Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Hong Kong Med J ; 29(4): 287-294, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37409372

RESUMO

INTRODUCTION: This study investigated the awareness, perceptions, and acceptance of human papillomavirus (HPV) vaccination for children among parents in Hong Kong. It also explored factors associated with, and differences in, vaccine acceptance and hesitancy between parents of girls and boys. METHODS: Parents of boys or girls in Primary 5 to 6 were invited to participate in an online survey through an established health and lifestyle e-platform. RESULTS: Overall, 851 parents completed the survey: 419 (49.2%) had daughters, 348 (40.9%) had sons, and 84 (9.9%) had children of both genders. Parents who enrolled their children into the Childhood Immunisation Programme were more likely to accept HPV vaccination (79.7% vs 33.7%, odds ratio [OR]=7.70; 95% confidence interval [CI]=5.39-11.01; P<0.001); parents of girls were more likely to accept than parents of boys (86.0% vs 71.8%, OR=2.40; 95% CI=1.67-3.46; P<0.001). Among parents of girls and boys, the main reasons for HPV vaccination acceptance were prevention of cancers (girls: 68.8% and boys: 68.7%), prevention of sexually transmitted diseases (girls: 67.3% and boys: 68.3%), and optimal timing before initiation of sexual activity (girls: 62.8% and boys: 59.8%). Vaccine hesitancy was mainly associated with concerns about serious side-effects (girls: 66.7% and boys: 68.0%) and the belief that their children were too young (girls: 60.0% and boys: 54.0%). CONCLUSION: Parents in Hong Kong are hesitant about HPV vaccination for their sons. This barrier could be removed by providing information to correct vaccine safety misconceptions and offering a gender-neutral vaccination programme through the school-based Childhood Immunisation Programme.


Assuntos
Infecções por Papillomavirus , Vacinas contra Papillomavirus , Criança , Humanos , Masculino , Feminino , Hong Kong , Infecções por Papillomavirus/prevenção & controle , Papillomavirus Humano , Aceitação pelo Paciente de Cuidados de Saúde , Conhecimentos, Atitudes e Prática em Saúde , Pais , Vacinação
2.
Med Res Rev ; 37(6): 1318-1349, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28586517

RESUMO

Numerous studies have proved that cell-nonautonomous regulation of neoplastic cells is a distinctive and essential characteristic of tumorigenesis. Two way communications between the tumor and the stroma, or within the tumor significantly influence disease progression and modify treatment responses. In the tumor microenvironment (TME), malignant cells utilize paracrine signaling initiated by adjacent stromal cells to acquire resistance against multiple types of anticancer therapies, wherein extracellular vesicles (EVs) substantially promote such events. EVs are nanoscaled particles enclosed by phospholipid bilayers, and can mediate intercellular communications between cancerous cells and the adjacent microenvironment to accelerate pathological proceeding. Here we review the most recent studies of EV biology and focus on key cell lineages of the TME and their EV cargoes that are biologically active and responsible for cancer resistance, including proteins, RNAs, and other potentially essential components. Since EVs are emerging as novel but critical elements in establishing and maintaining hallmarks of human cancer, timely and insightful understanding of their molecular properties and functional mechanisms would pave the road for clinical diagnosis, prognosis, and effective targeting in the global landscape of precision medicine. Further, we address the potential of EVs as promising biomarkers in cancer clinics and summarize the technical improvements in EV preparation, analysis, and imaging. We highlight the practical issues that should be exercised with caution to guide the development of targeting agents and therapeutic methodologies to minimize cancer resistance driven by EVs, thereby allowing to effectively control the early steps of disease exacerbation.


Assuntos
Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/patologia , Neoplasias/metabolismo , Neoplasias/terapia , Animais , Biomarcadores Tumorais/metabolismo , Humanos , Neoplasias/patologia , Microambiente Tumoral
3.
Br J Cancer ; 115(10): 1223-1233, 2016 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-27685445

RESUMO

BACKGROUND: Cetuximab, a monoclonal antibody against EGFR used for the treatment of colorectal cancer (CRC), is ineffective in many patients. The aim of this study was to identify the signalling pathways activated by cetuximab in CRC cells and define new biomarker of response. METHODS: We used in vitro, in vivo models and clinical CRC samples to assess the role of p38 and FOXO3a in cetuximab mechanism of action. RESULTS: We show that cetuximab activates the MAPK p38. Specifically, p38 inhibition reduced cetuximab efficacy on cell growth and cell death. At the molecular level, cetuximab activates the transcription factor FOXO3a and promotes its nuclear translocation via p38-mediated phosphorylation, leading to the upregulation of its target genes p27 and BIM and the subsequent induction of apoptosis and inhibition of cell proliferation. Finally, we found that high FOXO3a and p38 expression levels are associated with better response rate and improved outcome in cetuximab-treated patients with CRC harbouring WT KRAS. CONCLUSIONS: We identify FOXO3a as a key mediator of cetuximab mechanism of action in CRC cells and define p38 as its activator in this context. Moreover, high FOXO3a and p38 expression could predict the response to cetuximab in patients with CRC harbouring WT KRAS.


Assuntos
Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cetuximab/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Proteína Forkhead Box O3/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Células CACO-2 , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Nus , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Proteínas ras/metabolismo
4.
Ann Oncol ; 24(4): 924-30, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23233650

RESUMO

BACKGROUND: This phase II, open-label, multicentre study aimed to evaluate changes in cell proliferation and biomarkers, as well as efficacy of lapatinib in treatment-naïve patients with HER-2-negative primary breast cancer. PATIENTS AND METHODS: Patients received 1500 mg lapatinib for 28-42 days before surgery with repeat biopsies and measurements. The primary end point was inhibition of cell proliferation measured by Ki67; the secondary end points included clinical response, adverse events and changes in FOXO3a, FOXM1, p-AKT and HER-3. RESULTS: Overall, there was no significant reduction in Ki67 with treatment (assessment carried out in 28 of 31 subjects enrolled). However, four patients (14%) showed a reduction in Ki67 ≥50%. Four of 25 patients (16%) had a partial response to treatment judged by sequential ultrasound measurements. Response, in terms of either Ki67 or ultrasound, did not relate to changes in any biomarker assessed at baseline, including the estrogen receptor (ER) and epidermal growth factor receptor (EGFR). However, all four clinical responders were HER-3 positive, as were three of four Ki67 responders. CONCLUSIONS: Overall, a pre-surgical course of lapatinib monotherapy had little effect on this group of patients; however, in subsets of patients, especially those with HER-3-positive tumors, we observed either reduction in proliferation (Ki67) or tumor size; EGFR/ER status had no impact.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Quinazolinas/administração & dosagem , Adulto , Idoso , Biópsia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Receptores ErbB/metabolismo , Feminino , Proteína Forkhead Box M1 , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/metabolismo , Humanos , Antígeno Ki-67/metabolismo , Lapatinib , Pessoa de Meia-Idade , Proteína Oncogênica v-akt/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-3/metabolismo , Receptores de Estrogênio/metabolismo
5.
Curr Opin Cell Biol ; 6(6): 859-66, 1994 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7880534

RESUMO

Transcriptional control during the G1/S transition is important in regulating cell cycle progression. The transcription factor DRTF1/E2F is believed to play a crucial role in this process by integrating the activity of the machinery that drives the cell cycle with the transcription apparatus. Being the point of convergence for growth-promoting and growth-inhibitory signals, it is a pivotal cellular target for molecules which subvert normal cell cycle control, such as oncoviral proteins. Recent studies have indicated that members of two distinct families of proteins, DP and E2F, interact combinatorially as DP/E2F heterodimers in DRTF1/E2F. The activities of both DP and E2F proteins are under cell cycle control, being influenced by the level of phosphorylation imparted through the cell cycle regulated activity of cyclin-dependent kinases. Both DP and E2F proteins are endowed with proto-oncogenic activity and, conversely, have been implicated in regulating apoptosis. Current evidence suggests therefore that the activity of DRTF1/E2F is instrumental in regulating progression through the cell cycle.


Assuntos
Proteínas de Transporte , Proteínas de Ciclo Celular , Ciclo Celular/fisiologia , Proteínas de Ligação a DNA , Fatores de Transcrição/fisiologia , Transcrição Gênica/fisiologia , Animais , Apoptose , Ciclo Celular/genética , Divisão Celular/fisiologia , Sobrevivência Celular , Fatores de Transcrição E2F , Homeostase/fisiologia , Humanos , Neoplasias/fisiopatologia , Proteína 1 de Ligação ao Retinoblastoma , Fator de Transcrição DP1
6.
Oncogene ; 38(25): 5111-5112, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30867566

RESUMO

In the published version of this article, the images for cytoplasmic and nuclear FGF7 in MDA-MB-231 cells were duplicated and mistaken for total FGF7 in SKBR-3 and MDA-MB-231 cells.

7.
Leukemia ; 21(2): 304-10, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17170725

RESUMO

Mesenchymal stem cells (MSC) have received much attention in the field of hematopoietic stem cell transplantation because not only do they support hematopoiesis but also exhibit a profound immunosuppressive activity that can be exploited to prevent undesired alloreactivity. We have previously shown that their immunosuppressive activity is mainly exerted at the level of T-cell proliferation. Here, we show that MSC exhibit a similar antiproliferative activity on tumor cells of hematopoietic and non hematopoietic origin. In vitro, MSC produced the transient arrest of tumor cells in the G(1) phase of cell cycle; this was accompanied by a reduction in the apoptotic rate even when survival factors were limiting. However, when tumor cells were injected into non-obese diabetic-severe combined immunodeficient mice in conjunction with MSC, their growth was much faster as compared to the group receiving only tumor cells. To explain the discrepancy between the in vitro and in vivo behavior, we suggest that MSC have the ability to form a cancer stem cell niche in which tumor cells can preserve the potential to proliferate and sustain the malignant process. We conclude that the clinical use of MSC in conditions in which a malignant disease is involved should be handled with extreme caution.


Assuntos
Apoptose/fisiologia , Divisão Celular/fisiologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Neoplasias/patologia , Adulto , Animais , Autopsia , Linhagem Celular Tumoral , Sobrevivência Celular , Fase G1 , Humanos , Imunofenotipagem , Células Jurkat , Células K562 , Fígado/patologia , Linfonodos/patologia , Camundongos , Camundongos SCID , Pessoa de Meia-Idade , Baço/patologia , Transplante Heterólogo
8.
Oncogene ; 37(8): 1062-1074, 2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29106390

RESUMO

Glycolysis is critical for cancer stem cell reprogramming; however, the underlying regulatory mechanisms remain elusive. Here, we show that pyruvate dehydrogenase kinase 1 (PDK1) is enriched in breast cancer stem cells (BCSCs), whereas depletion of PDK1 remarkably diminishes ALDH+ subpopulations, decreases stemness-related transcriptional factor expression, and inhibits sphere-formation ability and tumor growth. Conversely, high levels of PDK1 enhance BCSC properties and are correlated with poor overall survival. In mouse xenograft tumor, PDK1 is accumulated in hypoxic regions and activates glycolysis to promote stem-like traits. Moreover, through screening hypoxia-related long non-coding RNAs (lncRNAs) in PDK1-positive tissue, we find that lncRNA H19 is responsible for glycolysis and BCSC maintenance. Furthermore, H19 knockdown decreases PDK1 expression in hypoxia, and ablation of PDK1 counteracts H19-mediated glycolysis and self-renewal ability in vitro and in vivo. Accordingly, H19 and PDK1 expression exhibits strong correlations in primary breast carcinomas. H19 acting as a competitive endogenous RNA sequesters miRNA let-7 to release Hypoxia-inducible factor 1α, leading to an increase in PDK1 expression. Lastly, aspirin markedly attenuates glycolysis and cancer stem-like characteristics by suppressing both H19 and PDK1. Thus, these novel findings demonstrate that the glycolysis gatekeeper PDK1 has a critical role in BCSC reprogramming and provides a potential therapeutic strategy for breast malignancy.


Assuntos
Neoplasias da Mama/patologia , Regulação Neoplásica da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia/fisiopatologia , Células-Tronco Neoplásicas/patologia , Proteínas Serina-Treonina Quinases/metabolismo , RNA Longo não Codificante/genética , Animais , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proliferação de Células , Feminino , Glicólise , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células-Tronco Neoplásicas/metabolismo , Prognóstico , Proteínas Serina-Treonina Quinases/genética , Piruvato Desidrogenase Quinase de Transferência de Acetil , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Oncogene ; 37(8): 1119, 2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29251717

RESUMO

This corrects the article DOI: 10.1038/onc.2017.368.

10.
Oncogene ; 25(15): 2170-80, 2006 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-16301994

RESUMO

Lymphocyte proliferation is key to the regulation of the immune system. Cyclin D2 is the first cell cycle protein induced following stimulation through the T-cell receptor, the B-cell receptor or cytokines. The promoter of this cyclin integrates a diverse range of signals. Through investigating the regulation of this promoter by interleukin-2 and phosphatidylinositol 3-kinase, we have identified a role for the transcription factor CREB, cAMP response element-binding protein. Mutation of the CREB-binding site reduced cyclin D2 promoter activity 5-10-fold. CREB-1 is phosphorylated at serine 133, a critical site for activity, in both T cells and Epstein-Barr virus immortalized B cells. The introduction of an S133A mutant of CREB-1 reduces IL-2 induction of cyclin D2 promoter activity, demonstrating a role for this phosphorylation site in promoter activity. Two inhibitors of protein kinase A reduce lymphocyte proliferation and CREB-1 phosphorylation. This study demonstrates that the cyclin D2 promoter is capable of being regulated by PI3K and CREB and identifies CREB-1 and protein kinase A as potential targets for altering lymphocyte proliferation.


Assuntos
Linfócitos B/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/farmacologia , Ciclinas/metabolismo , Regiões Promotoras Genéticas , Linfócitos T/efeitos dos fármacos , Linfócitos B/metabolismo , Linfócitos B/virologia , Western Blotting , Carbazóis/farmacologia , Proliferação de Células/efeitos dos fármacos , Transformação Celular Viral , Células Cultivadas , Ciclina D2 , Ciclinas/genética , Ensaio de Desvio de Mobilidade Eletroforética , Inibidores Enzimáticos/farmacologia , Humanos , Indóis/farmacologia , Interleucina-2/metabolismo , Isoquinolinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Pirróis/farmacologia , Sulfonamidas/farmacologia , Linfócitos T/metabolismo , Transcrição Gênica
11.
Mol Cell Biol ; 20(24): 9138-48, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11094066

RESUMO

Interleukin-3 (IL-3), IL-5, and granulocyte-macrophage colony-stimulating factor regulate the survival, proliferation, and differentiation of hematopoietic lineages. Phosphatidylinositol 3-kinase (PI3K) has been implicated in the regulation of these processes. Here we investigate the molecular mechanism by which PI3K regulates cytokine-mediated proliferation and survival in the murine pre-B-cell line Ba/F3. IL-3 was found to repress the expression of the cyclin-dependent kinase inhibitor p27(KIP1) through activation of PI3K, and this occurs at the level of transcription. This transcriptional regulation occurs through modulation of the forkhead transcription factor FKHR-L1, and IL-3 inhibited FKHR-L1 activity in a PI3K-dependent manner. We have generated Ba/F3 cell lines expressing a tamoxifen-inducible active FKHR-L1 mutant [FKHR-L1(A3):ER*]. Tamoxifen-mediated activation of FKHR-L1(A3):ER* resulted in a striking increase in p27(KIP1) promoter activity and mRNA and protein levels as well as induction of the apoptotic program. The level of p27(KIP1) appears to be critical in the regulation of cell survival since mere ectopic expression of p27(KIP1) was sufficient to induce Ba/F3 apoptosis. Moreover, cell survival was increased in cytokine-starved bone marrow-derived stem cells from p27(KIP1) null-mutant mice compared to that in cells from wild-type mice. Taken together, these observations indicate that inhibition of p27(KIP1) transcription through PI3K-induced FKHR-L1 phosphorylation provides a novel mechanism of regulating cytokine-mediated survival and proliferation.


Assuntos
Proteínas de Ciclo Celular , Citocinas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Inibidores Enzimáticos/metabolismo , Interleucina-3/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica/genética , Proteínas Supressoras de Tumor , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Linfócitos B/citologia , Linfócitos B/metabolismo , Linhagem Celular , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/antagonistas & inibidores , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Inibidores Enzimáticos/farmacologia , Eosinófilos/efeitos dos fármacos , Eosinófilos/metabolismo , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead , Regulação da Expressão Gênica , Genes Reporter/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Hidroxitestosteronas/farmacologia , Interleucina-3/genética , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação , PTEN Fosfo-Hidrolase , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , RNA/metabolismo , Transdução de Sinais , Sirolimo/farmacologia , Tamoxifeno/farmacologia , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética
12.
Mol Biol Cell ; 5(10): 1081-92, 1994 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7865876

RESUMO

It is widely believed that in mammalian cells the cellular transcription factor (DRTF1/E2F integrates cell-cycle events with the transcription apparatus by interacting with important regulators of the cell cycle, such as the retinoblastoma gene product (pRb) and related proteins, cyclins, and cyclin-dependent kinases. Here, we have defined DRTF1/E2F in Xenopus laevis that, like its mammalian counterpart, specifically binds to the E2F site, is regulated during development, and interacts with pRb and related proteins. We have isolated cDNAs that encode the functional homologue of mammalian DP-1, X1 DP-1, together with a close relative, X1 DP-2. X1 DP-1, which is highly conserved with murine DP-1, is a major DNA binding component of X1 DRTF1/E2F. Both DP-1 and DP-2 synergistically interact with members of the E2F family of proteins, E2F-1, E2F-2, and E2F-3, to generate DNA binding complexes that specifically recognize the E2F site and functionally interact with E2F-1 in E2F site-dependent transcriptional activation of cellular genes. DP-1 and DP-2 encode maternally stored transcripts that are expressed during early development. In the adult however, the expression of DP-1 and DP-2 is tissue restricted. This study therefore defines a new family of transcription factors, the DP proteins, members of which can interact combinatorially with E2F proteins to generate an array of DNA binding complexes that integrate cell-cycle progression with the transcription apparatus through the E2F binding site. The tissue-specific expression of DP family members suggests that the combination of DP/E2F heterodimers that constitute DRTF1/E2F is influenced by the phenotype of the cell.


Assuntos
Proteínas de Transporte , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila , Transativadores/genética , Xenopus laevis/genética , Sequência de Aminoácidos , Animais , Sequência Conservada , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Fator de Transcrição E2F3 , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Dados de Sequência Molecular , Mutação , Conformação Proteica , Proteína 1 de Ligação ao Retinoblastoma , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Distribuição Tecidual , Transativadores/química , Transativadores/metabolismo , Fator de Transcrição DP1 , Fatores de Transcrição/química , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional , Proteínas de Xenopus , Xenopus laevis/crescimento & desenvolvimento , Xenopus laevis/metabolismo
13.
Oncogenesis ; 6(2): e298, 2017 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-28218735

RESUMO

Aurora kinase A (AURKA) has been implicated in the regulation of cell cycle progression, mitosis and a key number of oncogenic signaling pathways in various malignancies. However, little is known about its role in gastric cancer prognosis and genotoxic resistance. Here we found that AURKA was highly overexpressed in gastric cancer and inversely correlated with disease prognosis. Overexpression of AURKA exacerbated gastric cancer drug resistance through upregulating the expression of the anti-apoptotic protein Survivin. Conversely, we demonstrated that AURKA depletion caused a decrease in Survivin protein levels by increasing its ubiquitylation and degradation. Mass spectrometric analysis revealed that upon AURKA depletion, Survivin bound to the FBXL7 E3 ubiquitin ligase, which induced ubiquitin-proteasome degradation of Survivin. In addition, we showed that AURKA regulated FBXL7 both at the levels of transcription and translation. Moreover, proteomic analysis of nuclear AURKA-interacting proteins identified Forkhead box protein P1 (FOXP1). We next showed that AURKA was required for FBXL7 transcription and that AURKA negatively regulated FOXP1-mediated FBXL7 expression. The physiological relevance of the regulation of Survivin by AURKA through the FOXP1-FBXL7 axis was further underscored by the significant positive correlations between AURKA and Survivin expression in gastric cancer patient samples. Moreover, the AURKA depletion or kinase inhibition-induced apoptotic cell death could be reversed by Survivin ectopic overexpression, further supporting that AURKA regulated Survivin to enhance drug resistance. In agreement, inhibition of AURKA synergistically enhanced the cytotoxic effect of DNA-damaging agents in cancer cells by suppressing Survivin expression. Taken together, our data suggest that AURKA restricts Survivin ubiquitylation and degradation in gastric cancer to promote drug resistance and hence the AURKA-Survivin axis can be targeted to promote the efficacy of DNA-damaging agents in gastric cancer.

14.
Oncogene ; 36(24): 3428-3440, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28114286

RESUMO

Substantial evidence suggests that breast cancer initiation, recurrence and drug resistance is supported by breast cancer stem cells (BCSCs). Recently, we reported a novel role of Aurora kinase A (AURKA) in BCSCs, as a transactivating co-factor in the induction of the c-Myc oncoprotein. However, the mode of action and transcriptional network of nuclear AURKA in BCSCs remain unknown. Here, we report that nuclear AURKA can be recruited by Forkhead box subclass M1 (FOXM1) as a co-factor to transactivate FOXM1 target genes in a kinase-independent manner. In addition, we show that AURKA and FOXM1 participate in a tightly coupled positive feedback loop to enhance BCSC phenotype. Indeed, kinase-dead AURKA can effectively transactivate the FOXM1 promoter through a Forkhead response element, whereas FOXM1 can activate AURKA expression at the transcriptional level in a similar manner. Consistently, breast cancer patient samples portrayed a strong and significant correlation between the expression levels of FOXM1 and AURKA. Moreover, both FOXM1 and AURKA were essential for maintaining the BCSC population. Finally, we demonstrated that the AURKA inhibitor AKI603 and FOXM1 inhibitor thiostrepton acted synergistically to inhibit cytoplasmic AURKA activity and disrupt the nuclear AURKA/FOXM1-positive feedback loop, respectively, resulting in a more effective inhibition of the tumorigenicity and self-renewal ability of BCSCs. Collectively, our study uncovers a previously unknown tightly coupled positive feedback signalling loop between AURKA and FOXM1, crucial for BCSC self-renewal. Remarkably, our data reveal a novel potential therapeutic strategy for targeting both the cytoplasmic and nuclear AURKA function to effectively eliminate BCSCs, so as to overcome both breast cancer and drug resistance.


Assuntos
Aurora Quinase A/genética , Neoplasias da Mama/patologia , Núcleo Celular/metabolismo , Resistencia a Medicamentos Antineoplásicos , Proteína Forkhead Box M1/genética , Células-Tronco Neoplásicas/patologia , Animais , Aurora Quinase A/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Retroalimentação Fisiológica , Feminino , Proteína Forkhead Box M1/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Camundongos , Células-Tronco Neoplásicas/metabolismo , Regiões Promotoras Genéticas
15.
Leukemia ; 31(12): 2543-2551, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28400619

RESUMO

Long non-coding RNAs (lncRNAs) play a pivotal role in tumorigenesis, exemplified by the recent finding that lncRNA maternally expressed gene 3 (MEG3) inhibits tumor growth in a p53-dependent manner. Acute myeloid leukemia (AML) is the most common malignant myeloid disorder in adults, and TP53 mutations or loss are frequently detected in patients with therapy-related AML or AML with complex karyotype. Here, we reveal that MEG3 is significantly downregulated in AML and suppresses leukemogenesis not only in a p53-dependent, but also a p53-independent manner. In addition, MEG3 is proven to be transcriptionally activated by Wilms' tumor 1 (WT1), dysregulation of which by epigenetic silencing or mutations is causally involved in AML. Therefore MEG3 is identified as a novel target of the WT1 molecule. Ten-eleven translocation-2 (TET2) mutations frequently occur in AML and significantly promote leukemogenesis of this disorder. In our study, TET2, acting as a cofactor of WT1, increases MEG3 expression. Taken together, our work demonstrates that TET2 dysregulated WT1-MEG3 axis significantly promotes AML leukemogenesis, paving a new avenue for diagnosis and treatment of AML patients.


Assuntos
Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , RNA Longo não Codificante/genética , Transdução de Sinais , Proteínas WT1/metabolismo , Apoptose/genética , Proliferação de Células , Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Dioxigenases , Regulação Leucêmica da Expressão Gênica , Humanos , Íntrons , Leucemia Mieloide Aguda/mortalidade , Leucemia Mieloide Aguda/patologia , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo , Transcrição Gênica
16.
Oncogene ; 36(3): 304-317, 2017 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-27345399

RESUMO

Aberrant p62 overexpression has been implicated in breast cancer development. Here, we found that p62 expression was elevated in breast cancer stem cells (BCSCs), including CD44+CD24- fractions, mammospheres, ALDH1+ populations and side population cells. Indeed, short-hairpin RNA (shRNA)-mediated knockdown of p62 impaired breast cancer cells from self-renewing under anchorage-independent conditions, whereas ectopic overexpression of p62 enhanced the self-renewal ability of breast cancer cells in vitro. Genetic depletion of p62 robustly inhibited tumor-initiating frequencies, as well as growth rates of BCSC-derived tumor xenografts in immunodeficient mice. Consistently, immunohistochemical analysis of clinical breast tumor tissues showed that high p62 expression levels were linked to poorer clinical outcome. Further gene expression profiling analysis revealed that p62 was positively correlated with MYC expression level, which mediated the function of p62 in promoting breast cancer stem-like properties. MYC mRNA level was reduced upon p62 deletion by siRNA and increased with p62 overexpression in breast cancer cells, suggesting that p62 positively regulated MYC mRNA. Interestingly, p62 did not transactivate MYC promoter. Instead, p62 delayed the degradation of MYC mRNA by repressing the expression of let-7a and let-7b, thus promoting MYC mRNA stabilization at the post-transcriptional level. Consistently, let-7a and let-7b mimics attenuated p62-mediated MYC mRNA stabilization. Together, these findings unveiled a previously unappreciated role of p62 in the regulation of BCSCs, assigning p62 as a promising therapeutic target for breast cancer treatments.


Assuntos
Neoplasias da Mama/patologia , MicroRNAs/genética , Células-Tronco Neoplásicas/patologia , Proteínas Proto-Oncogênicas c-myc/genética , RNA Mensageiro/genética , Proteína Sequestossoma-1/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Camundongos , Pessoa de Meia-Idade , Transplante de Neoplasias , Células-Tronco Neoplásicas/metabolismo , Prognóstico , Estabilidade de RNA , RNA Mensageiro/química , Proteína Sequestossoma-1/genética , Regulação para Cima
17.
Cancer Res ; 57(24): 5550-6, 1997 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-9407966

RESUMO

As a first step in evaluating the tumor suppressor activity of the manganese superoxide dismutase (MnSOD) gene on established tumors in vivo, we used adenovirus-mediated gene transfer as a means of delivering the MnSOD cDNA to hamster cheek pouch carcinoma (HCPC-1) cells in vitro. HCPC-1 cells were transduced with the adenovirus-MnSOD construct (AdMnSOD) at multiplicities of infectivity (MOI) of 0, 10, 25, 50, 100, 150, and 200 MOI or with the adenovirus-LacZ reporter gene construct (AdLacZ) at 100 MOI. Dose-dependent increases in MnSOD immunoreactivity were seen on Western blotting and indirect immunofluorescence microscopy with increasing AdMnSOD titers. Maximal immunoreactivity was observed at 100 MOI AdMnSOD with both techniques. Moreover, we observed a concomitant 6-7-fold increase in MnSOD activity compared with parental cell levels that also peaked at 100 MOI AdMnSOD. To determine the effect of transgene-expressed MnSOD on tumor cell behavior, we examined cell growth, plating efficiency, and anchorage-dependent growth in soft agar. Cell number measured on day 13 decreased approximately 50% with 100 MOI AdMnSOD (P < 0.05) compared with parental cells. Moreover, cell doubling time increased from 38 to 44 h with 100 MOI AdMnSOD. Plating efficiency and cell growth in soft agar decreased approximately two-thirds with 100 MOI AdMnSOD (P < 0.001). These assays of the transformed phenotype in vitro all appeared to show maximal effect with 100 MOI AdMnSOD. As tumor growth in vivo is most predictable by a combination of these in vitro data, our results suggest that if MnSOD can be effectively delivered to a tumor in vivo using the adenovirus paradigm, effective tumor growth suppression can be observed.


Assuntos
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/terapia , Superóxido Dismutase/genética , Adenoviridae/genética , Animais , Carcinoma de Células Escamosas/enzimologia , Adesão Celular/fisiologia , Divisão Celular/fisiologia , Transformação Celular Neoplásica , Bochecha , Cricetinae , DNA Complementar/administração & dosagem , DNA Complementar/genética , Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Mesocricetus , Fenótipo , Superóxido Dismutase/biossíntese , Transdução Genética , Transgenes
18.
Cancer Res ; 61(21): 8005-13, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11691826

RESUMO

Leukemia cells bearing the Philadelphia (Ph) chromosome express a Bcr-Abl fusion protein with deregulated protein tyrosine kinase (PTK) activity, which plays a central role in the malignant transformation. Many different signal transduction pathways are activated by Bcr-Abl, but little is known about their downstream targets in specific cell lineages. We show here that Ph-positive cell lines as well as primary cells derived from chronic myeloid leukemia (CML) in lymphoid blast crisis or from acute lymphoblastic leukemia (ALL) consistently express high levels of cyclin D2, whereas expression of this protein is low or absent in comparable Ph-negative lines and Ph-positive myeloid lines. Inhibition of Bcr-Abl with STI571 resulted in down-regulation of cyclin D2 and reduction of the number of cells in S phase, although complete G1 arrest was not induced. The expression of cyclin D2 in Ph-positive lymphoblasts was mediated via the phosphatidyl-inositol-3 kinase pathway. Analogous results were seen in murine BaF/3 cells transfected with a BCR-ABL expression vector. In contrast to the human cell lines, murine Baf/BCR-ABL cells exposed to STI571 inhibitor were all arrested in G1. This arrest could be abrogated by exogenous expression of cyclin D2 from a transfected cDNA construct. We conclude that a direct connection exists between Bcr-Abl PTK activity and cell cycle progression in which cyclin D2 plays a critical role. However, cell cycle progression in human Ph-positive lymphoid cells is not entirely dependent on Bcr-Abl PTK, and additional genetic lesions must be present.


Assuntos
Ciclinas/biossíntese , Proteínas de Fusão bcr-abl/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Linfócitos/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Proteínas Tirosina Quinases/metabolismo , Benzamidas , Crise Blástica , Ciclina D2 , Ciclinas/genética , Regulação para Baixo , Proteínas de Fusão bcr-abl/antagonistas & inibidores , Proteínas de Fusão bcr-abl/genética , Fase G1/efeitos dos fármacos , Humanos , Mesilato de Imatinib , Leucemia Mielogênica Crônica BCR-ABL Positiva/enzimologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Linfócitos/enzimologia , Fosfatidilinositol 3-Quinases/fisiologia , Piperazinas , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/genética , Pirimidinas/farmacologia , Fase S/efeitos dos fármacos , Transdução de Sinais , Transfecção
19.
Autophagy ; 12(10): 1804-1816, 2016 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-27532863

RESUMO

Bone remodeling is a continuous physiological process that requires constant generation of new osteoblasts from mesenchymal stem cells (MSCs). Differentiation of MSCs to osteoblast requires a metabolic switch from glycolysis to increased mitochondrial respiration to ensure the sufficient energy supply to complete this process. As a consequence of this increased mitochondrial metabolism, the levels of endogenous reactive oxygen species (ROS) rise. In the current study we analyzed the role of forkhead box O3 (FOXO3) in the control of ROS levels in human MSCs (hMSCs) during osteogenic differentiation. Treatment of hMSCs with H2O2 induced FOXO3 phosphorylation at Ser294 and nuclear translocation. This ROS-mediated activation of FOXO3 was dependent on mitogen-activated protein kinase 8 (MAPK8/JNK) activity. Upon FOXO3 downregulation, osteoblastic differentiation was impaired and hMSCs lost their ability to control elevated ROS levels. Our results also demonstrate that in response to elevated ROS levels, FOXO3 induces autophagy in hMSCs. In line with this, impairment of autophagy by autophagy-related 7 (ATG7) knockdown resulted in a reduced capacity of hMSCs to regulate elevated ROS levels, together with a reduced osteoblast differentiation. Taken together our findings are consistent with a model where in hMSCs, FOXO3 is required to induce autophagy and thereby reduce elevated ROS levels resulting from the increased mitochondrial respiration during osteoblast differentiation. These new molecular insights provide an important contribution to our better understanding of bone physiology.


Assuntos
Autofagia , Diferenciação Celular , Proteína Forkhead Box O3/metabolismo , Homeostase , Osteogênese , Autofagia/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/farmacologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Osteogênese/efeitos dos fármacos , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
20.
Oncogene ; 35(8): 990-1002, 2016 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-25961928

RESUMO

FOXM1 has been implicated in taxane resistance, but the molecular mechanism involved remains elusive. In here, we show that FOXM1 depletion can sensitize breast cancer cells and mouse embryonic fibroblasts into entering paclitaxel-induced senescence, with the loss of clonogenic ability, and the induction of senescence-associated ß-galactosidase activity and flat cell morphology. We also demonstrate that FOXM1 regulates the expression of the microtubulin-associated kinesin KIF20A at the transcriptional level directly through a Forkhead response element (FHRE) in its promoter. Similar to FOXM1, KIF20A expression is downregulated by paclitaxel in the sensitive MCF-7 breast cancer cells and deregulated in the paclitaxel-resistant MCF-7Tax(R) cells. KIF20A depletion also renders MCF-7 and MCF-7Tax(R) cells more sensitive to paclitaxel-induced cellular senescence. Crucially, resembling paclitaxel treatment, silencing of FOXM1 and KIF20A similarly promotes abnormal mitotic spindle morphology and chromosome alignment, which have been shown to induce mitotic catastrophe-dependent senescence. The physiological relevance of the regulation of KIF20A by FOXM1 is further highlighted by the strong and significant correlations between FOXM1 and KIF20A expression in breast cancer patient samples. Statistical analysis reveals that both FOXM1 and KIF20A protein and mRNA expression significantly associates with poor survival, consistent with a role of FOXM1 and KIF20A in paclitaxel action and resistance. Collectively, our findings suggest that paclitaxel targets the FOXM1-KIF20A axis to drive abnormal mitotic spindle formation and mitotic catastrophe and that deregulated FOXM1 and KIF20A expression may confer paclitaxel resistance. These findings provide insights into the underlying mechanisms of paclitaxel resistance and have implications for the development of predictive biomarkers and novel chemotherapeutic strategies for paclitaxel resistance.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos , Fatores de Transcrição Forkhead/fisiologia , Cinesinas/genética , Mitose , Paclitaxel/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Feminino , Proteína Forkhead Box M1 , Fatores de Transcrição Forkhead/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Humanos , Cinesinas/metabolismo , Camundongos , Mitose/efeitos dos fármacos , Regiões Promotoras Genéticas , Fuso Acromático/fisiologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA