongguo Fei Ai Za Zhi ; 24(7): 453-460, 2021 Jul 20.
Artículo en Chino | MEDLINE | ID: mdl-34134185

RESUMEN

BACKGROUND: Lung cancer is the leading cause of death worldwide, and lung adenocarcinoma is the main subtype of lung cancer. DEP domain-containing 1 (DEPDC1) has been proved to be closely related to the occurrence and development of most tumors, and the overexpression of DEPDC1 in lung adenocarcinoma has been preliminarily confirmed. This study aims to explore the relationship between the expression of DEPDC1 and the clinical prognosis of lung adenocarcinoma, and to preliminarily explore the possibility of DEPDC1 as a potential biomarker and therapeutic target of lung adenocarcinoma. METHODS: The bioinformatics website GEPIA database was used to collect relevant information, and the prognostic was analyzed online. Patient data were collected for statistical analysis, and immunohistochemical staining was performed on the collected samples. Subsequently, lung adenocarcinoma cells were cultured in vitro, and the knockout efficiency was verified by Western blot and reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and cell proliferation experiments were performed. RESULTS: The expression of DEPDC1 in lung adenocarcinoma tissues is significantly higher than that in adjacent normal tissues. The high expression of DEPDC1 is correlated with the tumor size and clinical stage of lung adenocarcinoma and knocking down DEPDC1 inhibits the proliferation of A549 and H1975 cells. CONCLUSIONS: DEPDC1 plays an important role in the progression and evolution of lung adenocarcinoma. And it is expected to become an important therapeutic target and a potential new biomarker for lung adenocarcinoma.


Asunto(s)
Adenocarcinoma del Pulmón , Proteínas Activadoras de GTPasa/genética , Neoplasias Pulmonares , Proteínas de Neoplasias/genética , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/metabolismo , Adenocarcinoma del Pulmón/patología , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Proliferación Celular/genética , Proliferación Celular/fisiología , Progresión de la Enfermedad , Femenino , Proteínas Activadoras de GTPasa/biosíntesis , Proteínas Activadoras de GTPasa/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/metabolismo , Pronóstico
6.
Int J Biol Sci ; 17(7): 1808-1820, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33994864

RESUMEN

Increasing evidence reveals that the Rho GTPase-activating protein is a crucial negative regulator of Rho family GTPase involved in tumorigenesis. The Rho GTPase-activating protein 25 (ARHGAP25) has been shown to specifically inactivate the Rho family GTPase Rac1, which plays an important role in pancreatic adenocarcinoma (PAAD) progression. Therefore, here we aimed to clarify the expression and functional role of ARHGAP25 in PAAD. The ARHGAP25 expression was lower in PAAD tissues than that in normal pancreatic tissues based on bioinformatics analysis and immunohistochemistry staining. Overexpression of ARHGAP25 inhibited cell growth of AsPC-1 human pancreatic cancer cells in vitro, while opposite results were observed in BxPC-3 human pancreatic cancer cells with ARHGAP25 knockdown. Consistently, in vivo tumorigenicity assays also confirmed that ARHGAP25 overexpression suppressed tumor growth. Mechanically, overexpression of ARHGAP25 inactivated AKT/mTOR signaling pathway by regulating Rac1/PAK1 signaling, which was in line with the results from the Gene set enrichment analysis on The Cancer Genome Atlas dataset. Furthermore, we found that ARHGAP25 reduced HIF-1α-mediated glycolysis in PAAD cells. Treatment with PF-04691502, a dual PI3K/mTOR inhibitor, hampered the increased cell growth and glycolysis due to ARHGAP25 knockdown in PAAD cells. Altogether, these results conclude that ARHGAP25 acts as a tumor suppressor by inhibiting the AKT/mTOR signaling pathway, which might provide a therapeutic target for PAAD.


Asunto(s)
Adenocarcinoma/genética , Carcinogénesis/genética , Proteínas Activadoras de GTPasa/genética , Regulación Neoplásica de la Expresión Génica , Proteína Oncogénica v-akt/genética , Neoplasias Pancreáticas/genética , Serina-Treonina Quinasas TOR/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Femenino , Proteínas Activadoras de GTPasa/biosíntesis , Glucólisis/genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Neoplasias Experimentales , Proteína Oncogénica v-akt/biosíntesis , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/biosíntesis
7.
PLoS One ; 16(5): e0251684, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33999959

RESUMEN

ARHGAP36 is an atypical Rho GTPase-activating protein (GAP) family member that drives both spinal cord development and tumorigenesis, acting in part through an N-terminal motif that suppresses protein kinase A and activates Gli transcription factors. ARHGAP36 also contains isoform-specific N-terminal sequences, a central GAP-like module, and a unique C-terminal domain, and the functions of these regions remain unknown. Here we have mapped the ARHGAP36 structure-activity landscape using a deep sequencing-based mutagenesis screen and truncation mutant analyses. Using this approach, we have discovered several residues in the GAP homology domain that are essential for Gli activation and a role for the C-terminal domain in counteracting an N-terminal autoinhibitory motif that is present in certain ARHGAP36 isoforms. In addition, each of these sites modulates ARHGAP36 recruitment to the plasma membrane or primary cilium. Through comparative proteomics, we also have identified proteins that preferentially interact with active ARHGAP36, and we demonstrate that one binding partner, prolyl oligopeptidase-like protein, is a novel ARHGAP36 antagonist. Our work reveals multiple modes of ARHGAP36 regulation and establishes an experimental framework that can be applied towards other signaling proteins.


Asunto(s)
Cilios , Proteínas Activadoras de GTPasa , Transducción de Señal , Animales , Cilios/química , Cilios/genética , Cilios/metabolismo , Proteínas Activadoras de GTPasa/biosíntesis , Proteínas Activadoras de GTPasa/química , Proteínas Activadoras de GTPasa/genética , Células HEK293 , Humanos , Ratones , Células 3T3 NIH , Dominios Proteicos , Isoformas de Proteínas , Relación Estructura-Actividad
8.
Life Sci ; 265: 118788, 2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33245966

RESUMEN

AIM: This study aimed to investigate the regulatory role of differentially-expressed circular RNAs (circRNAs) in mouse cardiomyocytes during doxorubicin (DOX)-induced cardiotoxicity. MAIN METHODS: Two groups of mice were injected with equal volumes (0.1 mL) of normal saline and DOX. Mouse heart tissue was isolated and digested for total RNA extraction and then subjected to next-generation RNA-sequencing. Expression profiles of circRNAs and circRNA-miRNA-mRNA networks were also constructed. Overall, 48 upregulated and 16 downregulated circRNAs were found to be statistically significant (p < 0.05) in the DOX-injected group. Bioinformatics analysis revealed several potential biological pathways that might be related to apoptosis caused by DOX-induced cardiotoxicity. In addition, using qRT-PCR, we found that a circRNA coded by the Arhgap12 gene, termed circArhgap12, was upregulated in the mouse heart tissue upon DOX intervention. CircArhgap12 enhanced apoptotic cell rate, as assessed using terminal-deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay, and increased reactive oxygen species and malondialdehyde release as well as superoxide dismutase and caspase-3 activation. Using a luciferase reporter assay, we found that circArhgap12 could sponge miR-135a-5p. In rat primary cardiomyocytes, we found that si-circArhgap12 promoted apoptosis and oxidative stress by sponging the miR-135a-5p inhibitor. Using bioinformatics analysis and luciferase reporter assay, we found that miR-135a-5p might have a potential target site for ADCY1 mRNA. KEY FINDINGS: Our research demonstrated that the expression profile of circRNAs was modified significantly and that circArhgap12 might play a competitive role among endogenous RNAs in mouse cardiomyocytes during DOX-induced cardiotoxicity. SIGNIFICANCE: Our study may provide a preliminary understanding of DOX-induced cardiotoxicity modulated by circRNA and its competing endogenous RNAs network.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Cardiotoxicidad/metabolismo , Doxorrubicina/toxicidad , Proteínas Activadoras de GTPasa/biosíntesis , MicroARNs/biosíntesis , ARN Circular/biosíntesis , Animales , Cardiotoxicidad/genética , Células Cultivadas , Proteínas Activadoras de GTPasa/genética , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , ARN Circular/genética , Distribución Aleatoria , Ratas
9.
J Cell Mol Med ; 24(18): 10648-10662, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32725802

RESUMEN

Enhancer of zeste homolog 2 (EZH2), an oncogene, is a commonly up-regulated epigenetic factor in human cancer. Hepatocellular carcinoma deletion gene 1 (DLC1) is an antioncogene that is either expressed at low levels or not expressed in many malignant tumours. Curcumin is a promising anticancer drug that has antitumour effects in many tumours, but its mechanism of action is unclear. Our research demonstrated that EZH2 was up-regulated in breast cancer (BC) tissues and cells, whereas DLC1 was down-regulated, and the expression of EZH2 and DLC1 was negatively correlated in BC. By analysing the characteristics of clinical cases, we found that positive expression of EZH2 and negative expression of DLC1 may be predictors of poor prognosis in patients with triple-negative breast cancer (TNBC). Moreover, knockdown of EZH2 expression restored the expression of DLC1 and inhibited the migration, invasion and proliferation, promoted the apoptosis, and blocked the cell cycle of MDA-MB-231 cells. Furthermore, we found that curcumin restored the expression of DLC1 by inhibiting EZH2; it also inhibited the migration, invasion and proliferation of MDA-MB-231 cells, promoted their apoptosis and blocked the cell cycle. Finally, xenograft tumour models were used to demonstrate that curcumin restored DLC1 expression by inhibiting EZH2 and also inhibited the growth and promoted the apoptosis of TNBC cells. In conclusion, our results suggest that curcumin can inhibit the migration, invasion and proliferation, promote the apoptosis, block the cycle of TNBC cells and restore the expression of DLC1 by inhibiting the expression of EZH2.


Asunto(s)
Adenocarcinoma/patología , Antineoplásicos Fitogénicos/farmacología , Curcumina/farmacología , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Proteínas Activadoras de GTPasa/biosíntesis , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias de la Mama Triple Negativas/patología , Proteínas Supresoras de Tumor/biosíntesis , Adulto , Anciano , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , División Celular/efectos de los fármacos , División Celular/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proteína Potenciadora del Homólogo Zeste 2/biosíntesis , Proteína Potenciadora del Homólogo Zeste 2/genética , Femenino , Proteínas Activadoras de GTPasa/genética , Técnicas de Silenciamiento del Gen , Código de Histonas , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Pronóstico , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Proteínas Supresoras de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Artículo en Inglés | MEDLINE | ID: mdl-31904427

RESUMEN

The sea cucumber is an economically important aquaculture species in China, where it encounter hypo-saline conditions caused by freshwater outflow from rivers and rainfall. MicroRNAs (miRNA) are small noncoding RNAs of about 22 nucleotides, which are crucial regulators of gene expression at the post-transcriptional level and are involved in a variety of physiological and pathophysiological processes. miR-10 is differentially expressed in salinity acclimation, and has a seed-region match with TBC1D5. The expression profiles of miR-10 and TBC1D5 indicate that miR-10 negatively regulates the expression of TBC1D5 in coelomocytes and sea cucumbers with a miR-10 agomir or antagomir. During salinity acclimation, up-regulation of miR-10 was induced after transfection in coelomocytes with a miR-10 inhibitor, while down-regulation of TBC1D5 was induced. The miR-10 expression maximum in coelomocytes appeared at 48 h post-transfection with a miR-10 inhibitor, was later than that of in sea cucumbers, which appeared 24 h after miR-10 antagomir injection. There was no longer a negative relationship between miR-10 and TBC1D5 expression in coelomocytes and sea cucumbers with miR-10 mimics or agomir during salinity acclimation. The miR-10 antagomir or agomir only affected sodium and NKA enzyme activities, and has little effect on other chloride and potassium ions. Our results demonstrate miR-10 directly regulates TBC1D5 by targeting its 3'-UTR, and that miR-10 suppression substantially increases TBC1D5 mRNA levels in vivo and in vitro. Furthermore, miR-10 and TBC1D5 fluctuating expression patterns after treatment with a miR-10 inhibitor or mimics during salinity acclimation may indicate that they are required for adaptation to salinity stress caused by environmental change. Especially, the miR-10 up-regulation in coelomocytes with miR-10 inhibitor during salinity acclimation indicated that they are required for adaptation to salinity stress caused by environmental change. We propose that miR-10 participates in a regulatory circuit that allows for rapid gene program transitions in response to osmotic stress.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Estrés Salino/fisiología , Stichopus/fisiología , Aclimatación/fisiología , Animales , Acuicultura , Células Cultivadas , Proteínas Activadoras de GTPasa/biosíntesis , Proteínas Activadoras de GTPasa/genética , MicroARNs/antagonistas & inhibidores , MicroARNs/biosíntesis , Stichopus/genética , Stichopus/metabolismo , Transfección , Regulación hacia Arriba
11.
Am J Physiol Heart Circ Physiol ; 318(2): H413-H424, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31886719

RESUMEN

We previously showed that ARHGAP42 is a smooth muscle cell (SMC)-selective, RhoA-specific GTPase activating protein that regulates blood pressure and that a minor allele single nucleotide variation within a DNAse hypersensitive regulatory element in intron1 (Int1DHS) increased ARHGAP42 expression by promoting serum response factor binding. The goal of the current study was to identify additional transcriptional and posttranscriptional mechanisms that control ARHGAP42 expression. Using deletion/mutation, gel shift, and chromatin immunoprecipitation experiments, we showed that recombination signal binding protein for immunoglobulin κ-J region (RBPJ) and TEA domain family member 1 (TEAD1) binding to a conserved core region was required for full IntDHS transcriptional activity. Importantly, overexpression of the notch intracellular domain (NICD) or plating SMCs on recombinant jagged-1 increased IntDHS activity and endogenous ARHGAP42 expression while siRNA-mediated knockdown of TEAD1 inhibited ARHGAP42 mRNA levels. Re-chromatin immunoprecipitation experiments indicated that RBPJ and TEAD1 were bound to the Int1DHS enhancer at the same time, and coimmunoprecipitation assays indicated that these factors interacted physically. Our results also suggest TEAD1 and RBPJ bound cooperatively to the Int1DHS and that the presence of TEAD1 promoted the recruitment of NICD by RBPJ. Finally, we showed that ARHGAP42 expression was inhibited by micro-RNA 505 (miR505) which interacted with the ARHGAP42 3'-untranslated region (UTR) to facilitate its degradation and by AK124326, a long noncoding RNA that overlaps with the ARHGAP42 transcription start site on the opposite DNA strand. Since siRNA-mediated depletion of AK124326 was associated with increased H3K9 acetylation and RNA Pol-II binding at the ARHGAP42 gene, it is likely that AK124326 inhibits ARHGAP42 transcription.NEW & NOTEWORTHY First, RBPJ and TEAD1 converge at an intronic enhancer to regulate ARHGAP42 expression in SMCs. Second, TEAD1 and RBPJ interact physically and bind cooperatively to the ARHGAP42 enhancer. Third, miR505 interacts with the ARHGAP42 3'-UTR to facilitate its degradation. Finally, LncRNA, AK124326, inhibits ARHGAP42 transcription.


Asunto(s)
Presión Sanguínea/genética , Presión Sanguínea/fisiología , Proteínas Activadoras de GTPasa/genética , Regulación de la Expresión Génica/genética , Miocitos del Músculo Liso/metabolismo , Procesamiento Proteico-Postraduccional/genética , Regiones no Traducidas 3'/genética , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas Activadoras de GTPasa/biosíntesis , Eliminación de Gen , Técnicas de Silenciamiento del Gen , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , MicroARNs/genética , Mutación , Proteínas Nucleares/genética , ARN Largo no Codificante/genética , ARN Interferente Pequeño , Factores de Transcripción de Dominio TEA , Factores de Transcripción/genética
12.
Cell Prolif ; 53(1): e12717, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31825138

RESUMEN

OBJECTIVES: DISHEVELLED, EGL-10, PLECKSTRIN (DEP) domain-containing 1B (DEPDC1B) promotes dismantling of focal adhesions and coordinates detachment events during cell cycle progression. DEPDC1B is overexpressed in several cancers with expression inversely correlated with patient survival. Here, we analysed the role of DEPDC1B in the regulation of murine and human skeletal myogenesis. MATERIALS AND METHODS: Expression dynamics of DEPDC1B were examined in murine and human myoblasts and rhabdomyosarcoma cells in vitro by RT-qPCR and/or immunolabelling. DEPDC1B function was mainly tested via siRNA-mediated gene knockdown. RESULTS: DEPDC1B was expressed in proliferating murine and human myoblasts, with expression then decreasing markedly during myogenic differentiation. SiRNA-mediated knockdown of DEPDC1B reduced myoblast proliferation and induced entry into myogenic differentiation, with deregulation of key cell cycle regulators (cyclins, CDK, CDKi). DEPDC1B and ß-catenin co-knockdown was unable to rescue proliferation in myoblasts, suggesting that DEPDC1B functions independently of canonical WNT signalling during myogenesis. DEPDC1B can also suppress RHOA activity in some cell types, but DEPDC1B and RHOA co-knockdown actually had an additive effect by both further reducing proliferation and enhancing myogenic differentiation. DEPDC1B was expressed in human Rh30 rhabdomyosarcoma cells, where DEPDC1B or RHOA knockdown promoted myogenic differentiation, but without influencing proliferation. CONCLUSION: DEPDC1B plays a central role in myoblasts by driving proliferation and preventing precocious myogenic differentiation during skeletal myogenesis in both mouse and human.


Asunto(s)
Proliferación Celular , Proteínas Activadoras de GTPasa/biosíntesis , Regulación Neoplásica de la Expresión Génica , Mioblastos Esqueléticos/metabolismo , Proteínas de Neoplasias/metabolismo , Rabdomiosarcoma/metabolismo , Animales , Línea Celular Tumoral , Humanos , Ratones , Mioblastos Esqueléticos/patología , Rabdomiosarcoma/patología
13.
Int J Biol Markers ; 34(4): 348-355, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31544570

RESUMEN

BACKGROUND: The oncogene IQ motif-containing GTPase activating protein 3 (IQGAP3) is ubiquitously overexpressed in several human cancers. This study was designed to explore the expression and role of IQGAP3 in colorectal cancer. METHODS: We first assessed the IQGAP3 expression level in colorectal cancer. The correlation of IQGAP3 expression with the clinicopathological characteristics and prognosis was then assessed. At last, we investigated the function of IQGAP3 in colorectal cancer by knocking down its expression in colorectal cancer cell lines. RESULTS: Consistent with the conclusions drawn from The Cancer Genome Atlas database, IQGAP3 was upregulated in colorectal cancer at the tissue level and cellular level. Based on immunohistochemistry results of the tissue microarrays, we demonstrated that higher expression of IQGAP3 was associated with higher tumor node metastasis stage (P = 0.005), higher incidence of lymph node metastasis (P = 0.004), and shorter overall survival (P = 0.022). Knockdown of IQGAP3 in colorectal cancer cell lines remarkably decreased their proliferation and migration abilities. CONCLUSION: Our data provide evidence that IQGAP3 significantly promote malignant progression of colorectal cancer and could serve as a potential therapeutic target.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Proteínas Activadoras de GTPasa/biosíntesis , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Femenino , Proteínas Activadoras de GTPasa/genética , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Persona de Mediana Edad , Pronóstico
14.
J Am Heart Assoc ; 8(2): e010057, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30630384

RESUMEN

Background Vascular endothelial cell (EC) alignment in the direction of flow is an adaptive response that protects against aortic diseases, such as atherosclerosis. The Rho GTP ases are known to regulate this alignment. Herein, we analyze the effect of ARHGAP 18 on the regulation of EC alignment and examine the effect of ARHGAP 18 deficiency on the development of atherosclerosis in mice. Methods and Results We used in vitro analysis of ECs under flow conditions together with apolipoprotein E-/- Arhgap 18-/- double-mutant mice to study the function of ARHGAP 18 in a high-fat diet-induced model of atherosclerosis. Depletion of ARHGAP 18 inhibited the alignment of ECs in the direction of flow and promoted inflammatory phenotype, as evidenced by disrupted junctions and increased expression of nuclear factor-κB and intercellular adhesion molecule-1 and decreased endothelial nitric oxide synthase. Mice with double deletion in ARHGAP 18 and apolipoprotein E and fed a high-fat diet show early onset of atherosclerosis, with lesions developing in atheroprotective regions. Conclusions ARHGAP 18 is a protective gene that maintains EC alignments in the direction of flow. Deletion of ARHGAP 18 led to loss of EC ability to align and promoted atherosclerosis development.


Asunto(s)
Enfermedades de la Aorta/genética , Velocidad del Flujo Sanguíneo/fisiología , Endotelio Vascular/metabolismo , Proteínas Activadoras de GTPasa/genética , Regulación de la Expresión Génica , Placa Aterosclerótica/genética , Animales , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Western Blotting , Modelos Animales de Enfermedad , Endotelio Vascular/patología , Proteínas Activadoras de GTPasa/biosíntesis , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , ARN/genética , Transducción de Señal
15.
Cancer Res ; 79(5): 905-917, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30674530

RESUMEN

Medulloblastoma and central nervous system primitive neuroectodermal tumors (CNS-PNET) are aggressive, poorly differentiated brain tumors with limited effective therapies. Using Sleeping Beauty (SB) transposon mutagenesis, we identified novel genetic drivers of medulloblastoma and CNS-PNET. Cross-species gene expression analyses classified SB-driven tumors into distinct medulloblastoma and CNS-PNET subgroups, indicating they resemble human Sonic hedgehog and group 3 and 4 medulloblastoma and CNS neuroblastoma with FOXR2 activation. This represents the first genetically induced mouse model of CNS-PNET and a rare model of group 3 and 4 medulloblastoma. We identified several putative proto-oncogenes including Arhgap36, Megf10, and Foxr2. Genetic manipulation of these genes demonstrated a robust impact on tumorigenesis in vitro and in vivo. We also determined that FOXR2 interacts with N-MYC, increases C-MYC protein stability, and activates FAK/SRC signaling. Altogether, our study identified several promising therapeutic targets in medulloblastoma and CNS-PNET. SIGNIFICANCE: A transposon-induced mouse model identifies several novel genetic drivers and potential therapeutic targets in medulloblastoma and CNS-PNET.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Cerebelosas/genética , Meduloblastoma/genética , Tumores Neuroectodérmicos Primitivos/genética , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Transformación Celular Neoplásica/genética , Neoplasias Cerebelosas/metabolismo , Neoplasias Cerebelosas/patología , Elementos Transponibles de ADN/genética , Femenino , Factores de Transcripción Forkhead/genética , Proteínas Activadoras de GTPasa/biosíntesis , Proteínas Activadoras de GTPasa/genética , Humanos , Masculino , Meduloblastoma/metabolismo , Meduloblastoma/patología , Proteínas de la Membrana/genética , Ratones , Ratones Desnudos , Mutagénesis Insercional/métodos , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Tumores Neuroectodérmicos Primitivos/metabolismo , Tumores Neuroectodérmicos Primitivos/patología , Pronóstico
16.
Sci Rep ; 9(1): 390, 2019 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-30674964

RESUMEN

AGAP2 (Arf GAP with GTP-binding protein-like domain, Ankyrin repeat and PH domain 2) isoform 2 is considered a proto-oncogene, but not much is known about AGAP2 gene expression regulation. To get some insight into this process, AGAP2 proximal promoter was cloned and characterised using reporter assays. We have identified SP1 as a transcription factor bound to AGAP2 promoter and required for AGAP2 expression in two different types of cancer cells (KU812, a chronic myeloid leukaemia cell line; and DU145, a prostate cancer cell line): silencing SP1 decreased AGAP2 protein levels. We have also found that all-trans retinoic acid (ATRA) treatment increased AGAP2 protein levels in both cell lines whilst curcumin treatment reduced ATRA-mediated AGAP2 increase. Furthermore, chromatin immunoprecipitation studies revealed the presence of RARα, RXRα and the lysine acetyl transferase PCAF in AGAP2 promoter. Our results provide a novel understanding of AGAP2 expression regulation that could be beneficial to those patients with cancers where AGAP2 is overexpressed.


Asunto(s)
Proteínas de Unión al GTP/biosíntesis , Proteínas Activadoras de GTPasa/biosíntesis , Regulación Enzimológica de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias de la Próstata/metabolismo , Receptor alfa de Ácido Retinoico/metabolismo , Factor de Transcripción Sp1/metabolismo , Línea Celular Tumoral , Proteínas de Unión al GTP/genética , Proteínas Activadoras de GTPasa/genética , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Masculino , Proteínas de Neoplasias/genética , Neoplasias de la Próstata/genética , Proto-Oncogenes Mas , Receptor alfa de Ácido Retinoico/genética , Factor de Transcripción Sp1/genética
17.
Int J Cancer ; 144(12): 3043-3055, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-30511377

RESUMEN

Considering the importance of microRNAs (miRNAs) in regulating cellular processes, we performed microarray analysis and revealed miR-4324 as one of the most differentially expressed miRNAs in bladder cancer (BCa). Then, we discovered that miR-4324 was a negative regulator of Rac GTPase activating protein 1 (RACGAP1) and that RACGAP1 functioned as an oncogenic protein in BCa. Our studies indicated that ectopic overexpression of miR-4324 in BCa cells significantly suppressed cell proliferation and metastasis and enhanced chemotherapy sensitivity to doxorubicin by repressing RACGAP1 expression. Further studies showed that estrogen receptor 1 (ESR1) increased the expression of miR-4324 by binding to its promoter, while the downregulation of ESR1 in BCa was caused by hypermethylation of its promoter. p-STAT3 induced the enrichment of DNMT3B by binding to the ESR1 promoter and then induced methylation of the ESR1 promoter. In turn, RACGAP1 induced STAT3 phosphorylation, increasing p-STAT3 expression and promoting its translocation to the nucleus. Therefore, the miR-4324-RACGAP1-STAT3-ESR1 feedback loop could be a critical regulator of BCa progression.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , MicroARNs/metabolismo , Factor de Transcripción STAT3/metabolismo , Neoplasias de la Vejiga Urinaria/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/fisiología , Metilación de ADN , Progresión de la Enfermedad , Regulación hacia Abajo , Doxorrubicina/farmacología , Receptor alfa de Estrógeno/genética , Retroalimentación Fisiológica , Proteínas Activadoras de GTPasa/biosíntesis , Proteínas Activadoras de GTPasa/genética , Xenoinjertos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Metástasis de la Neoplasia , Regiones Promotoras Genéticas , Factor de Transcripción STAT3/genética , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/patología
18.
Med Sci Monit ; 24: 8669-8677, 2018 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-30499465

RESUMEN

BACKGROUND STAT3 has emerged as a novel potential target for sorafenib, a multikinase inhibitor, in the context of cancer therapy. ARHGAP24 is a Rac-specific Rho GTPase-activating protein (Rho GAP), which can convert Rho GTPases to an inactive state. It has been proved to be an oncosuppressor protein in renal cancer. In the present study, we investigated its anti-cancer effect in breast cancer (BC). MATERIAL AND METHODS Quantitative real-time PCR (qRT-PCR) and Western blot analysis were performed to detect the expression of ARHGAP24 in clinical tissue samples. Then, BC MDA-MB-231 cells were virally transduced with ARHGAP24 silencing or overexpression lentiviral vectors in the absence or presence of sorafenib. Cell viability and metastatic ability were evaluated by using the Cell Counting Kit-8 (CCK-8) and Transwell assays. Proteins belonging to the STAT3 pathway were detected by Western blot. RESULTS ARHGAP24 decreased in BC tissues compared with the adjacent normal tissues. Forced expression of ARHGAP24 and sorafenib treatment significantly suppressed the viability, migration, and invasion of MDA-MB-231 cells. Conversely, elimination of the endogenous ARHGAP24 with shRNA promoted cell viability, migration, and invasion. The phosphorylation of STAT3 and the expression of MMP-2 and MMP-9 were attenuated by ARHGAP24 ectopic expression and sorafenib treatment. Furthermore, forced expression of ARHGAP24 significantly enhanced sorafenib-induced decrease of cell viability, migration, and invasion of MDA-MB-231 cells, while elimination of the endogenous ARHGAP24 with shRNA inhibited it. CONCLUSIONS ARHGAP24 can suppress the development of MDA-MB-231 cells via the STAT3 signaling pathway, and sorafenib inhibits cell viability, migration, invasion, and STAT3 activation in MDA-MB-231 cells through ARHGAP24.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Factor de Transcripción STAT3/metabolismo , Sorafenib/farmacología , Anciano , Anciano de 80 o más Años , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Proteínas Activadoras de GTPasa/biosíntesis , Proteínas Activadoras de GTPasa/genética , Humanos , MicroARNs/genética , Persona de Mediana Edad , Invasividad Neoplásica , Transducción de Señal/efectos de los fármacos
19.
Pharmacol Rep ; 70(5): 963-971, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30107346

RESUMEN

BACKGROUND: The most frequent type of renal cell carcinoma is called clear-cell renal cell carcinoma (ccRCC) which is associated with a poor prognosis. It has been observed that miR-137 is aberrantly expressed in many different kinds of human malignancies including ccRCC. This research aims to examine the role of miR-137 in ccRCC. METHODS: Quantitative RT-PCR (qRT-PCR) was applied to measure miR-137 expression in ccRCC and adjacent noncancerous tissue. Gene expression was determined by western blot. Cell Counting Kit-8 (CCK-8) assay, flow cytometry and Transwell assay were used to determine the effects of miR-137 on cell growth, apoptosis and invasion, respectively. Moreover, xenograft and pulmonary metastasis animal models were established to investigate the role of miR-137 in vivo. RESULTS: Our findings show that there was significant downregulation of miR-137 in ccRCC tissue relative to corresponding non-cancerous tissue. Ectopic miR-137 expression in ccRCC cells led to suppression of cell growth and invasion, as well as apoptosis induction. In contrast, knockdown of miR-137 enhances proliferation and invasion, inhibits apoptosis. It also confirms that miR-137 plays a tumor supressor role in vivo. Mechanically, miR-137 directly targets the 3'-UTR of RLIP76 which is an established oncogene in ccRCC. CONCLUSION: MiR-137 serves as a tumor suppressor, which can be considered a potential therapeutic target in ccRCC.


Asunto(s)
Neoplasias Pulmonares/prevención & control , MicroARNs/farmacología , Transportadoras de Casetes de Unión a ATP/biosíntesis , Anciano , Apoptosis/genética , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Proliferación Celular/genética , Femenino , Proteínas Activadoras de GTPasa/biosíntesis , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares/secundario , Masculino , MicroARNs/biosíntesis , Persona de Mediana Edad , Invasividad Neoplásica/genética , ARN Interferente Pequeño/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Histol Histopathol ; 33(6): 597-608, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29297565

RESUMEN

BACKGROUND: The outcome of patients with metastatic soft tissue sarcoma (STS) remains unfavourable and new therapeutic strategies are needed. The aim of this study was to determine the role of RhoGAP, XTP1/DEPDC1B and SDP35/DEPDC1A, as possible prognostic markers, to be used to identify candidate patients for more effective and personalized therapies. MATERIALS-METHODS: SDP35/DEPDC1A and XTP1/DEPDC1B transcriptional levels were evaluated by Real-Time PCR in 86 primary STS and 22 paired lung metastasis. 17 normal tissues were used as control. Protein expression was evaluated by tissue microarray, including 152 paraffin-embedded STS samples and by western blot in 22 lung metastases and paired primary STS. Non-parametric and parametric analysis were used to establish the differences in gene and protein expression and prognostic factors were tested with Kaplan Meier and Cox's regression analyses. RESULTS: SDP35/DEPDC1A and XTP1/DEPDC1B gene were down-regulated in adjacent normal tissues while sarcoma specimens presented high mRNA levels, significantly related to metastasis-free survival. Gene expression further increased in paired metastatic lesions. Immunohistochemical staining showed a variable expression in intensity and distribution, with a significantly higher probability of metastatic disease in patients up-regulating SDP35/DEPDC1A. Western blotting assessed high levels of proteins in STS specimens and indicated a stronger expression of SDP35/DEPDC1A in metastases when compared to primary tumours. Multivariate analyses highlighted that SDP35/DEPDC1A abundance, grade III and no history of radiation therapy were significant independent risk factors. CONCLUSIONS: Our results demonstrated that increased expression of SDP35/DEPDC1A and XPT1/DEPDC1B correlates with metastatic progression and identified SDP35/DEPDC1A as an independent marker for prediction of poor prognosis.


Asunto(s)
Biomarcadores de Tumor/análisis , Proteínas Activadoras de GTPasa/biosíntesis , Proteínas de Neoplasias/biosíntesis , Sarcoma/patología , Neoplasias de los Tejidos Blandos/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Supervivencia sin Enfermedad , Femenino , Proteínas Activadoras de GTPasa/análisis , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/análisis , Pronóstico , Modelos de Riesgos Proporcionales , Sarcoma/mortalidad , Neoplasias de los Tejidos Blandos/mortalidad , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...