Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Biochem Biophys Res Commun ; 509(3): 746-752, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30621914

RESUMO

MicroRNA-501-3p (miR-501-3p) has been reported as a novel cancer-related miRNA in many types of cancer. However, the precise biological function of miR-501-3p in prostate cancer remains unknown. In this study, we aimed to investigate the regulatory effect and mechanism of miR-501-3p on cell growth of prostate cancer cells. We found that miR-501-3p expression was significantly downregulated in prostate cancer tissues and cell lines. Gain-of-function experiments showed that upregulation of miR-501-3p expression significantly decreased cell proliferation and colony formation, and induced cell cycle arrest in the G0/G1 phase. Bioinformatics analysis predicted that cell cycle-related and expression-elevated protein in tumor (CREPT) was a potential target gene of miR-501-3p., and the results of our luciferase reporter assay confirmed that miR-501-3p bound to the 3'-untranslated region of CREPT at the predicted binding site. Moreover, miR-501-3p was shown to negatively regulate CREPT expression in prostate cancer cells. Correlation analysis showed that miR-501-3p was inversely correlated with CREPT expression in prostate cancer tissues. Knockdown studies revealed that miR-501-3p regulated the expression of cyclin D1 by targeting CREPT. Additionally, the inhibitory effect of miR-501-3p on prostate cancer cell growth was partially reversed by CREPT overexpression. Overall, these results suggest that miR-501-3p restricts prostate cancer cell growth by targeting CREPT to inhibit the expression of cyclin D1. These findings indicate that the miR-501-3p/CREPT/cyclin D1 axis plays a crucial role in the progression of prostate cancer and may serve as potential therapeutic target.


Assuntos
Ciclina D1/genética , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Neoplasias da Próstata/genética , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Masculino , Proteínas de Neoplasias/genética , Neoplasias da Próstata/patologia
2.
J Cell Biochem ; 119(1): 1083-1092, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28695988

RESUMO

The CREPT (cell cycle-related and expression elevated protein in tumor, also known as RPRD1B) and p15RS (p15INK4b -related sequence, also known as RPRD1A) have been shown to regulate cell proliferation and alter the cell cycle through Wnt/ß-catenin pathway downstream genes in human. Although several studies have revealed the mechanism by which CREPT and p15RS regulate cell proliferation in human and mammals, it is still unclear how these genes function in poultry. In order to determine the function of CREPT and p15RS in chicken, we examined the expression of CREPT and p15RS in a variety of chicken tissues and DF-1 cells. Then, we determined the effect of overexpression or depletion of CREPT or p15RS, by transiently transfecting chicken DF-1 cells with overexpression and short hairpin RNA (shRNA) vectors respectively, on the regulation of cell proliferation. The results showed that CREPT and p15RS had different expression patterns and opposite effects on the cell cycling and proliferation. Knockdown of p15RS expression or overexpression of CREPT facilitated cell proliferation by promoting the cell-cycle transition from G0/G1 to S-phase and G2/M, whereas knockdown of CREPT or overexpression of p15RS inhibited cell proliferation. Mechanistically, CREPT and p15RS control DF-1 cell proliferation by regulating the expression of Wnt/ß-catenin pathway downstream regulatory genes, including ß-catenin, TCF4, and Cyclin D1. In conclusion, CREPT and p15RS regulate cell proliferation and the cell-cycle transition in chicken DF-1 cells by regulating the transcription of Wnt/ß-catenin pathway downstream regulatory genes.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Transcrição Gênica , Via de Sinalização Wnt , Animais , Ciclo Celular , Proteínas de Ciclo Celular/química , Linhagem Celular , Proliferação de Células , Galinhas , Humanos , Modelos Moleculares , Mapas de Interação de Proteínas , Distribuição Tecidual
3.
Cancer Sci ; 109(4): 1012-1023, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29397041

RESUMO

The cell cycle-related and expression-elevated protein in tumor (CREPT) is overexpressed in several human malignancies. However, the clinical relevance of CREPT expression and its biological role in non-small-cell lung cancer (NSCLC) remains unclear. In this study, we detected the expression of CREPT in both NSCLC tissues and cell lines by immunohistochemistry, Western blot analysis, and RT-PCR. The correlation between CREPT expression and clinicopathologic features was analyzed in 271 NSCLC patients. The prognostic value of CREPT expression was evaluated by Kaplan-Meier analysis and Cox regression analysis. CREPT was overexpressed in Calu-1 cell lines by using plasmid vector and its biological function was explored both in vitro and in vivo. We found that CREPT was significantly overexpressed in NSCLC compared with paired adjacent non-tumor tissues, and the expression level of CREPT was correlated with tumor differentiation, lymph node metastasis, and clinical stage. Kaplan-Meier analysis showed that the recurrence-free survival and overall survival of high CREPT expression groups were significantly shorter than those of the low CREPT expression group. Multivariate analysis identified that CREPT might be an independent biomarker for the prediction of NSCLC prognosis. Overexpression of CREPT increased cell proliferation and enhanced the migration and invasion ability of Calu-1 cells (a human NSCLC cell line with relative low CRPET expression) in vitro. Moreover, CREPT overexpression promoted tumor growth in a nude mice model. These results suggest that CREPT is closely relevant to the proliferation of NSCLC cells and it might be a potential prognostic marker in NSCLC patients.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/fisiologia , Proliferação de Células/fisiologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Proteínas de Neoplasias/metabolismo , Células A549 , Animais , Biomarcadores Tumorais/metabolismo , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Intervalo Livre de Doença , Feminino , Humanos , Metástase Linfática/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Prognóstico
4.
Biochem Biophys Res Commun ; 496(4): 1183-1190, 2018 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-29402413

RESUMO

CREPT (cell-cycle related and expression-elevated protein in tumor) was reported to be associated with growth of several human cancers; however, its clinical significance and regulatory mechanism still remain unclear in human gastric cancer. In the present study, we found CREPT was significantly increased in gastric cancer tissues compared to the matched adjacent normal tissues. CREPT silence inhibited the proliferation of gastric cancer cells through inducing G0/G1 phase cell cycle arrest, which was linked to the reduction of Cyclin D1 and Cyclin D-dependent kinase 4 (CDK4), and the elevation of p53 and p21. In addition, CREPT knockdown (KD) decreased migration of gastric cancer cells through up-regulating E-cadherin and down-regulating vimentin, N-cadherin and matrix metalloproteinase 1 (MMP-1) expressions. Further, CREPT KD induced apoptosis in gastric cancer cells, as evidenced by the increase of cleaved Caspase-3 and poly (ADP-ribose) polymerase (PARP). Intriguingly, suppressing p53 expressions significantly abolished CREPT silence-induced apoptosis, and reduction of cell viability. Moreover, CREPT KD caused reactive oxygen species (ROS) generation using discounted cash flow (DCF) analysis, which was reversed by ROS scavenger, N-acetyl-l-cysteine (NAC), pretreatment. Of note, NAC pretreatment abrogated apoptotic cell death in CREPT KD gastric cancer cells. In vivo, suppressing CREPT reduced the gastric tumor growth in gastric cancer xenograft models. Altogether, our results provided a novel insight into CREPT in regulating gastric cancer progression through apoptosis regulated by ROS/p53 pathways.


Assuntos
Apoptose , Pontos de Checagem do Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Proteínas de Neoplasias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Proteína Supressora de Tumor p53/metabolismo , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Transdução de Sinais
5.
Mol Carcinog ; 57(10): 1408-1420, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29938829

RESUMO

CREPT (Cell-cycle-related and expression-elevated protein in tumor)/RPRD1B, a novel protein that enhances the transcription of Cyclin D1 to promote cell proliferation during tumorigenesis, was demonstrated highly expressed in most of tumors. However, it remains unclear how CREPT is regulated in colorectal cancers. In this study, we report that miR-383 negatively regulates CREPT expression. We observed that CREPT was up-regulated but the expression of miR-383 was down regulated in both colon cancer cell lines and colon tumor tissues. Intriguingly, we found that enforced expression of miR-383 inhibited the expression of CREPT at both the mRNA and protein level. Using a luciferase reporter, we showed that miR-383 targeted the 3'-UTR of CREPT mRNA directly. Consistently we observed that over expression of miR-383 shortened the half-life of CREPT mRNA in varieties of colorectal cancer cells. Furthermore, restoration of miR-383 inhibited cell growth and colony formation of colon cancer cells accompanied by inhibition of expression of CREPT and related downstream genes. Finally, we demonstrated that stable over expression of miR-383 in colon cancer cells decreased the growth of the tumors. Our results revealed that the abundant expression of CREPT in colorectal cancers is attributed to the decreased level of miR-383. This study shed a new light on the potential therapeutic therapy strategy for colorectal cancers using introduced miRNA.


Assuntos
Proteínas de Ciclo Celular/genética , Neoplasias Colorretais/genética , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , MicroRNAs/genética , Proteínas de Neoplasias/genética , Regiões 3' não Traduzidas/genética , Idoso , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/genética , Neoplasias Colorretais/metabolismo , Feminino , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/metabolismo , Estabilidade de RNA/genética
6.
Biochem Biophys Res Commun ; 493(1): 263-269, 2017 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-28893536

RESUMO

CREPT (also known as RPRD1B) function as an oncogene and is highly expressed in several kinds of cancers. However, the distribution and clinical significance of CREPT in breast cancer (BC) still not clarified. In this study, we found that the CREPT expression is greatly upregulated in BC tissues and cell lines. Moreover, the CREPT expression was significantly associated with tumor differentiation and metastasis. Next, the functional assay of CREPT showed that CREPT could promote BC proliferation and invasion both in vitro and in vivo. Dual-luciferase reporter assay indicated that miR-138 regulated the expression of CREPT by binding to its 3'-UTR. miR-138 is downregulated and inversely correlated with CREPT expression in BCs. Overexpression of miR-138 suppressed tumor growth and invasion, these effects could be reversed by re-expressing CREPT. Mechanistically, CREPT regulated ß-catenin/TCF4/cyclin D1 pathway in BC. In conclusion, the data suggested that miR-138/CREPT involved BC progression, providing potential therapeutic targets for BC.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proteínas de Ciclo Celular/metabolismo , Regulação Neoplásica da Expressão Gênica , MicroRNAs/metabolismo , Proteínas de Neoplasias/metabolismo , Animais , Proliferação de Células , Feminino , Humanos , Células MCF-7 , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Células Tumorais Cultivadas
7.
Biochem Biophys Res Commun ; 480(3): 436-442, 2016 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-27773816

RESUMO

CREPT (cell cycle-related and expression elevated protein in tumor) is highly expressed in many kinds of cancer, and has been shown to be prognostic in certain cancers. However, the clinical significance of CREPT in colorectal cancer (CRC) has not been sufficiently investigated. In this study, we examined the CREPT expression in 225 clinical CRC tissues and paired adjacent normal tissues, and analyzed the correlation between CREPT expression and other clinicopathological features. We also evaluated the biological function of CREPT both in vitro and in vivo using knockdown or overexpressing CRC cells. Our results showed that CREPT expressed in 175 of 225 (77.8%) CRC patients and the CREPT expression was significantly associated with tumor differentiation (P = 0.000), Dukes' stages (P = 0.013) and metastasis (P = 0.038). Patients with high CREPT expression tended to have shorter survival time. Multivariate analysis showed that positive CREPT expression can be used as an independent predictor for CRC prognosis. CREPT knockdown cells showed inhibited cell proliferation and arrested cell cycle, while CREPT overexpressing cells showed increased proliferation and promoted cell cycle. In addition, CREPT overexpression significantly promoted tumor growth in vivo. Mechanism study showed that CREPT may regulate cell proliferation and cell cycle through the regulation on cyclin D3, CDK4 and CDK6.


Assuntos
Biomarcadores Tumorais/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/mortalidade , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Idoso , Idoso de 80 Anos ou mais , Células CACO-2 , Proliferação de Células , Sobrevivência Celular , China/epidemiologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Células HT29 , Humanos , Pessoa de Meia-Idade , Prevalência , Prognóstico , Reprodutibilidade dos Testes , Fatores de Risco , Sensibilidade e Especificidade , Taxa de Sobrevida , Regulação para Cima
8.
J Biol Chem ; 289(33): 22589-22599, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24982424

RESUMO

CREPT (cell cycle-related and expression elevated protein in tumor)/RPRD1B (regulation of nuclear pre-mRNA domain-containing protein 1B), highly expressed during tumorigenesis, was shown to enhance transcription of CCND1 and to promote cell proliferation by interacting with RNA polymerase II. However, which signaling pathway is involved in CREPT-mediated activation of gene transcription remains unclear. In this study, we reveal that CREPT participates in transcription of the Wnt/ß-catenin signaling activated genes through the ß-catenin and the TCF4 complex. Our results demonstrate that CREPT interacts with both ß-catenin and TCF4, and enhances the association of ß-catenin with TCF4, in response to Wnt stimulation. Furthermore, CREPT was shown to occupy at TCF4 binding sites (TBS) of the promoters of Wnt-targeted genes under Wnt stimulation. Interestingly, depletion of CREPT resulted in decreased occupancy of ß-catenin on TBS, and over-expression of CREPT enhances the activity of the ß-catenin·TCF4 complex to initiate transcription of Wnt target genes, which results in up-regulated cell proliferation and invasion. Our study suggests that CREPT acts as an activator to promote transcriptional activity of the ß-catenin·TCF4 complex in response to Wnt signaling.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Neoplasias/metabolismo , Fatores de Transcrição/metabolismo , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Proteínas de Ciclo Celular/genética , Proliferação de Células/fisiologia , Células HEK293 , Células HeLa , Humanos , Camundongos , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Células NIH 3T3 , Proteínas de Neoplasias/genética , Fator de Transcrição 4 , Fatores de Transcrição/genética , Transcrição Gênica/fisiologia , Regulação para Cima/fisiologia , beta Catenina/genética
9.
Theranostics ; 14(2): 662-680, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38169511

RESUMO

Rationale: Cancer local recurrence increases the mortality of patients, and might be caused by field cancerization, a pre-malignant alteration of normal epithelial cells. It has been suggested that cancer-derived small extracellular vesicles (CDEs) may contribute to field cancerization, but the underlying mechanisms remain poorly understood. In this study, we aim to identify the key regulatory factors within recipient cells under the instigation of CDEs. Methods: In vitro experiments were performed to demonstrate that CDEs promote the expression of CREPT in normal epithelial cells. TMT-based quantitative mass spectrometry was employed to investigate the proteomic differences between normal cells and tumor cells. Loss-of-function approaches by CRISPR-Cas9 system were used to assess the role of CREPT in CDEs-induced field cancerization. RNA-seq was performed to explore the genes regulated by CREPT during field cancerization. Results: CDEs promote field cancerization by inducing the expression of CREPT in non-malignant epithelial cells through activating the ERK signaling pathway. Intriguingly, CDEs failed to induce field cancerization when CREPT was deleted, highlighting the importance of CREPT. Transcriptomic analyses revealed that CDEs elicited inflammatory responses, primarily through activation of the TNF signaling pathway. CREPT, in turn, regulates the transduction of downstream signals of TNF by modulating the expression of TNFR2 and PI3K, thereby promoting inflammation-to-cancer transition. Conclusion: CREPT not only serves as a biomarker for field cancerization, but also emerges as a target for preventing the cancer local recurrence.


Assuntos
Vesículas Extracelulares , Neoplasias , Humanos , Linhagem Celular Tumoral , Proteômica , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células/genética , Proteínas de Neoplasias/genética , Vesículas Extracelulares/metabolismo , Neoplasias/genética
10.
Cancers (Basel) ; 14(19)2022 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-36230720

RESUMO

Histone deacetylases 1 (HDAC1), an enzyme that functions to remove acetyl molecules from ε-NH3 groups of lysine in histones, eliminates the histone acetylation at the promoter regions of tumor suppressor genes to block their expression during tumorigenesis. However, it remains unclear why HDAC1 fails to impair oncogene expression. Here we report that HDAC1 is unable to occupy at the promoters of oncogenes but maintains its occupancy with the tumor suppressors due to its interaction with CREPT (cell cycle-related and expression-elevated protein in tumor, also named RPRD1B), an oncoprotein highly expressed in tumors. We observed that CREPT competed with HDAC1 for binding to oncogene (such as CCND1, CLDN1, VEGFA, PPARD and BMP4) promoters but not the tumor suppressor gene (such as p21 and p27) promoters by a chromatin immunoprecipitation (ChIP) qPCR experiment. Using immunoprecipitation experiments, we deciphered that CREPT specifically occupied at the oncogene promoter via TCF4, a transcription factor activated by Wnt signaling. In addition, we performed a real-time quantitative PCR (qRT-PCR) analysis on cells that stably over-expressed CREPT and/or HDAC1, and we propose that HDAC1 inhibits CREPT to activate oncogene expression under Wnt signaling activation. Our findings revealed that HDAC1 functions differentially on tumor suppressors and oncogenes due to its interaction with the oncoprotein CREPT.

11.
Int J Oral Maxillofac Surg ; 51(1): 27-32, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33838964

RESUMO

Cyclin D1 is the most essential progressive regulator of the cell cycle, and its transcription is enhanced by CREPT (cell cycle-related and expression-elevated protein in tumour). These molecules regulate cell growth, and their aberrant expression can cause malignant transformation. In this study, the expression of these molecules was explored to investigate the molecular alterations in oral precancerous lesions and squamous cell carcinoma. Cyclin D1 and CREPT expression was examined immunohistochemically in tissue specimens from 55 patients with oral epithelial precursor lesions (OEPLs) and 84 patients with oral squamous cell carcinoma (OSCC). Associations between the results and clinicopathological variables were examined. Cyclin D1 and CREPT expression levels were higher in OSCC than in OEPLs. Furthermore, there were statistically significant differences in cyclin D1 expression among the different grades of OEPLs and OSCC lesions. In OSCC, there were statistically significant differences in CREPT expression according to sex, T stage, and degree of differentiation. In addition, the expression of both molecules was significantly correlated with postoperative metastasis and modes of invasion. The expression of cyclin D1 and CREPT was found to depend upon the state of development and progression of the oral epithelial lesions, and clinicopathological behaviours might be affected by these molecules in OSCC.


Assuntos
Carcinoma de Células Escamosas , Neoplasias Bucais , Lesões Pré-Cancerosas , Proteínas de Ciclo Celular , Ciclina D1 , Humanos , Proteínas de Neoplasias , Prognóstico
12.
Cell Oncol (Dordr) ; 44(2): 345-355, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33125631

RESUMO

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive human malignancies. Cell-cycle-related and expression-elevated protein in tumor (CREPT) plays an important role in the phosphorylation of RNA Pol II, and has been implicated in the development of several types of cancer. As yet, however, there have been no reports on its role in PDAC. Here, we aimed to explore the value of CREPT as a prognostic biomarker in PDAC. METHODS: CREPT expression was assessed by immunohistochemistry (IHC) on a tissue microarray containing samples from 375 PDAC patients. Kaplan-Meier and Cox regression analyses were performed to explore the independent prognostic value of CREPT expression for the disease-free survival (DFS) and overall survival (OS) of PDAC patients. A Cell Counting Kit-8 (CCK8) assay was used to determine the growth rates and gemcitabine sensitivities of PDAC cells, while a Transwell assay was used to determine the migration and invasion abilities of PDAC cells. Subcutaneous xenografts were used to explore the effect of CREPT expression on tumor growth in vivo. RESULTS: We found that CREPT is highly expressed in tumor tissues and may serve as an independent prognostic biomarker for DFS and OS of PDAC patients. In vitro assays revealed that CREPT expression promotes the proliferation, migration, invasion and gemcitabine resistance of PDAC cells, and in vivo assays showed that CREPT expression knockdown led to inhibition of PDAC tumor growth. CONCLUSIONS: We conclude that high CREPT expression enhances the proliferation, migration, invasion and gemcitabine resistance of PDAC cells. In addition, we conclude that CREPT may serve as an independent prognostic biomarker and therapeutic target for PDAC patients.


Assuntos
Adenocarcinoma/metabolismo , Biomarcadores Tumorais/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/metabolismo , Adenocarcinoma/genética , Animais , Biomarcadores Tumorais/genética , Carcinoma Ductal Pancreático/genética , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Intervalo Livre de Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Análise Multivariada , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/genética , Prognóstico , Análise de Sobrevida , Regulação para Cima/genética , Neoplasias Pancreáticas
13.
Theranostics ; 10(8): 3708-3721, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32206117

RESUMO

Cancers remain a threat to human health due to the lack of effective therapeutic strategies. Great effort has been devoted to the discovery of drug targets to treat cancers, but novel oncoproteins still need to be unveiled for efficient therapy. Methods: We show that CREPT is highly expressed in pancreatic cancer and is associated with poor disease-free survival. CREPT overexpression promotes but CREPT deletion blocks colony formation and proliferation of pancreatic cancer cells. To provide a proof of concept for CREPT as a new target for the inhibition of pancreatic cancer, we designed a cell-permeable peptide-based proteolysis targeting chimera (PROTAC), named PRTC, based on the homodimerized leucine-zipper-like motif in the C-terminus domain of CREPT to induce its degradation in vivo. Results: PRTC has high affinity for CREPT, with Kd = 0.34 +/- 0.11 µM and is able to permeate into cells because of the attached membrane-transportable peptide RRRRK. PRTC effectively induces CREPT degradation in a proteasome-dependent manner. Intriguingly, PRTC inhibits colony formation, cell proliferation, and motility in pancreatic cancer cells and ultimately impairs xenograft tumor growth, comparable to the effect of CREPT deletion. Conclusions: PRTC-induced degradation of CREPT leads to inhibition of tumor growth, which is promising for the development of new drugs against pancreatic cancer. In addition, using an interacting motif based on the dimerized structure of proteins may be a new way to design a PROTAC aiming at degrading any protein without known interacting small molecules or peptides.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Peptídeos/uso terapêutico , Proteólise , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus
14.
Biochimie ; 162: 116-124, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30995540

RESUMO

Cell cycle-related and expression elevated protein in tumor (CREPT) is emerging as a novel cancer-related gene that is dysregulated in many kinds of malignancies. However, the expression and biological role of CREPT in glioma remains unclear. In the present study, we aimed to explore the potential function and regulation mechanism of CREPT in glioma. Results showed that CRETP expression was significantly upregulated in glioma cell lines. Depletion of CREPT by siRNA-mediated gene silencing markedly decreased the proliferative and invasive capabilities of glioma cells. Bioinformatics analysis predicted CREPT as a target gene of microRNA-596 (miR-596), which was further verified by real-time quantitative polymerase chain reaction and Western blot analysis. miR-596 was significantly decreased in glioma tissues and cell lines, and inversely correlated with CREPT expression in clinical specimens. Knockdown of CREPT or overexpression of miR-596 significantly restricted the activation of Wnt/ß-catenin signaling in glioma cells. Moreover, overexpression of CREPT partially reversed the miR-596-mediated inhibitory effect on proliferation, invasion and Wnt/ß-catenin signaling in glioma cells. Overall, these results demonstrate that CREPT exerts an oncogenic role in glioma and its expression is regulated by miR-596. Our study highlights the important role miR-596/CREPT/Wnt/ß-catenin signaling axis may play in glioma.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Glioma/patologia , MicroRNAs/fisiologia , Proteínas de Neoplasias/fisiologia , Via de Sinalização Wnt , beta Catenina/metabolismo , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Glioma/genética , Células HEK293 , Humanos , Invasividade Neoplásica , Proteínas de Neoplasias/genética , Oncogenes , beta Catenina/genética
15.
Chem Biol Interact ; 302: 74-82, 2019 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-30738779

RESUMO

Accumulating evidence has suggested that microRNA-449b-5p (miR-449b-5p) plays an important role in the development and progression of multiple cancers. However, little is known about the role of miR-449b-5p in breast cancer. In this study, we aimed to investigate the expression level, biological function and underlying mechanism of miR-449b-5p in breast cancer. Our results showed that miR-449b-5p expression was frequently down-regulated in breast cancer cell lines and tissues. The overexpression of miR-449b-5p significantly inhibited growth and invasion, and induced the cell cycle arrest of breast cancer cells. In contrast, the inhibition of miR-449b-5p showed the opposite effect. Interestingly, bioinformatic analysis predicted that cell cycle-related and expression-elevated protein in tumor (CREPT), an important oncogene in breast cancer, was a potential target gene of miR-449b-5p. The overexpression of miR-449b-5p decreased CREPT expression while miR-449b-5p inhibition promoted CREPT expression in breast cancer cells. Restoration of CREPT expression in miR-449b-5p mimics transfected cells partially reversed the suppressive effect of miR-449b-5p on breast cancer cell growth and invasion. Notably, our results showed that miR-449b-5p overexpression decreased the expression of ß-catenin and suppressed the activation of Wnt/ß-catenin/TCF-4 signaling via targeting CREPT. In addition, blocking Wnt/ß-catenin partially reversed the promotion effect of miR-449b-5p inhibition on breast cancer cell growth and invasion. Overall, these results reveal a tumor suppressive role of miR-449b-5p that restricts the growth and invasion of breast cancer cells through targeting CREPT and inhibiting CREPT-mediated activation of Wnt/ß-catenin signaling. Our study suggests that the miR-449b-5p/CREPT/Wnt/ß-catenin axis may play an important role in the pathogenesis of breast cancer and miR-449-5p may serve as a potential therapeutic target for breast cancer.


Assuntos
Neoplasias da Mama/patologia , Proteínas de Ciclo Celular/metabolismo , MicroRNAs/metabolismo , Proteínas de Neoplasias/metabolismo , Via de Sinalização Wnt , Regiões 3' não Traduzidas , Antagomirs/metabolismo , Sítios de Ligação , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Humanos , Células MCF-7 , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Fator de Transcrição 4/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
16.
Cancers (Basel) ; 12(1)2019 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-31877646

RESUMO

Melanoma is one of the most aggressive cancers, and patients with distant metastases have dire outcomes. We observed previously that melanoma progression is driven by a high migratory potential of melanoma cells, which survive and proliferate under harsh environmental conditions. In this study, we report that CREPT (cell-cycle related and expression-elevated protein in tumor), an oncoprotein highly expressed in other cancers, is overexpressed in melanoma cells but not melanocytes. Overexpression of CREPT stimulates cell proliferation, migration, and invasion in several melanoma cell lines. Further, we show that CREPT enhances melanoma progression through upregulating and activating Ras homolog family member A (RhoA)-induced actin organization and focal adhesion assembly. Our study reveals a novel role of CREPT in promoting melanoma progression. Targeting CREPT may be a promising strategy for melanoma treatment.

17.
Int J Clin Exp Pathol ; 12(9): 3301-3310, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31934173

RESUMO

CREPT has been shown to be highly expressed in most tumors and is associated with a poor prognosis, but the histologic characteristics of CREPT expression and its impact on clinical outcomes in esophageal squamous cell carcinoma (ESCC) are unclear. Therefore, we retroactively evaluated tissue microarrays (TMA) from 300 surgical cases, including 300 ESCC tissues and 161 adjacent non-tumor tissues, and pretreatment tumor biopsies from 113 concurrent chemoradiotherapy (CCRT) cases by immunohistochemistry (IHC). Notably, CREPT was increasingly expressed from non-cancerous tissues to atypical hyperplasia to tumor tissues (P < 0.01). Furthermore, patients were divided into low CREPT (≤ 8 scores) and high CREPT (> 8 scores) groups. Patients with high CREPT expressions had a worse overall survival (OS) (5-year OS: 40.9% vs. 50.1%, P=0.040) and disease-free survival (DFS) (5-year DFS: 29.5 vs. 43.0%; P=0.020) than those with low expressions. Nevertheless, only in the high CREPT subgroup did adjuvant therapy (AT) prolong the OS (5-year OS: 53.8 vs. 28.9%; P=0.020), especially for adjuvant radiotherapy (ART) (5-year OS: 85.7 vs. 28.9%; P=0.037; 5-year DFS: 85.7 vs. 22.3%; P=0.020). Surprisingly, high CREPT expressions endowed CCRT-treated patients with higher complete response rates (50% vs. 26%; P=0.018) and a favorable OS (3-year OS: 54.3 vs. 28.1%; P=0.046) compared to low expression. Overall, our findings indicate that CREPT is highly expressed in ESCC tissue compared with non-cancerous tissue and this feature is associated with a poor prognosis. Otherwise, patients with high CREPT expression were more sensitive to AT and CCRT. Moreover, CREPT could be a predictive immunohistochemical biomarker used to guide individualized clinical treatment.

18.
Journal of Medical Research ; (12): 164-169, 2023.
Artigo em Chinês | WPRIM | ID: wpr-1023559

RESUMO

Objective To detect the expression difference of dynactin 2(DCTN2)and cell-cycle-related and expression-elevat-ed protein in tumor(CREPT)in gastric cancer and paracancer tissues,and to explore the relationship between DCTN2 and CREPT ex-pressions and the clinicopathological characteristics and prognosis of gastric cancer patients.Methods A total of 90 patients who received gastric cancer surgery from March 2014 to September 2015 in Affiliated Hospital of Xuzhou Medical University and were confirmed by his-tology were enrolled.The expression levels of DCTN2 and CREPT in gastric cancer tissues and paracancer tissues were detected by immu-nohistochemistry,and the Spearman correlation analysis was used to analyze the relationship between DCTN2 and CREPT in gastric cancer tissues;the relationship between their levels and clinicopathological characteristics were analyzed;Kaplan-Meier and Log-rank analysis were used to analyze the relationship between the expression levels of DCTN2 and CREPT and the overall survival time of gastric cancer patients;univariate and multivariate COX regression analysis were used to analyze the independent risk factors influencing prognosis of gastric cancer patients.Results The expression levels of DCTN2 and CREPT in gastric cancer tissues were significantly higher than that in paracancer tissues,and DCTN2 was positively correlated with CREPT in gastric cancer tissues(P<0.05).The expression levels of DCTN2 and CREPT were correlated with the tumor differentiation,TNM stage,invasion depth,lymph node metastasis(P<0.05),but were not significantly correlated with gender,age and distant metastasis(P>0.05);the results of prognosis analysis showed that the 1-year,3-year and 5-year cumulative survival rate and overall survival rate of gastric cancer patients in the DCTN2high-expression group were significantly lower than those in the DCTN2 low-expression group,and the 5-year cumulative survival rate and overall sur-vival rate of gastric cancer patients in the CREPT high-expression group were significantly lower than those in the CREPT low-expres-sion group(P<0.05).Univariate and multivariate COX regression analysis showed that both DCTN2 and CREPT were independent risk factors on the prognosis of gastric cancer patients.Conclusion DCTN2 and CREPT are highly expressed in gastric cancer tissues and their high expression were closely related to the poor prognosis of gastric cancer patients.DCTN2 and CREPT were expected to be potential markers and new therapeutic targets for gastric cancer diagnosis,and help to judge the prognosis of gastric cancer patients.

19.
World J Gastroenterol ; 24(4): 475-483, 2018 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-29398868

RESUMO

AIM: To investigate expression of cell cycle-related and expression-elevated protein in tumor (CREPT) in colorectal cancer (CRC) and determine its prognostic value in response to 5-fluorouracil (5-FU). METHODS: The relative expression of CREPT in CRC tumor samples was determined using immunohistochemistry. The protein content in cell lines was analyzed by immunoblotting. Cell viability was measured with the CCK-8 assay. Cell cycle and apoptosis analyses were performed with flow cytometry. RESULTS: CREPT was overexpressed in CRC tissues and correlated with histological grade. Clinicopathological analysis indicated that CREPT was positively related to tumor progression. Exogenous expression of CREPT stimulated cell proliferation and accelerated the cell cycle. More importantly, high expression of CREPT sensitized CRC cells to 5-FU treatment. Furthermore, we demonstrated that 5-FU elicited significant apoptosis in CREPT-positive cells. CONCLUSION: Aberrant overexpression of CREPT contributes to tumorigenesis of CRC by promoting cell proliferation and accelerating the cell cycle, and confers sensitivity to 5-FU. CREPT is a potential prognostic biomarker for 5-FU in CRC.


Assuntos
Adenoma/patologia , Antimetabólitos Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/metabolismo , Proteínas de Ciclo Celular/metabolismo , Neoplasias Colorretais/patologia , Fluoruracila/uso terapêutico , Proteínas de Neoplasias/metabolismo , Adenoma/tratamento farmacológico , Adenoma/mortalidade , Antimetabólitos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinogênese , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/mortalidade , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Citometria de Fluxo , Fluoruracila/farmacologia , Humanos , Gradação de Tumores , Prognóstico , Análise de Sobrevida , Regulação para Cima
20.
Am J Transl Res ; 8(5): 2097-113, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27347318

RESUMO

It has been reported that CREPT acts as a highly expressed oncogene in a variety of tumors, affecting cyclin D1 signal pathways. However, the distribution and clinical significance of CREPT in NSCLC remains poorly understood. Our study focused on the role of CREPT on the regulation ofnon-small cell lung cancer (NSCLC). We found that CREPT mRNA and protein expression was significantly increased in NSCLC compared with adjacent lung tissues and was increased in various NSCLC cell lines compared with the normal human bronchial epithelial (HBE) cell line. siRNA-induced knockingdown of CREPT significantly inhibited the proliferation and migration of NSCLC cell lines by arresting cell cycle in S phase. Moreover, CREPT knocking down affected the expression of cell cycle proteins including c-mycand CDC25A. Finally, we found there were obvious correlations between CREPT with c-myc expression in histological type, differentiation, and pTNM stages of NSCLC (P<0.05, rs>0.3). Immunohistofluorescence studies demonstrated a co-localization phenomenon when CREPT and c-myc were expressed. Thus, we propose that CREPT may promote NSCLC cell growth and migration through the c-myc and CDC25A signaling molecules.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa