Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 312
Filtrar
1.
J Cell Physiol ; 235(11): 8176-8186, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-31960969

RESUMEN

Circular RNA (circRNA) is a subclass of noncoding RNA (ncRNA) detected within mammalian tissues and cells. However, its regulatory role during the proliferation phase of rat liver regeneration (LR) remains unreported. This study was designed to explore their regulatory mechanisms in cell proliferation of LR. The circRNA expression profile was detected by high-throughput sequencing. It was indicated that 260 circRNAs were differentially expressed during the proliferation phase of rat LR. Among them, circ-14723 displayed a significantly differential expression. We further explored its regulatory mechanism in rat hepatocytes (BRL-3A cells). First, EdU, flow cytometry and western blot (WB) indicated that knocking down circ-14723 inhibited BRL-3A cells proliferation. Second, RNA-Pulldown and dual-luciferase report assay showed that circ-14723 could sponge rno-miR-16-5p. At last, WB showed that the reported target genes of rno-miR-16-5p, CCND1, and CCNE1 were downregulated after knocking down circ-14723. In conclusion, we found that circ-14723 exerted a critical role in G1/S arrest to promote cell proliferation via rno-miR-16-5p/CCND1 and CCNE1 axis in rat LR. This finding further revealed the regulatory mechanisms of circRNA on cell proliferation of LR, and might provide a potential target for clinical problems.


Asunto(s)
Proliferación Celular/genética , Regulación de la Expresión Génica/genética , Hepatocitos/metabolismo , Regeneración Hepática/genética , MicroARNs/genética , ARN Circular/genética , Animales , Ciclina D1/biosíntesis , Ciclina D1/genética , Ciclina E/biosíntesis , Ciclina E/genética , Masculino , Ratas , Ratas Sprague-Dawley
2.
Int Ophthalmol ; 40(2): 343-350, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31571090

RESUMEN

PURPOSE: To investigate the overexpression of genes in sebaceous gland carcinoma (SGC) of the eyelid compared to sebaceous adenoma of the eyelid in order to elucidate the molecular mechanism underlying pathogenesis. METHODS: We performed histopathological examination of eyelid tissues surgically removed from four patients diagnosed with SGC (cases 1-3) and sebaceous adenoma (case 4) of the eyelid. Next, we performed global gene expression analysis of surgical tissue samples using a GeneChip® system and the Ingenuity Pathways Knowledge Base. The results of the GeneChip® analysis were explored with quantitative real-time polymerase chain reaction (qRT-PCR) analysis. RESULTS: In the SGC samples, we found that 211, 199, and 199 genes, respectively, showed ≥ 2.0-fold higher expression than those in the sebaceous adenoma sample (case 4); 194 genes were common to all three SGC samples. For the 194 genes with upregulated expression, functional category analysis showed that SGC of the eyelid employed a unique gene network, including cyclin-dependent kinase inhibitor 2A (CDKN2A), cyclin-dependent kinase 1 (CDK1), and cyclin E1 (CCNE1), which are related to cell cycle progression, incidence of tumor, and cell viability. Furthermore, qRT-PCR analysis showed that the expression levels of CDKN2A, CDK1, and CCNE1 were significantly upregulated in all SGC cases compared to those in the sebaceous adenoma case. These data were similar to the results of microarray analysis. CONCLUSION: Overexpression of cell cycle-related genes CDKN2A, CDK1, CCNE1, and their gene network may help elucidate the pathogenic pathway of SGC of the eyelid at the molecular level.


Asunto(s)
Adenocarcinoma Sebáceo/genética , Proteína Quinasa CDC2/genética , Ciclina E/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Neoplasias de los Párpados/genética , Regulación Neoplásica de la Expresión Génica , Proteínas Oncogénicas/genética , Neoplasias de las Glándulas Sebáceas/genética , Adenocarcinoma Sebáceo/metabolismo , Adenocarcinoma Sebáceo/patología , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Proteína Quinasa CDC2/biosíntesis , Ciclina E/biosíntesis , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Neoplasias de los Párpados/metabolismo , Neoplasias de los Párpados/patología , Femenino , Humanos , Inmunohistoquímica , Masculino , Proteínas Oncogénicas/biosíntesis , ARN Neoplásico/genética , Neoplasias de las Glándulas Sebáceas/metabolismo , Neoplasias de las Glándulas Sebáceas/patología , Glándulas Sebáceas/metabolismo , Glándulas Sebáceas/patología
3.
Cancer Control ; 26(1): 1073274819875736, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31533461

RESUMEN

Cyclin E and hepatocyte growth factor (HGF) have been observed as a multifaceted factor in many cancers, and the assessment of microvascular density (MVD) and micro-lymphatic vessel density (MLVD) has been used to quantify tumor angiogenesis and lymphangiogenesis. The aim of this study was to explore the association between expression of cyclin E, HGF, MVD, and MLVD, and clinicopathologic parameters in esophageal squamous cell carcinoma (ESCC). The expression of cyclin E, HGF, MVD, and MLVD were detected using immunohistochemically anticyclin E, HGF, CD34, and lymphatic vessel endothelial hyaluronan receptor 1 in 168 surgically resected ESCC cases and 30 normal esophageal mucosal samples. The expression levels of cyclin E, HGF, MVD, and MLVD were higher compared to controls. High cyclin E and HGF expression was found more frequently in the tumors larger than 5 cm (P < .001), with poorer differentiation (P = .034) and higher tumor node metastasis (TNM) staging (P = .009) compared to their counterparts. Both MVD and MLVD values were found to be higher in the tumors larger than 5 cm (P < .001), with poorer differentiation (P < .001) and higher TNM staging (P < .001) compared to their counterparts. Furthermore, the expression of MVD and MLVD in both the high cyclin E and high HGF expression groups was significantly higher compared to the low cyclin E and HGF expression groups (P < .001). This study demonstrated that high cyclin E and HGF expression is closely correlated with tumor size, tumor differentiation degree, and TNM stage in patients with ESCC. These findings proposed that cyclin E and HGF could serve as novel molecular markers for preoperational evaluation of ESCC.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Carcinoma de Células Escamosas/metabolismo , Ciclina E/biosíntesis , Neoplasias Esofágicas/metabolismo , Factor de Crecimiento de Hepatocito/biosíntesis , Vasos Linfáticos/metabolismo , Neovascularización Patológica/metabolismo , Anciano , Carcinoma de Células Escamosas/irrigación sanguínea , Carcinoma de Células Escamosas/patología , Neoplasias Esofágicas/irrigación sanguínea , Neoplasias Esofágicas/patología , Femenino , Humanos , Inmunohistoquímica , Vasos Linfáticos/patología , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neovascularización Patológica/patología , Pronóstico
4.
J Exp Clin Cancer Res ; 38(1): 376, 2019 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-31455378

RESUMEN

BACKGROUND: Metformin has been reported to function as the anti-tumor inhibiting the growth of different types of cancers, including bladder cancer. But there are few reports on the roles of Yap1, the key molecule of Hippo pathway, in the metformin induced inhibition of bladder cancer (BLCA). We are wondering if the inhibitory effect of metformin on bladder cancer is fulfilled via Yap1 and exploring the related mechanism. METHODS: MTS and colony formation assays were used to explore the cellular viabilities and proliferation of BLCA cells challenged by metformin at different concentrations, in vitro. Flow Cytometry (FCM) was used to analyze the cell cycle and the cellular apoptosis of the BLCA cells. Western Blot was performed to detect the expressions of AMPKα, Yap1, CCND1, CCNE1/2 and CDK2/4/6 in the metformin-treated BLCA cell lines. RNAi method was used for the related genetic functional analysis. The relationships among Yap1, TEADs and CCNE1/2 were predicted and evaluated using bioinformatics, dual-luciferase reporter and co-immunoprecipitation (Co-IP) assays. For in vivo experiments, a xenograft model was used to investigate the effects of metformin on the proliferation of BLCA cells. And Immunohistochemistry (IHC) assay was performed to assess the expressions of CCNE1/2 and Yap1 proteins in the tumor tissues from the model. RESULTS: Metformin could inhibit the proliferation of the BLCA cells via inducing the G1 cell cycle arrest without apoptosis. And metformin upregulated the phosphorylated AMPKα and decreased the expressions of Yap1 and CCND1, CCNE1/2 and CDK4/6. AMPK inhibition by compound C (CC) restored the cell proliferation and the G1 cell cycle arrest induced by metformin, in vivo. Knockdown of YAP1 inhibited the proliferation of BLCA cells and caused the cell cycle arrest at G1 phase by decreasing the expressions of CCNE1/2 and other G1 phase related molecules, which has been restored by the Yap 5SA mutant. Bioinformatics analysis showed that trans-factor TEAD4 was highly expressed and positively associated with the expressions of CCNE1 and CCNE2 in BLCA and only TEAD4 was precipitated by Yap1 in the BLCA cells. Further studies demonstrated that Yap1 positively regulated both CCNE1 and CCNE2 expressions via forming complex with TEAD4. Furthermore, we observed that metformin inhibited the cell proliferation by decreasing the expressions of Yap1 and both CCNE1 and CCNE2 in xenograft model. CONCLUSIONS: The results of our study reveal a new potential regulatory pathway in which metformin inhibits cell proliferation via AMPKα/Yap1/TEAD4/CCNE1/2 axis in BLCA cells, providing new insights into novel molecular therapeutic targets for BLCA.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Ciclina E/antagonistas & inhibidores , Ciclinas/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Metformina/farmacología , Proteínas Musculares/metabolismo , Proteínas Oncogénicas/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina E/biosíntesis , Ciclina E/genética , Ciclina E/metabolismo , Ciclinas/biosíntesis , Ciclinas/genética , Ciclinas/metabolismo , Proteínas de Unión al ADN/genética , Femenino , Fase G1/efectos de los fármacos , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Musculares/genética , Proteínas Oncogénicas/biosíntesis , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Factores de Transcripción de Dominio TEA , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Transfección , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Proteínas Señalizadoras YAP
5.
Int J Cancer ; 145(5): 1299-1311, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31093963

RESUMEN

Molecular subtypes of breast cancer are defined on the basis of gene expression and genomic/epigenetic pattern differences. Different subtypes are thought to originate from distinct cell lineages, but the early activation of an oncogene could also play a role. It is difficult to discriminate the respective inputs of oncogene activation or cell type of origin. In this work, we wished to determine whether activation of distinct oncogenic pathways in human mammary epithelial cells (HMEC) could lead to different patterns of genetic and epigenetic changes. To this aim, we transduced shp53 immortalized HMECs in parallel with the CCNE1, WNT1 and RASv12 oncogenes which activate distinct oncogenic pathways and characterized them at sequential stages of transformation for changes in their genetic and epigenetic profiles. We show that initial activation of CCNE1, WNT1 and RASv12, in shp53 HMECs results in different and reproducible changes in mRNA and micro-RNA expression, copy number alterations (CNA) and DNA methylation profiles. Noticeably, HMECs transformed by RAS bore very specific profiles of CNAs and DNA methylation, clearly distinct from those shown by CCNE1 and WNT1 transformed HMECs. Genes impacted by CNAs and CpG methylation in the RAS and the CCNE1/WNT1 clusters showed clear differences, illustrating the activation of distinct pathways. Our data show that early activation of distinct oncogenic pathways leads to active adaptive events resulting in specific sets of CNAs and DNA methylation changes. We, thus, propose that activation of different oncogenes could have a role in reshaping the genetic landscape of breast cancer subtypes.


Asunto(s)
Neoplasias de la Mama/genética , Glándulas Mamarias Humanas/fisiología , Oncogenes , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Ciclina E/biosíntesis , Ciclina E/genética , Metilación de ADN , Epigénesis Genética , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células Epiteliales/fisiología , Femenino , Dosificación de Gen , Regulación Neoplásica de la Expresión Génica , Genoma Humano , Xenoinjertos , Humanos , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Ratones , Ratones Desnudos , Ratones SCID , Proteínas Oncogénicas/biosíntesis , Proteínas Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras)/biosíntesis , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteína Wnt1/biosíntesis , Proteína Wnt1/genética
6.
mBio ; 10(2)2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-31040242

RESUMEN

Toxoplasma gondii is an obligate intracellular parasite that establishes a favorable environment in the host cells in which it replicates. We have previously reported that it uses MYR-dependent translocation of dense granule proteins to elicit a key set of host responses related to the cell cycle, specifically, E2F transcription factor targets, including cyclin E. We report here the identification of a novel Toxoplasma effector protein that is exported from the parasitophorous vacuole in a MYR1-dependent manner and localizes to the host's nucleus. Parasites lacking this inducer of host cyclin E (HCE1) are unable to modulate E2F transcription factor target genes and exhibit a substantial growth defect. Immunoprecipitation of HCE1 from infected host cells showed that HCE1 efficiently binds elements of the cyclin E regulatory complex, namely, DP1 and its partners E2F3 and E2F4. Expression of HCE1 in Neospora caninum, or in uninfected human foreskin fibroblasts (HFFs), showed localization of the expressed protein to the host nuclei and strong cyclin E upregulation. Thus, HCE1 is a novel effector protein that is necessary and sufficient to impact the E2F axis of transcription, resulting in co-opting of host functions to the advantage of ToxoplasmaIMPORTANCE Like most Apicomplexan parasites, Toxoplasma gondii has the remarkable ability to invade and establish a replicative niche within another eukaryotic cell, in this case, any of a large number of cell types in almost any warm-blooded animals. Part of the process of establishing this niche is the export of effector proteins to co-opt host cell functions in favor of the parasite. Here we identify a novel effector protein, HCE1, that the parasites export into the nucleus of human cells, where it modulates the expression of multiple genes, including the gene encoding cyclin E, one of the most crucial proteins involved in controlling when and whether a human cell divides. We show that HCE1 works through binding to specific transcription factors, namely, E2F3, E2F4, and DP1, that normally carefully regulate these all-important pathways. This represents a new way in which these consummately efficient infectious agents co-opt the human cells that they so efficiently grow within.


Asunto(s)
Ciclina E/biosíntesis , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Miosina Tipo I/metabolismo , Proteínas Protozoarias/metabolismo , Toxoplasma/fisiología , Factores de Virulencia/metabolismo , Células Cultivadas , Factores de Transcripción E2F/metabolismo , Fibroblastos/parasitología , Humanos , Unión Proteica , Transporte de Proteínas
7.
J Clin Oncol ; 37(14): 1169-1178, 2019 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-30807234

RESUMEN

PURPOSE: A large-panel gene expression analysis was conducted to identify biomarkers associated with the effectiveness of adding palbociclib to fulvestrant. METHODS: The PALOMA-3 ( ClinicalTrials.gov identifier: NCT01942135) trial randomly assigned 521 endocrine-pretreated patients with metastatic breast cancer to receive palbociclib plus fulvestrant or placebo plus fulvestrant. Primary analysis was first conducted on 10 genes on the basis of pathway biology and evidence from previous studies followed by a systematic panel-wide search among 2,534 cancer-related genes. The association of gene expression with the effect of palbociclib on progression-free survival (PFS) was evaluated using Cox proportional hazards regression analysis, with gene expression as a continuous variable or dichotomized by median. An independent breast cancer cohort from the Preoperative Palbociclib (POP) Clinical Trial ( ClinicalTrials.gov identifier: NCT02008734) was used for validation, in 61 patients with primary breast cancer treated with 2 weeks of palbociclib. RESULTS: In the PALOMA-3 trial, 302 patients had tumor tissue analyzed (palbociclib arm, 194 patients; placebo arm, 108 patients). Palbociclib efficacy was lower in patients with high versus low cyclin E1 (CCNE1) mRNA expression (median PFS: palbociclib arm, 7.6 v 14.1 months; placebo arm, 4.0 v 4.8 months, respectively; interaction P unadjusted = .00238; false discovery rate-adjusted P = .0238). CCNE1 mRNA was more predictive in metastatic than in archival primary biopsy tissue samples. No significant interaction was found between treatment and expression levels of CDK4, CDK6, cyclin D1, and RB1. Palbociclib was efficacious in both luminal A and luminal B tumors. High CCNE1 mRNA expression was associated with poor antiproliferative activity of palbociclib in the POP trial (P = .005). CONCLUSION: Addition of palbociclib to fulvestrant demonstrated efficacy in all biomarker groups, although high CCNE1 mRNA expression was associated with relative resistance to palbociclib.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Ciclina E/genética , Fulvestrant/uso terapéutico , Proteínas Oncogénicas/genética , Biomarcadores de Tumor/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclina E/biosíntesis , Femenino , Fulvestrant/administración & dosificación , Perfilación de la Expresión Génica , Humanos , Metástasis de la Neoplasia , Proteínas Oncogénicas/biosíntesis , Piperazinas/administración & dosificación , Supervivencia sin Progresión , Modelos de Riesgos Proporcionales , Piridinas/administración & dosificación , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo
8.
Gynecol Oncol ; 151(2): 327-336, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30209015

RESUMEN

OBJECTIVES: Readily apparent cyclin E1 expression occurs in 50% of HGSOC, but only half are linked to 19q12 locus amplification. The amplified/cyclin E1hi subset has intact BRCA1/2, unfavorable outcome, and is potentially therapeutically targetable. We studied whether non-amplified/cyclin E1hi HGSOC has similar characteristics. We also assessed the expression of cyclin E1 degradation-associated proteins, FBXW7 and USP28, as potential drivers of high cyclin E1 expression in both subsets. METHODS: 262 HGSOC cases were analyzed by in situ hybridization for 19q12 locus amplification and immunohistochemistry for cyclin E1, URI1 (another protein encoded by the 19q12 locus), FBXW7 and USP28 expression. Tumors were classified by 19q12 amplification status and correlated to cyclin E1 and URI1 expression, BRCA1/2 germline mutation, FBXW7 and USP28 expression, and clinical outcomes. Additionally, we assessed the relative genomic instability of amplified/cyclin E1hi and non-amplified/cyclin E1hi groups of HGSOC datasets from The Cancer Genome Atlas. RESULTS: Of the 82 cyclin E1hi cases, 43 (52%) were amplified and 39 (48%) were non-amplified. Unlike amplified tumors, non-amplified/cyclin E1hi tumor status was not mutually exclusive with gBRCA1/2 mutation. The non-amplified/cyclin E1hi group had significantly increased USP28, while the amplified/cyclin E1hi cancers had significantly lower FBXW7 expression consistent with a role for both in stabilizing cyclin E1. Notably, only the amplified/cyclin E1hi subset was associated with genomic instability and had a worse outcome than non-amplified/cyclin E1hi group. CONCLUSIONS: Amplified/cyclin E1hi and non-amplified/cyclin E1hi tumors have different pathological and biological characteristics and clinical outcomes indicating that they are separate subsets of cyclin E1hi HGSOC.


Asunto(s)
Cromosomas Humanos Par 19 , Ciclina E/genética , Cistadenocarcinoma Seroso/genética , Proteínas Oncogénicas/genética , Neoplasias Ováricas/genética , Adulto , Anciano , Anciano de 80 o más Años , Proteína BRCA1/biosíntesis , Proteína BRCA1/genética , Proteína BRCA2/biosíntesis , Proteína BRCA2/genética , Ciclina E/biosíntesis , Cistadenocarcinoma Seroso/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD/biosíntesis , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Femenino , Amplificación de Genes , Inestabilidad Genómica , Humanos , Persona de Mediana Edad , Proteínas Oncogénicas/biosíntesis , Neoplasias Ováricas/metabolismo , Ubiquitina Tiolesterasa/biosíntesis , Ubiquitina Tiolesterasa/genética
9.
Proc Natl Acad Sci U S A ; 115(37): 9282-9287, 2018 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-30150405

RESUMEN

E-type cyclins E1 (CcnE1) and E2 (CcnE2) are regulatory subunits of cyclin-dependent kinase 2 (Cdk2) and thought to control the transition of quiescent cells into the cell cycle. Initial findings indicated that CcnE1 and CcnE2 have largely overlapping functions for cancer development in several tumor entities including hepatocellular carcinoma (HCC). In the present study, we dissected the differential contributions of CcnE1, CcnE2, and Cdk2 for initiation and progression of HCC in mice and patients. To this end, we tested the HCC susceptibility in mice with constitutive deficiency for CcnE1 or CcnE2 as well as in mice lacking Cdk2 in hepatocytes. Genetic inactivation of CcnE1 largely prevented development of liver cancer in mice in two established HCC models, while ablation of CcnE2 had no effect on hepatocarcinogenesis. Importantly, CcnE1-driven HCC initiation was dependent on Cdk2. However, isolated primary hepatoma cells typically acquired independence on CcnE1 and Cdk2 with increasing progression in vitro, which was associated with a gene signature involving secondary induction of CcnE2 and up-regulation of cell cycle and DNA repair pathways. Importantly, a similar expression profile was also found in HCC patients with elevated CcnE2 expression and poor survival. In general, overall survival in HCC patients was synergistically affected by expression of CcnE1 and CcnE2, but not through Cdk2. Our study suggests that HCC initiation specifically depends on CcnE1 and Cdk2, while HCC progression requires expression of any E-cyclin, but no Cdk2.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Transformación Celular Neoplásica/metabolismo , Ciclina E/biosíntesis , Quinasa 2 Dependiente de la Ciclina/biosíntesis , Reparación del ADN , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/metabolismo , Proteínas Oncogénicas/biosíntesis , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Ciclina E/genética , Quinasa 2 Dependiente de la Ciclina/genética , Ciclinas/biosíntesis , Ciclinas/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Ratones , Ratones Noqueados , Proteínas Oncogénicas/genética
10.
Int J Med Microbiol ; 308(7): 819-828, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30033344

RESUMEN

Ureaplasma spp. are known to be associated with human genitourinary tract diseases and perinatal diseases and Ureaplasma spp. Lipid-associated membrane proteins (LAMPs) play important roles in their related diseases. However, the exact mechanism underlying pathogenesis of Ureaplasma spp. LAMPs is largely unknown. In this study, we explored the pathogenic mechanisms of Ureaplasma spp. LAMPs by elucidating their role in modulating the cell cycle and related signaling pathways in human monocytic cell U937, which is highly related to the inflammatory and protective effect in infectious diseases. We utilized the two ATCC reference strains (Ureaplasma parvum serovar 3 str. ATCC 27,815 (UPA3) and Ureaplasma urealyticum serovar 8 str. ATCC 27,618 (UUR8)) and nine clinical isolates which including both UPA and UUR to study the effects of Ureaplasma spp. LAMPs on U937 in vitro. We found that LAMPs derived from UUR8 and both UPA and UUR of clinical strains markedly inhibited the cell proliferation, while UPA3 LAMPs suppressed slightly. Besides, the result of flow cytometry analysis indicated all the Ureaplasma spp. LAMPs could arrest U937 cells in G1 phase. Next, we found that the cell cycle arrest was associated with increased levels of p53 and p21, and a concomitant decrease in the levels of CDK2, CDK4, CDK6 and cyclin E1 at both transcriptional and translational levels after treatment with LAMPs derived from UUR8 or clinical strains, while only cyclin E1 was down-regulated after treatment with UPA3 LAMPs. Further study showed that p53 down-regulation had almost no effect on the distribution of cell cycle and the expression of p21. In conclusion, this study demonstrated that LAMPs derived from UUR8 and clinical strains could inhibit the proliferation of U937 cells by inducing G1 cell cycle arrest through increasing the p21 expression in a p53-independent manner, while UPA3 LAMPs could induce the cell cycle arrest slightly. Our study could contribute to the understanding of Ureaplasma spp. pathogenesis, which has potential value for the treatment of Ureaplasma spp. infections.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/fisiología , Lipoproteínas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Infecciones por Ureaplasma/patología , Ureaplasma/patogenicidad , Línea Celular Tumoral , Proliferación Celular/fisiología , Ciclina E/biosíntesis , Quinasa 2 Dependiente de la Ciclina/biosíntesis , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Quinasa 6 Dependiente de la Ciclina/biosíntesis , Humanos , Proteínas Oncogénicas/biosíntesis , Células U937 , Ureaplasma/aislamiento & purificación , Enfermedades Urológicas/microbiología
11.
Life Sci ; 205: 131-135, 2018 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-29750990

RESUMEN

AIMS: Cerebral atrophy resulting from neurodegeneration is highly prevalent in individuals with diabetes; however, the underlying mechanisms for diabetic neurodegeneration are not fully understood. Here we hypothesized that hyperglycemia induces molecular signaling that favors induction of proliferation in post mitotic, fully differentiated hippocampal neurons. MATERIALS AND METHODS: Streptozotocin (150 mg/kg) was intraperitoneally injected to four months old male mice to induce diabetes. Hippocampal tissue was subjected to molecular analysis of wingless-related integration site, extracellular signal regulated kinase, and brain derived neurotrophic factor signaling, and cell cycle regulation. KEY FINDINGS: Hyperglycemia did not alter wingless-related integration site signaling or cyclin E levels in the hippocampus. There were reductions in extracellular signal regulated kinase activation and brain derived neurotrophic factor levels along with elevated cyclin D1 levels. SIGNIFICANCE: These findings indicate that hyperglycemic conditions can stimulate cell cycle progression in the hippocampus in vivo. These new insights into the disease mechanisms could support the development of novel therapeutics aimed to provide neuroprotection in diabetic patients.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Hipocampo/patología , Transducción de Señal , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Diferenciación Celular/efectos de los fármacos , Ciclina D1/biosíntesis , Ciclina D1/genética , Ciclina E/biosíntesis , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Mitosis/efectos de los fármacos , Neuronas/efectos de los fármacos , Proteínas Oncogénicas/biosíntesis , Vía de Señalización Wnt/efectos de los fármacos
12.
J Antibiot (Tokyo) ; 71(9): 808-817, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29752478

RESUMEN

Bafilomycin C1, which was isolated from Streptomyces albolongus in our previous work, exhibited strong cytotoxicity against several cancer cell lines. This study aimed to evaluate its antitumor effect on human hepatocellular cancer SMMC7721 cells and the underlying mechanism in vitro and in vivo. MTT assay revealed that bafilomycin C1 retarded SMMC7721 cell growth and proliferation. Western blot and real-time qPCR analysis revealed that bafilomycin C1 caused partial G0/G1 phase cell-cycle arrest, downregulated the expression of cyclin D3, cyclin E1, CDK2, CDK4, and CDK6 and upregulated the expression of p21. Moreover, bafilomycin C1 caused mitochondrial membrane dysfunction through oxidative stress. Furthermore, bafilomycin C1 decreased the expression of Bcl-2; increased the expression of Bax, p53, and P-p53; and increased cleavage of caspase-9 and caspase-3, thereby inducing the intrinsic caspase-dependent apoptotic pathway. In vivo experiments in mice suggested that bafilomycin C1 suppressed tumor growth with few side effects. Cell-cycle arrest and induced apoptosis in tumor tissues in a mouse model treated with bafilomycin C1 were demonstrated by histological analyses, western blot and TUNEL. These findings indicate that bafilomycin C1 may be a promising candidate for hepatic cellular cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/tratamiento farmacológico , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Neoplasias Hepáticas/tratamiento farmacológico , Macrólidos/farmacología , Animales , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina D3/biosíntesis , Ciclina E/biosíntesis , Quinasa 2 Dependiente de la Ciclina/biosíntesis , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Quinasa 6 Dependiente de la Ciclina/biosíntesis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Células Hep G2 , Humanos , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/metabolismo , Membranas Mitocondriales/patología , Proteínas Oncogénicas/biosíntesis , Estrés Oxidativo/efectos de los fármacos , Streptomyces/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Development ; 145(10)2018 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-29695611

RESUMEN

Adult C. elegans germline stem cells (GSCs) and mouse embryonic stem cells (mESCs) exhibit a non-canonical cell cycle structure with an abbreviated G1 phase and phase-independent expression of Cdk2 and cyclin E. Mechanisms that promote the abbreviated cell cycle remain unknown, as do the consequences of not maintaining an abbreviated cell cycle in these tissues. In GSCs, we discovered that loss of gsk-3 results in reduced GSC proliferation without changes in differentiation or responsiveness to GLP-1/Notch signaling. We find that DPL-1 transcriptional activity inhibits CDK-2 mRNA accumulation in GSCs, which leads to slower S-phase entry and progression. Inhibition of dpl-1 or transgenic expression of CDK-2 via a heterologous germline promoter rescues the S-phase entry and progression defects of the gsk-3 mutants, demonstrating that transcriptional regulation rather than post-translational control of CDK-2 establishes the abbreviated cell cycle structure in GSCs. This highlights an inhibitory cascade wherein GSK-3 inhibits DPL-1 and DPL-1 inhibits cdk-2 transcription. Constitutive GSK-3 activity through this cascade maintains an abbreviated cell cycle structure to permit the efficient proliferation of GSCs necessary for continuous tissue output.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/embriología , Quinasa 2 Dependiente de la Ciclina/biosíntesis , Células Germinativas/citología , Glucógeno Sintasa Quinasa 3/metabolismo , Fase S/fisiología , Células Madre/citología , Factores de Transcripción/genética , Animales , Caenorhabditis elegans/citología , Proteínas de Caenorhabditis elegans/metabolismo , Diferenciación Celular/genética , Proliferación Celular/genética , Ciclina E/biosíntesis , Quinasa 2 Dependiente de la Ciclina/genética , Glucógeno Sintasa Quinasa 3/genética , Interferencia de ARN , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Receptores Notch/metabolismo , Transducción de Señal/genética , Transcripción Genética/genética
14.
Tumour Biol ; 39(6): 1010428317705515, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28618963

RESUMEN

Noxin (also called chromosome 11 open reading frame 82 or DNA damage-induced apoptosis suppressor) is associated with anti-apoptosis and cell proliferation in response to stress signals. However, to our knowledge, the role of Noxin in regulating cell proliferation is still controversial and there are no reports of the function and clinicopathological association in breast cancer. In this study, immunohistochemistry results showed that Noxin expression was significantly correlated with advanced tumor-node-metastasis stage ( p = 0.027), positive regional lymph node metastasis ( p = 0.002), and poor overall survival ( p = 0.002). Proliferation assay results showed that Noxin obviously promoted the ability of proliferation of normal breast cells. Subsequent western blot results revealed that Cyclin D1 and Cyclin E1 were upregulated by overexpressing Noxin, whereas Cyclin D1 and Cyclin E1 were downregulated after depleting Noxin. The levels of phosphorylated P38 and activating transcription factor 2 were obviously increased after overexpressing Noxin, and their expression was downregulated accordingly by transfecting Noxin-small interfering RNA. Moreover, P38 inhibitor counteracted the elevating expression of phosphorylated activating transcription factor 2, Cyclin D1, and Cyclin E1 induced by Noxin overexpression and thereby reversed the effect of Noxin overexpression on facilitating cell growth. Taken together, our studies indicated that Noxin was overexpressed in breast cancer and its positive expression was significantly correlated with advance tumor-node-metastasis stage, positive lymph node metastasis, and poor prognosis. Noxin facilitated the expression of Cyclin D1 and Cyclin E1 through activating P38-activating transcription factor 2 signaling pathway, thus enhanced cell growth of breast cancer.


Asunto(s)
Factor de Transcripción Activador 2/genética , Proteínas Reguladoras de la Apoptosis/genética , Neoplasias de la Mama/genética , Ciclina D1/biosíntesis , Ciclina E/biosíntesis , Proteínas Oncogénicas/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Factor de Transcripción Activador 2/biosíntesis , Adulto , Apoptosis/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Ciclo Celular/genética , Proteínas de Ciclo Celular , Proliferación Celular/genética , Ciclina D1/genética , Ciclina E/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis Linfática , Células MCF-7 , Persona de Mediana Edad , Estadificación de Neoplasias , Proteínas Oncogénicas/genética , ARN Interferente Pequeño/genética , Transducción de Señal/genética , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
15.
Sci Rep ; 7(1): 240, 2017 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-28325912

RESUMEN

The development and differentiation of steroidogenic organs are controlled by Ad4BP/SF-1 (adrenal 4 binding protein/steroidogenic factor 1). Besides, lysosomal activity is required for steroidogenesis and also enables adrenocortical cell to survive during stress. However, the role of lysosomal activity on steroidogenic cell growth is as yet unknown. Here, we showed that lysosomal activity maintained Ad4BP/SF-1 protein stability for proper steroidogenic cell growth. Treatment of cells with lysosomal inhibitors reduced steroidogenic cell growth in vitro. Suppression of autophagy did not affect cell growth indicating that autophagy was dispensable for steroidogenic cell growth. When lysosomal activity was inhibited, the protein stability of Ad4BP/SF-1 was reduced leading to reduced S phase entry. Interestingly, treatment of cells with lysosomal inhibitors reduced glycolytic gene expression and supplying the cells with pyruvate alleviated the growth defect. ChIP-sequence/ChIP studies indicated that Ad4BP/SF-1 binds to the upstream region of Ccne1 (cyclin E1) gene during G1/S phase. In addition, treatment of zebrafish embryo with lysosomal inhibitor reduced the levels of the interrenal (adrenal) gland markers. Thus lysosomal activity maintains steroidogenic cell growth via stabilizing Ad4BP/SF-1 protein.


Asunto(s)
Proliferación Celular , Ciclina E/biosíntesis , Lisosomas/metabolismo , Proteínas Oncogénicas/biosíntesis , Factor Esteroidogénico 1/metabolismo , Animales , Células Cultivadas , Glucólisis , Ratones , Pez Cebra/embriología
16.
Tumour Biol ; 39(3): 1010428317695027, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28351306

RESUMEN

MTA3 overexpression has been implicated in carcinogenesis. The aim of the present study was to explore the clinical significance and biological roles of MTA3 in human colorectal cancer and colorectal cancer cells. A total of 80 cases of colorectal cancer tissues were examined by immunohistochemistry for MTA3 protein expression. We analyzed the relationship between MTA3 and clinical factors and the results showed that MTA3 was overexpressed in 51.25% (41/80) cancer cases. There was significant associations between MTA3 overexpression and advanced TNM stage (p = 0.0086) and Ki67 index (p = 0.001). We overexpressed MTA3 in LoVo cells and depleted its expression in HCT15 cells. The results showed that MTA3 promoted cancer cell proliferation, invasion, migration, and cell cycle progression, and inhibited 5-fluorouracil-induced apoptosis in LoVo cell line. MTA3 depletion in HCT15 cell line showed the opposite effects. In addition, we found that MTA3 positively regulated cell cycle proteins including cyclin D1 and cyclin E. It also upregulated Bcl2 and downregulated Bax expression. Furthermore, we found that MTA3 could activate Wnt signaling pathway by upregulating Wnt target proteins. Our results demonstrated that MTA3 overexpression contributes to colorectal cancer carcinogenesis, progression, and chemoresistance. MTA3 could serve as a potential therapeutic target in colorectal cancer.


Asunto(s)
Carcinogénesis/genética , Proliferación Celular/genética , Neoplasias Colorrectales/genética , Proteínas de Neoplasias/genética , Adulto , Anciano , Apoptosis/genética , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Ciclina D1/biosíntesis , Ciclina E/biosíntesis , Progresión de la Enfermedad , Femenino , Fluorouracilo/administración & dosificación , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/biosíntesis , Estadificación de Neoplasias , Proteínas Oncogénicas/biosíntesis , Vía de Señalización Wnt/efectos de los fármacos
17.
Neoplasma ; 63(5): 705-16, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27468874

RESUMEN

Increasing evidences have shown that B-cell translocation gene 3 (BTG3) inhibits metastasis of multiple cancer cells. However, the role of BTG3 in colorectal cancer (CRC) and its possible mechanism have not yet been reported. In our study, we evaluated BTG3 expression in several CRC cell lines. Then, pcDNA3.1-BTG3 was transfected into SW480 cells. We found that BTG3 was upregulated in SW480 cells after overexpression plasmid transfection. BTG3 overexpression significantly inhibited cell growth and decreased PCNA (proliferating cell nuclear antigen) and Ki67 levels. BTG3 overexpression markedly downregulated Cyclin D1 and Cyclin E1 levels, whereas elevated p27. Overexpression of BTG3 arrested the cell cycle at G1 phase, which was abrogated by p27 silencing. Furthermore, migration, invasion and EMT of SW480 cells were significantly suppressed by BTG3 overexpression. Further investigations showed the inhibition of Wnt/ß-catenin signaling pathway. We then used GSK3ß specific inhibitor SB-216763 to activate the Wnt/ß-catenin signaling pathway. We found that Wnt/ß-catenin signaling pathway activation reversed the effect of BTG3 overexpression on cell proliferation, cell cycle progression, invasion and EMT. In conclusion, BTG3 overexpression inhibited cell growth, induced cell cycle arrest and suppressed the metastasis of SW480 cells via the Wnt/ß-catenin signaling pathway. BTG3 may be considered as a therapeutic target in CRC treatment.


Asunto(s)
Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Colorrectales/patología , Proteínas/metabolismo , Vía de Señalización Wnt/genética , Células CACO-2 , Proteínas de Ciclo Celular , Línea Celular Tumoral , Ciclina D1/biosíntesis , Ciclina E/biosíntesis , Transición Epitelial-Mesenquimal/genética , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Humanos , Indoles/farmacología , Antígeno Ki-67/metabolismo , Maleimidas/farmacología , Invasividad Neoplásica/patología , Proteínas Oncogénicas/biosíntesis , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética
18.
Oncol Rep ; 36(1): 10-22, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27176634

RESUMEN

Long non-coding RNAs (lncRNAs) have been identified to be critical mediators in various tumors associated with cancer progression. Long non-coding RNA activated by TGF-ß (lncRNA-ATB) is a stimulator of epithelial-mesenchymal transition (EMT) and serves as a novel prognostic biomarker for hepatocellular carcinoma. However, the biological role and clinical significance of lncRNA-ATB in human prostate cancer have yet to be fully elucidated. The present study was designed to explore the expression of lncRNA-ATB in human prostate cancer patients and the role of lncRNA-ATB in prostate cancer cells. We showed that lncRNA-ATB expression was significantly upregulated in tumor tissues in patients with prostate cancer in comparison with adjacent non-tumor tissues. Further analysis indicted that high lncRNA-ATB expression may be an independent prognostic factor for biochemical recurrence (BCR)-free survival in prostate cancer patients. Overexpression of lncRNA-ATB promoted, and knockdown of lncRNA-ATB inhibited the growth of prostate cancer cells via regulations of cell cycle regulatory protein expression levels. In addition, lncRNA-ATB stimulated epithelial-mesenchymal transition (EMT) associated with ZEB1 and ZNF217 expression levels via ERK and PI3K/AKT signaling pathways. These results indicated that lncRNA-ATB may be considered as a new predictor in the clinical prognosis of patients with prostate cancer. Overexpression of lncRNA-ATB exerts mitogenic and EMT effects of prostate cancer via activation of ERK and PI3K/AKT signaling pathways.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Largo no Codificante/genética , Cadherinas/biosíntesis , Línea Celular Tumoral , Proliferación Celular/genética , Ciclina D1/biosíntesis , Ciclina E/biosíntesis , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Invasividad Neoplásica/genética , Proteínas Oncogénicas/biosíntesis , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/mortalidad , Neoplasias de la Próstata/terapia , Interferencia de ARN , ARN Interferente Pequeño/genética , Transducción de Señal/genética , Transactivadores/biosíntesis , Vimentina/biosíntesis , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/biosíntesis , Proteína de la Zonula Occludens-1/biosíntesis
19.
J Pharm Pharmacol ; 68(7): 901-11, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27138740

RESUMEN

OBJECTIVES: Our study aimed to evaluate the effect of cordycepin on human lung cancer cell lines A549 and NCI-H460. METHODS: Human lung cancer A549 cells and NCI-H460 cells were treated with different concentrations of cordycepin for different times. Cells incubated without cordycepin were defined as a control. The cell proliferation, migration and apoptosis were, respectively, determined by MTT assay, transwell migration assay and flow cytometry. Additionally, the expression levels of related proteins associated with cell cycle, epithelial-mesenchymal transition (EMT) and apoptosis were examined. KEY FINDINGS: The survival rate of A549 cells and NCI-H460 cells treated with cordycepin significantly decreased compared with untreated cells in a concentration-dependent manner, while the apoptosis rate increased. The migration number of cells treated with cordycepin significantly decreased as the increase in concentration. qRT-PCR and Western blot analysis showed that the aberrant expression of related molecules associated with cell cycle, migration and apoptosis was observed in the lung cancer cells, such as cyclin B, cyclin E, MMP-9, caspase-3 and Bcl-2. CONCLUSIONS: Cordycepin may exert inhibitory effects on the development of human lung cancer via inhibiting cell proliferation, suppressing migration and inducing apoptosis, suggesting that cordycepin may have therapeutic potential for the treatment of this disease.


Asunto(s)
Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Desoxiadenosinas/farmacología , Cadherinas/biosíntesis , Caspasa 3/biosíntesis , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ciclina B/biosíntesis , Ciclina D/biosíntesis , Ciclina E/biosíntesis , Relación Dosis-Respuesta a Droga , Humanos , Metaloproteinasa 9 de la Matriz/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Factores de Tiempo , Vimentina/biosíntesis
20.
Neoplasma ; 63(3): 385-93, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26925784

RESUMEN

The objective of the study was to investigate the impact of BTG2 on growth, migration and invasion of human clear cell renal cell carcinoma (ccRCC) cells. Endogenous expression of BTG2 was evaluated in the ccRCC cell lines (Caki-1, 786-O and Caki-2) and noncancerous human renal proximal tubular cell lines (HKC, HK-2 and RPTEC). BTG2 expression was decreased in the ccRCC cells compared with the noncancerous cells (P < 0.01). Then Caki-1 and 786-O cells described as suitable transfection hosts were used in transfection to carry out biological function studies. The three experimental groups were as follows: BTG2-ORF (transfected with BTG2-ORF plasmid), blank-Vector (transfected with pCMV6-Entry), and Cell-alone group (no DNA transfected in). BTG2 expression in the BTG2-ORF groups was significantly higher than that in the controls (P < 0.01). Cell growth was remarkably reduced and the number of migrating or invading cells was reduced in the BTG2-ORF groups compared with the controls (P < 0.01). Furthermore, Matrix Metalloproteinase-9 (MMP-9), Cyclin D1 and Cyclin E expression were reduced in the BTG2-ORF groups compared with the controls. Here, we have provided data for attenuated BTG2 expression in the ccRCC cells. Overexpressed BTG2 could inhibit cell proliferation, migration and invasion of human ccRCC, and the suppressive effects might be due to down-regulation of MMP-9, Cyclin D1 and Cyclin E expression.


Asunto(s)
Carcinoma de Células Renales/metabolismo , Proteínas Inmediatas-Precoces/biosíntesis , Neoplasias Renales/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Ciclina D1/biosíntesis , Ciclina E/biosíntesis , Puntos de Control de la Fase G1 del Ciclo Celular , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Neoplasias Renales/genética , Neoplasias Renales/patología , Metaloproteinasa 9 de la Matriz/biosíntesis , Invasividad Neoplásica , Fase de Descanso del Ciclo Celular , Transfección , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...