Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell Biochem ; 462(1-2): 25-31, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31440879

RESUMO

Resveratrol, a phytoalexin present in grapes and other edible foods, has been reported to have beneficial effects against various diseases including cancer. We previously reported that resveratrol and its derivative, caffeic acid-adducted resveratrol, selectively inhibit the three-dimensional (3D) proliferation of a human colorectal cancer cell line, HCT116 with activating KRAS mutation. Herein, we demonstrated that a novel compound, ferulic acid-bound resveratrol, also represses the 3D proliferation of HCT116 cells. We observed that resveratrol conjugated to two ferulic acids represses the 3D proliferation of HCT116 cells more strongly than resveratrol and resveratrol conjugated to one ferulic acid. Resveratrol conjugated to two ferulic acids also inhibited the 3D proliferation of MCF7 human breast cancer cells. We further uncovered that the resveratrol derivative increases the mRNA level of the tumor suppressor p15, a CDK inhibitor that functions as a brake of cell proliferation in HCT116 cells. These results imply that the resveratrol derivative represses 3D proliferation via increasing p15 expression in HCT116 cells.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Ácidos Cumáricos/farmacologia , Inibidor de Quinase Dependente de Ciclina p15/genética , Genes Supressores de Tumor , Resveratrol/farmacologia , Proliferação de Células/efeitos dos fármacos , Ácidos Cumáricos/química , Inibidor de Quinase Dependente de Ciclina p15/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Humanos , Concentração Inibidora 50 , Células MCF-7 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Resveratrol/química
2.
Mol Cell Biochem ; 442(1-2): 39-45, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-28936721

RESUMO

Resveratrol is a polyphenolic compound in many edible foods including grapes, peanuts, and berries. Several studies have revealed the beneficial effects of resveratrol against various diseases such as heart disease, diabetes, obesity, neurological disorders, and cancer. A recent study showed that resveratrol inhibits the proliferation of HCT116 human colorectal cancer cells in three-dimensional culture (3DC) via induction of luminal apoptosis in HCT116 cell spheroids. In this study, we showed that a novel compound, caffeic acid-adducted resveratrol, has a stronger inhibitory effect on the growth of HCT116 cell spheroids in 3DC than resveratrol. It showed almost the same inhibitory efficacy as 5-fluorouracil, a conventional anticancer drug. We further showed that the resveratrol derivative did not affect the growth of HKe3 cell spheroids derived from HCT116 cells by disruption of the activating mutant KRAS gene. These results suggest that the resveratrol derivative inhibits the growth of HCT116 cell spheroids via inhibition of an oncogenic KRAS-mediated signaling pathway.


Assuntos
Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Mutação , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estilbenos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Humanos , Células MCF-7 , Proteínas Proto-Oncogênicas p21(ras)/genética , Resveratrol
3.
EMBO J ; 31(10): 2365-77, 2012 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-22505024

RESUMO

The tumour suppressor gene product Mig-6 acts as an inhibitor of epidermal growth factor (EGF) signalling. However, its posttranslational modifications and regulatory mechanisms have not been elucidated. Here, we investigated the phosphorylation of human Mig-6 and found that Chk1 phosphorylated Mig-6 in vivo as well as in vitro. Moreover, EGF stimulation promoted phosphorylation of Mig-6 without DNA damage and the phosphorylation was inhibited by depletion of Chk1. EGF also increased Ser280-phosphorylated Chk1, a cytoplasmic-tethering form, via PI3K pathway. Mass spectrometric analyses suggested that Ser 251 of Mig-6 was a major phosphorylation site by Chk1 in vitro and in vivo. Substitution of Ser 251 to alanine increased inhibitory activity of Mig-6 against EGF receptor (EGFR) activation. Moreover, EGF-dependent activation of EGFR and cell growth were inhibited by Chk1 depletion, and were rescued by co-depletion of Mig-6. Our results suggest that Chk1 phosphorylates Mig-6 on Ser 251, resulting in the inhibition of Mig-6, and that Chk1 acts as a positive regulator of EGF signalling. This is a novel function of Chk1.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Proteínas Quinases/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Substituição de Aminoácidos , Quinase 1 do Ponto de Checagem , Regulação da Expressão Gênica , Humanos , Espectrometria de Massas , Mutagênese Sítio-Dirigida , Fosforilação , Serina/genética , Serina/metabolismo , Proteínas Supressoras de Tumor/química
4.
Mol Cell Biochem ; 412(1-2): 141-6, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26708220

RESUMO

ANRIL is a long noncoding RNA transcribed from the INK4 locus that encodes three tumor suppressor genes, p15, p16, and ARF. Previous studies demonstrated that ANRIL represses p15 and p16, which positively regulate the pRB pathway, leading to repression of cellular senescence of human normal fibroblasts. However, the role of ANRIL in cancer cell proliferation is less well understood. Here we report that ANRIL is involved in the proliferation of colorectal cancer HCT116 cells in two- and three-dimensional culture. Silencing ANRIL by both transfection with small interfering RNA and retrovirally produced small hairpin RNA reduced HCT116 cell proliferation in both two- and three-dimensional culture. HCT116 cells depleted for ANRIL were arrested in the S phase of cell cycle. Notably, silencing ANRIL did not result in the activation of expression of the INK4 locus. These results suggest that ANRIL positively regulates the proliferation of HCT116 cells in two- and three-dimensional culture in a p15/p16-pRB pathway-independent manner.


Assuntos
Proliferação de Células/fisiologia , Neoplasias Colorretais/patologia , RNA Longo não Codificante/fisiologia , Linhagem Celular Tumoral , Inativação Gênica , Humanos , RNA Longo não Codificante/genética
5.
Genes Cells ; 19(6): 504-16, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24774443

RESUMO

Cyclin D1, an oncogenic G1 cyclin, and YB-1, a transcription factor involved in cell growth, are both over-expressed in several human cancers. In human lung cancer, the functional association between YB-1 and cyclin D1 has never been elucidated. In this study, we show YB-1 is involved in the transcription of cyclin D1 in human lung cancer. Depletion of endogenous YB-1 by siRNA inhibited progression of G1 phase and down-regulated both the protein and mRNA levels of cyclin D1 in human lung cancer cells. Forced over-expression of YB-1 with a cyclin D1 reporter plasmid increased luciferase activity, and ChIP assay results showed YB-1 bound to the cyclin D1 promoter. Moreover, the amount of YB-1 mRNA positively correlated with cyclin D1 mRNA levels in clinical non-small-cell lung cancer (NSCLC) specimens. Immunohistochemical analysis also indicated YB-1 expression correlated with cyclin D1 expression in NSCLC specimens. In addition, most of the cases expressing both cyclin D1 and CDC6, another molecule controlled by YB-1, had co-existing YB-1 over-expression. Together, our results suggest that aberrant expression of both cyclin D1 and CDC6 by YB-1 over-expression may collaboratively participate in lung carcinogenesis.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Ciclina D1/genética , Neoplasias Pulmonares/metabolismo , Proteína 1 de Ligação a Y-Box/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Grandes/metabolismo , Carcinoma de Células Grandes/patologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Ciclina D1/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Proteína 1 de Ligação a Y-Box/genética
6.
Genes Cells ; 18(11): 999-1006, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24165022

RESUMO

Y box binding protein 1 (YB1) has multiple functions associated with drug resistance, cell proliferation and metastasis through transcriptional and translational regulation. Increased expression of YB1 is closely related to tumor growth and aggressiveness. We showed that YB1 protein levels were decreased through replicative and premature senescence and were correlated with increased expression levels of p16(INK) (4A) tumor suppressor gene. Depletion of YB1 was associated with increased levels of p16 in human and murine primary cells. Forced expression of YB1 in mouse embryonic fibroblasts resulted in decreased expression of p16 and increased cell proliferation. Senescence-associated expression of ß-galactosidase was repressed in YB1-over-expressing cells. Chromatin immunoprecipitation assays showed that YB1 directly associates with the p16 promoter. Taken together, all our findings indicate that YB1 directly binds to and represses p16 transcription, subsequently resulting in the promotion of cell growth and prevention of cellular senescence.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Fatores de Transcrição/genética , Proteína 1 de Ligação a Y-Box/metabolismo , Animais , Senescência Celular , Inibidor p16 de Quinase Dependente de Ciclina/genética , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Genes Supressores de Tumor , Humanos , Camundongos , Regiões Promotoras Genéticas , Fatores de Transcrição/metabolismo , Proteína 1 de Ligação a Y-Box/genética
7.
Cell Mol Life Sci ; 70(24): 4785-94, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23880895

RESUMO

The mammalian cell cycle is precisely controlled by cyclin-dependent kinases (CDKs) and related pathways such as the RB and p53 pathways. Recent research on long non-coding RNAs (lncRNAs) indicates that many lncRNAs are involved in the regulation of critical cell cycle regulators such as the cyclins, CDKs, CDK inhibitors, pRB, and p53. These lncRNAs act as epigenetic regulators, transcription factor regulators, post-transcription regulators, and protein scaffolds. These cell cycle-regulated lncRNAs mainly control cellular levels of cell cycle regulators via various mechanisms, and may provide diversity and reliability to the general cell cycle. Interestingly, several lncRNAs are induced by DNA damage and participate in cell cycle arrest or induction of apoptosis as DNA damage responses. Therefore, deregulations of these cell cycle regulatory lncRNAs may be involved in tumorigenesis, and they are novel candidate molecular targets for cancer therapy and diagnosis.


Assuntos
Pontos de Checagem do Ciclo Celular/genética , RNA Longo não Codificante/genética , Animais , Pontos de Checagem do Ciclo Celular/fisiologia , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Proteínas Inibidoras de Quinase Dependente de Ciclina/metabolismo , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/genética , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/genética , Ciclinas/metabolismo , Epigênese Genética , Humanos , Modelos Biológicos , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
8.
Anticancer Res ; 43(8): 3441-3446, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37500167

RESUMO

BACKGROUND/AIM: CDK inhibitor p16 plays a pivotal role in the induction of cellular senescence and functions as a tumor suppressor. Here, we demonstrate that histone H1.2 is involved in p16 repression. MATERIALS AND METHODS: Cells were transfected with siRNAs and subjected to quantitative reverse transcription-polymerase chain reaction, immunoblotting and chromatin immunoprecipitation (ChIP) assay. RESULTS: The decrease in H1.2 by oncogenic RAS was associated with increased levels of p16. Depletion of H1.2 selectively increased p16, but not alternative reading frame (ARF) mRNA. ChIP assay showed that H1.2 directly bound to the p16 promoter. Interestingly, silencing YB-1, a component of H1.2 complex, decreased the expression levels of H1.2, resulting in decreased binding of H1.2 on the p16 promoter. CONCLUSION: These results provide a model in which H1.2 is positively regulated by YB-1 and directly binds to and represses the transcription of p16.


Assuntos
Genes Supressores de Tumor , Histonas , Humanos , Histonas/genética , Histonas/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Regiões Promotoras Genéticas , Senescência Celular
9.
Genes Cells ; 16(11): 1110-20, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22017545

RESUMO

The cyclin-dependent kinase (CDK) inhibitor p27 level is associated with progression of renal damage. We previously reported that mRNA of Skp2, a component of Skp/Cullin/F-box (SCF)-ubiquitin ligase which targets to p27, was increased in unilateral ureteral obstructive kidneys in mice and that the nephritis was attenuated in Skp2-deficient mice. However, the details have not been fully clarified. Here, we found that not only Skp2 but also cdc kinase subunit 1 (Cks1), an essential cofactor for the SCF-Skp2 ubiquitin ligase in targeting p27, was increased in another chronic progressive model, anti-thymocyte serum (ATS) rat nephropathy. After induction of ATS nephropathy, Skp2(+) /Cks1(+) /Ki67(+) tubular epithelial cell numbers increased, and p27(+) tubular epithelial cells decreased transiently. Moreover, we found that TNFα was involved in expression of both Skp2 and Cks1 in NRK cell line as well as the in ATS nephropathy. Nuclear accumulations of NF-κB subunits RelB and p52 were increased in the tubular epithelial cells of the nephritic kidney. Both Skp2 and Cks1 were colocalized with RelB in these cells. These data suggest that both Skp2 and Cks1 are up-regulated by the TNFα-RelB/p52 pathway in the early stages of renal damage and are collaboratively involved in down-regulation of p27 in proliferative tubular dilation and the progression of chronic nephropathy.


Assuntos
Quinases relacionadas a CDC2 e CDC28/genética , Regulação da Expressão Gênica , NF-kappa B/metabolismo , Nefrite/genética , Proteínas Quinases Associadas a Fase S/genética , Proteínas Ligases SKP Culina F-Box/genética , Fator de Necrose Tumoral alfa/metabolismo , Animais , Soro Antilinfocitário/química , Linhagem Celular , Proliferação de Células , Doença Crônica , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Modelos Animais de Doenças , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Masculino , Camundongos , Nefrite/induzido quimicamente , Ratos , Ratos Wistar , Transdução de Sinais
10.
Nucleic Acid Ther ; 32(5): 438-447, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35404139

RESUMO

In this study, the efficiency of RNA interference of small interfering RNAs (siRNAs) bearing 5'-O-methyl-2'-deoxythymidine (X) and 5'-amino-2', 5'-dideoxythymidine (Z) at the 5'-end of the sense strand and the antisense strand of siRNA was investigated in HeLa cells stably expressing enhanced green fluorescent protein. The results indicated that when one strand of siRNA was modified with X or Z and the other was unmodified, the X or Z modification was predominant in the process of strand selection and the unmodified strand was selected as a guide strand. When both strands are modified with X or Z, the modified antisense strand with X or Z will be selected as a guide strand with a certain probability. The resulting mature RNA-induced silencing complex exerted reduced, but still moderate silencing activity remained. These results suggest that the modification of the sense strand with X or Z eliminates the off-target effects caused by the sense strand without affecting the silencing efficiency of the siRNA.


Assuntos
RNA de Cadeia Dupla , Complexo de Inativação Induzido por RNA , Humanos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Células HeLa , Interferência de RNA , Complexo de Inativação Induzido por RNA/metabolismo , Timidina
11.
Exp Cell Res ; 316(20): 3342-50, 2010 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-20937271

RESUMO

We aimed to clarify the transcription-regulation mechanisms of the mouse telomerase reverse transcriptase gene (mTERT). First, we searched for the promoter region required for transcriptional activation of mTERT and identified an enhancer cis-element (named mTERT-EE) located between -200 and -179bp of the mouse TERT gene (mTERT). EMSA results suggested that nuclear factor of activated T cells (NFAT) member proteins bind to mTERT-EE. We then identified NFAT5 as the factor binding to mTERT-EE and found that it activates the transcription of the mTERT core promoter. The results that siRNA directed against NFAT5 significantly reduced mTERT expression and mTERT core promoter activity and that the expressions of NFAT5 and mTERT were well correlated in various mouse tissues except liver suggest that NFAT5 dominantly and directly regulates mTERT expression. To clarify their functionality further, we investigated the effect of hypertonic stress, a known stimulus affecting the expression and transcriptional activity of NFAT5, on mTERT expression. The result indicated that hypertonic stress activates mTERT transcription via the activation and recruitment of NFAT5 to the mTERT promoter. These results provide useful information about the transcription-regulation mechanisms of mTERT.


Assuntos
Regulação da Expressão Gênica , Telomerase/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Estruturas Animais/metabolismo , Animais , Sítios de Ligação/genética , Extratos Celulares/química , Núcleo Celular/química , Sobrevivência Celular/genética , Ensaio de Desvio de Mobilidade Eletroforética , Elementos Facilitadores Genéticos/genética , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Genes Reporter/genética , Soluções Hipertônicas/farmacologia , Camundongos , Camundongos Endogâmicos , Mutação/genética , Fatores de Transcrição NFATC/metabolismo , Células NIH 3T3 , Regiões Promotoras Genéticas/genética , RNA Interferente Pequeno/genética , Estresse Fisiológico/genética , Fatores de Transcrição/genética
12.
Cancer Genomics Proteomics ; 18(4): 543-548, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34183387

RESUMO

BACKGROUND/AIM: The long noncoding RNA OIP5 antisense RNA 1 (OIP5-AS1) is overexpressed in various cancer types, such as lung cancer, hepatoblastoma and cervical cancer, and functions to accelerate cell proliferation, invasion and migration. Here, we investigated the roIe of OIP5-AS1 in cell-cycle progression of H1299 and A549 non-small cell lung cancer cells, and FaDu and CAL27 head and neck squamous cell carcinoma cells. MATERIALS AND METHODS: The cells were transfected with small interfering RNA and subjected to cell-cycle analysis and reverse-transcription quantitative polymerase chain reaction (RT-qPCR). RESULTS: Silencing of OIP5-AS1 suppressed the proliferation of H1299, A549, FaDu and CAL27 cells. RT-qPCR and cell-cycle analysis revealed that silencing OIP5-AS1 increased the expression of CDK inhibitors, such as p15, p16, p18 and p19, resulting in G1-phase arrest. CONCLUSION: OIP5-AS1 regulates G1-phase progression by repressing CDK inhibitors and, thus, promotes the proliferation of H1299, A549, FaDu and CAL27 cells.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Proliferação de Células/genética , Neoplasias de Cabeça e Pescoço/genética , Neoplasias Pulmonares/genética , RNA Longo não Codificante/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Ciclo Celular/genética , Linhagem Celular Tumoral , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia
13.
J Biol Chem ; 284(41): 27766-27779, 2009 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-19679664

RESUMO

The SCF(Fbw7) ubiquitin ligase complex plays important roles in cell growth, survival, and differentiation via the ubiquitin-proteasome-mediated regulation of protein stability. Fbw7 (also known as Fbxw7, Sel-10, hCdc4, or hAgo), a substrate recognition subunit of SCF(Fbw7) ubiquitin ligase, facilitates the degradation of several proto-oncogene products by the proteasome. Given that mutations in Fbw7 are found in various types of human cancers, Fbw7 is considered to be a potent tumor suppressor. In the present study, we show that E1A, an oncogene product derived from adenovirus, interferes with the activity of the SCF(Fbw7) ubiquitin ligase. E1A interacted with SCF(Fbw7) and attenuated the ubiquitylation of its target proteins in vivo. Furthermore, using in vitro purified SCF(Fbw7) component proteins, we found that E1A directly bound to Roc1/Rbx1 and CUL1 and that E1A inhibited the ubiquitin ligase activity of the Roc1/Rbx1-CUL1 complex but not that of another RING-type ubiquitin ligase, Mdm2. Ectopically expressed E1A interacted with cellular endogenous Roc1/Rbx1 and CUL1 and decelerated the degradation of several protooncogene products that were degraded by SCF(Fbw7) ubiquitin ligase. Moreover, after wild-type adenovirus infection, adenovirus-derived E1A interacted with endogenous Roc1/Rbx1 and decelerated degradation of the endogenous target protein of SCF(Fbw7). These observations demonstrated that E1A perturbs protein turnover regulated by SCF(Fbw7) through the inhibition of SCF(Fbw7) ubiquitin ligase. Our findings may help to explain the mechanism whereby adenovirus infection induces unregulated proliferation.


Assuntos
Proteínas E1A de Adenovirus/metabolismo , Adenovírus Humanos/metabolismo , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas F-Box/antagonistas & inibidores , Subunidades Proteicas/metabolismo , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Proteínas E1A de Adenovirus/genética , Adenovírus Humanos/genética , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Proteínas Culina/genética , Proteínas Culina/metabolismo , Ciclina D1/genética , Ciclina D1/metabolismo , Ciclina E/genética , Ciclina E/metabolismo , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Proteína 7 com Repetições F-Box-WD , Humanos , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Subunidades Proteicas/genética , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-myb/genética , Proteínas Proto-Oncogênicas c-myb/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Especificidade por Substrato , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
15.
Cancer Genomics Proteomics ; 17(4): 425-430, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32576587

RESUMO

BACKGROUND/AIM: The INK4 locus encodes three important genes p15INK4B, p16INK4A, and ARF, which function to suppress oncogenesis, and a long noncoding RNA, ANRIL, which, in contrast, functions to promote oncogenesis. Herein, we report a fifth genetic element on the INK4 locus, a long noncoding RNA with unknown function named associated negative regulation of cell proliferation (ANROC), which played a role in the suppression of cell proliferation. MATERIALS AND METHODS: Following ANROC silencing in cells by siRNA, quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and cell cycle analysis using flow cytometry were performed. RESULTS: ANROC expression was decreased by oncogenic RAS signalling. ANROC knockdown enhanced HeLa cell proliferation and induced cyclin B1 mRNA, which promotes G2/M progression of the cell cycle. Furthermore, flow cytometric analysis revealed that ANROC knockdown increased the percentage of cells in the S and G2/M phases of the cell cycle. CONCLUSION: ANROC functions to suppress cell cycle progression by suppressing cyclin B1 expression, thus inhibiting cell proliferation.


Assuntos
Ciclo Celular , Proliferação de Células , Ciclina B1/metabolismo , Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , RNA Longo não Codificante/genética , Neoplasias do Colo do Útero/patologia , Ciclina B1/genética , Feminino , Células HeLa , Humanos , RNA Longo não Codificante/antagonistas & inibidores , RNA Interferente Pequeno , Neoplasias do Colo do Útero/genética
16.
Nucleosides Nucleotides Nucleic Acids ; 39(1-3): 407-425, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32310030

RESUMO

Telomerase activity has been regarded as a critical step in cellular immortalization and carcinogenesis and because of this, regulation of telomerase represents an attractive target for anti-tumor specific therapeutics. Recently, one avenue of cancer research focuses on antisense strategy to target the oncogenes or cancer driver genes, in a sequence specific fashion to down-regulate the expression of the target gene. The protein catalytic subunit, human telomerase reverse transcriptase (hTERT) and the template RNA component (hTERC) are essential for telomerase function, thus theoretically, inhibition of telomerase activity can be achieved by interfering with either the gene expression of hTERT or the hTERC of the telomerase enzymatic complex. The present study showed that phosphorothioate antisense oligonucleotide (sASO)-nuclear localization signal (NLS) peptide conjugates targeting hTERC could inhibit telomerase activity very efficiently at 5 µM concentration but less efficiently at 1 µM concentration. On the other hand, siRNA targeting hTERT mRNA could strongly suppress hTERT expression at 200 nM concentration. It was also revealed that siRNA targeting hTERT could induce telomere attrition and then irreversible arrest of proliferation of cancer cells.


Assuntos
Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Sinais de Localização Nuclear/química , Oligonucleotídeos Antissenso , Fosfatos/química , Telomerase/antagonistas & inibidores , Telômero/química , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática , Células HeLa , Humanos , Peptídeos/química , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Telomerase/química , Células Tumorais Cultivadas
17.
Cancer Sci ; 100(8): 1374-81, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19459846

RESUMO

Cellular levels of products from both oncogenes and tumor suppressor genes in normal cells need to be critically regulated to avoid malignant transformation. These products are often controlled by the ubiquitin proteasome pathway, the specific degradation mechanism in the cell. E3 ubiquitin ligases polyubiquitylate their specific substrates by collaborating with E1 and E2, and then the modified substrates are degraded in the proteasome. Mdm2 targets p53 and retinoblastoma protein, two major tumor suppressor gene products, for ubiquitin-dependent degradation. SCF(Skp2) targets other tumor suppressor gene products and CDK inhibitors such as p130, Tob1, p27(Kip1), p57(Kip2), and p21(Cip1). Therefore, both E3 ligases act like oncogene products. In contrast, degradation of several oncogene products, such as Cyclin E, Notch, c-Myc, c-Jun, and c-Myb, are mediated by SCF(Fbw7). Fbw7 is often deleted or mutated in human cancers and acts like a tumor suppressor. As well as growth factor receptors and signal transduction regulators, DNA repair-related proteins are also regulated via the ubiquitin-proteasome pathway mediated by their specific E3 ligases. The stabilization of oncogene products and enhanced degradation of tumor suppressor gene products or DNA repair proteins might be associated with carcinogenesis and malignant progression, due to defects or the abnormal expression of their E3 ligases.


Assuntos
Genes Supressores de Tumor , Oncogenes , Proteínas Supressoras de Tumor/genética , Ubiquitina/metabolismo , Humanos
18.
Cancer Sci ; 100(5): 866-72, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19445020

RESUMO

Downregulation of the cyclin-dependent kinase inhibitory protein p27 is frequently observed in various cancers due to enhancement of its degradation. We recently reported that p53-inducible protein with RING-H2 domain (Pirh2) is a novel ubiquitin ligase for p27, required for the ubiquitylation and consequent degradation of p27 protein. However, there is no reports about the involvement of Pirh2 in both p27 downregulation and pathogenesis in human cancers. In the present study, we investigated them using cultured cell lines and surgical specimens derived from human head and neck squamous cell carcinoma (HNSCC). Depletion of Pirh2 by short interfering RNA induced accumulation of p27 and inhibited the growth of cultured HNSCC cells. By immunohistochemical analysis in 57 cases of HNSCC specimens, higher levels of Pirh2 expression (labeling index > or = 60%) were found in 61.4% of HNSCC in comparison with 0% of normal mucosa. In addition, 83.3% of HNSCC with lower p27 expression (labeling index < 20%) displayed high Pirh2 levels. Therefore, Pirh2 expression was inversely correlated with p27 expression. Finally, Pirh2 expression was well correlated with poor prognosis. These findings suggest that Pirh2 overexpression may have an important role in the development and maintenance of HNSCC at least partially through p27 degradation, and that Pirh2 may be a potential molecular target for human HNSCC.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Ubiquitina-Proteína Ligases/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/genética , Humanos , Imuno-Histoquímica , Prognóstico , Ubiquitina-Proteína Ligases/genética
19.
Anticancer Res ; 39(8): 4073-4077, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31366490

RESUMO

BACKGROUND/AIM: ANRIL is a long noncoding RNA located on INK4 locus, which encodes p15 and p16 that cause G1 phase arrest in the cell cycle. ANRIL positively regulates proliferation of several kinds of cancer cells such as lung and gastric cancers. This study, examined the effect of ANRIL in head and neck squamous cell carcinoma cells. MATERIALS AND METHODS: Cells were transfected with siRNA oligonucleotides targeting ANRIL. Transfected cells were subjected to cell-cycle and quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analysis. RESULTS: Depletion of ANRIL increased p15 mRNA in FaDu cells, and p15 and p16 mRNA in CAL27 cells and inhibited proliferation of these cells. Cell cycle analysis showed that depletion of ANRIL caused arrest at the G1 phase of the cell cycle. CONCLUSION: ANRIL promotes G1 phase progression by repressing p15 and p16, and thus promotes FaDu and CAL27 cell proliferation.


Assuntos
Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , RNA Longo não Codificante/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Pontos de Checagem da Fase G1 do Ciclo Celular/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia
20.
Anticancer Res ; 38(1): 77-81, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29277759

RESUMO

BACKGROUND/AIM: OPA-interacting protein 5 antisense transcript 1 (OIP5-AS1) is a long noncoding RNA located on human chromosome 15q15.1 and transcribed in the opposite direction to OIP5. Here, we report that OIP5-AS1 is involved in regulating cell proliferation. MATERIALS AND METHODS: HeLa cells were transfected with OIP5-AS1-targeting siRNA oligonucleotides and anti-sense oligonucleotides. The cells were harvested 72 h after transfection and subjected to quantitative reverse transcription-polymerase chain reaction (qRT-PCR), and cell-cycle and apoptosis analysis. RESULTS: OIP5-AS1 was expressed at a lower level in cells harbouring an oncogenic kirsten rat sarcoma viral oncogene homolog (K-RAS) mutation than in cells expressing wild-type K-RAS. Silencing OIP5-AS1 with siRNA oligonucleotides or anti-sense oligonucleotides reduced HeLa cell proliferation. Apoptosis and cell-cycle analysis showed that silencing OIP5-AS1 did not cause apoptosis, but did cause G2/M phase cell-cycle arrest. CONCLUSION: These results suggest that OIP5-AS1 positively regulates cell proliferation by promoting G2/M phase progression.


Assuntos
Proliferação de Células/genética , RNA Longo não Codificante/genética , Apoptose , Ciclo Celular , Células HCT116 , Células HeLa , Humanos , RNA Interferente Pequeno/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA