Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 518(2): 374-380, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31434609

RESUMO

Recent evidence support that the c-Jun activation domain-binding protein 1 (JAB1)/COPS5 has an oncogenic function in various tissues. We show that JAB1 amplification in human prostate cancer (PCa) correlates with reduced overall survival and disease-free progression. Immunohistochemical staining shows enhanced expression of JAB1 in the cytoplasmic compartment of PCa cells compared to the normal prostate epithelium, indicating the activity/function of JAB1 is altered in PCa. To test the function of JAB1 in PCa, we efficiently silenced JAB1 expression using four unique shRNAs in three PCa cell lines (LNCaP, C4-2, and PC-3) and an immortalized prostate epithelial cell line, RWPE-1. Our data clearly show that silencing JAB1 robustly suppresses the growth of PCa cells, but not RWPE-1 cells, suggesting that PCa cells become addicted to JAB1. To study the potential mechanism by which JAB1 controls PCa growth, we profiled gene expression changes by whole transcriptome microarray analysis of C4-2 cells silenced for JAB1 using a pool of 3 shRNAs compared to scrambled shRNA control. We identified 1268 gene changes ≥1.5 fold by silencing JAB1 in C4-2. Western blot confirmation and bioinformatics pathway analyses support that PCa cells become addicted to JAB1 through controlling the following signaling pathways: cell cycle, p53 signaling, DNA replication, TGF-ß/BMP, MAPK, TNF, and steroid hormone biosynthesis. We propose that UGT2B28, UGT2B10, UGT2B11, Skp2, EZH2, MDM2, BIRC5 (Survivin), UBE2C, and Smads 1/5/8, which are all associated with the abovementioned key oncogenic pathways, may play critical roles in the putative oncogenic function of JAB1 in PCa.


Assuntos
Complexo do Signalossomo COP9/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Oncogênicas/metabolismo , Peptídeo Hidrolases/metabolismo , Neoplasias da Próstata/metabolismo , Complexo do Signalossomo COP9/genética , Proliferação de Células , Sobrevivência Celular , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Peptídeo Hidrolases/genética , Neoplasias da Próstata/patologia , Taxa de Sobrevida , Células Tumorais Cultivadas
2.
J Biol Chem ; 291(11): 5512-5526, 2016 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-26786102

RESUMO

The transcription factor c-Fos controls many important cellular processes, including cell growth and apoptosis. c-Fos expression is rapidly elevated in the prostate upon castration-mediated androgen withdrawal through an undefined mechanism. Here we show that androgens (5α-dihydrotestosterone and R1881) suppress c-Fos protein and mRNA expression induced by 12-O-tetradecanoylphorbol-13-acetate (TPA) or EGF in human prostate cancer (PCa) cell lines. Such suppression transpires through a transcriptional mechanism, predominantly at the proximal serum response element of the c-fos promoter. We show that androgen signaling suppresses TPA-induced c-Fos expression through repressing a PKC/MEK/ERK/ELK-1 signaling pathway. Moreover, our results support the hypothesis that p38(MAPK), PI3K, and PKCδ are involved in the androgenic regulation of c-Fos through controlling MEK/ERK. Stable silencing of c-Fos and PKCδ with shRNAs suggests that R1881 promotes cell death induced by low-dose TPA through a mechanism that is dependent on both PKCδ and loss of c-Fos expression. Reciprocally, loss of either PKCδ or c-Fos activates p38(MAPK) while suppressing the activation of ERK1/2. We also provide the first demonstration that R1881 permits cell death induced by low-dose TPA in the LNCaP androgen-dependent PCa cell line and that TPA-induced cell death is independent of exogenous androgen in the castration-resistant variants of LNCaP, C4-2 and C4-2B. Acquisition of androgen-independent killing by TPA correlates with activation of p38(MAPK), suppression of ERK1/2, and loss of c-Fos. These results provide new insights into androgenic control of c-Fos and use of PKC inhibitors in PCa therapy.


Assuntos
Adenocarcinoma/tratamento farmacológico , Androgênios/farmacologia , Di-Hidrotestosterona/farmacologia , Metribolona/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Proteínas Proto-Oncogênicas c-fos/genética , Acetato de Tetradecanoilforbol/farmacologia , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Próstata/efeitos dos fármacos , Próstata/metabolismo , Próstata/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteína Quinase C/metabolismo , RNA Mensageiro/genética , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
J Biol Chem ; 289(1): 251-63, 2014 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-24265312

RESUMO

CREB-binding protein (CBP)/p300 interacting transactivator with glutamic acid (Glu) and aspartic acid (Asp)-tail 2 (Cited2) was recently shown to be essential for gluconeogenesis in the adult mouse. The metabolic function of Cited2 in mouse embryonic stem cells (mESCs) remains elusive. In the current study, the metabolism of glucose was investigated in mESCs, which contained a deletion in the gene for Cited2 (Cited2(Δ/-)). Compared with its parental wild type counterpart, Cited2(Δ/-) ESCs have enhanced glycolysis, alternations in mitochondria morphology, reduced glucose oxidation, and decreased ATP content. Cited2 is recruited to the hexokinase 1 (HK1) gene promoter to regulate transcription of HK1, which coordinates glucose metabolism in wild type ESCs. Reduced glucose oxidation and enhanced glycolytic activity in Cited2(Δ/-) ESCs correlates with defective differentiation during hypoxia, which is reflected in an increased expression of pluripotency marker (Oct4) and epiblast marker (Fgf5) and decreased expression of lineage specification markers (T, Gata-6, and Cdx2). Knockdown of hypoxia inducible factor-1α in Cited2(Δ/-) ESCs re-initiates the expression of differentiation markers T and Gata-6. Taken together, a deletion of Cited2 in mESCs results in abnormal mitochondrial morphology and impaired glucose metabolism, which correlates with a defective cell fate decision.


Assuntos
Células-Tronco Embrionárias/metabolismo , Glicólise/fisiologia , Mitocôndrias/metabolismo , Proteínas Repressoras/metabolismo , Transativadores/metabolismo , Transcrição Gênica/fisiologia , Trifosfato de Adenosina/biossíntese , Trifosfato de Adenosina/genética , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Hipóxia Celular/fisiologia , Células-Tronco Embrionárias/citologia , Glucose/genética , Glucose/metabolismo , Hexoquinase/biossíntese , Hexoquinase/genética , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Oxirredução , Proteínas Repressoras/genética , Transativadores/genética
4.
Biochem J ; 462(2): 315-27, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24844355

RESUMO

We show that HEXIM1 (hexamethylene bis-acetamide inducible 1) functions as an AR (androgen receptor) co-repressor as it physically interacts with the AR and is required for the ability of anti-androgens to inhibit androgen-induced target gene expression and cell proliferation. Oncomine™ database and IHC (immunohistochemistry) analyses of human prostate tissues revealed that expression of HEXIM1 mRNA and protein are down-regulated during the development and progression of prostate cancer. Enforced down-regulation of HEXIM1 in parental hormone-dependent LNCaP cells results in resistance to the inhibitory action of anti-androgens. Conversely, ectopic expression of HEXIM1 in the CRPC (castration-resistant prostate cancer) cell line, C4-2, enhances their sensitivity to the repressive effects of the anti-androgen bicalutamide. Novel insight into the mechanistic basis for HEXIM1 inhibition of AR activity is provided by the present studies showing that HEXIM1 induces expression of the histone demethylase KDM5B (lysine-specific demethylase 5B) and inhibits histone methylation, resulting in the inhibition of FOXA1 (forkhead box A1) licensing activity. This is a new mechanism of action attributed to HEXIM1, and distinct from what has been reported so far to be involved in HEXIM1 regulation of other nuclear hormone receptors, including the oestrogen receptor.


Assuntos
Antagonistas de Androgênios/farmacologia , Neoplasias da Próstata/metabolismo , Proteínas de Ligação a RNA/metabolismo , Receptores Androgênicos/metabolismo , Anilidas/farmacologia , Linhagem Celular Tumoral , Elementos Facilitadores Genéticos , Células Epiteliais/metabolismo , Expressão Gênica/efeitos dos fármacos , Histonas/metabolismo , Humanos , Histona Desmetilases com o Domínio Jumonji/metabolismo , Masculino , Metribolona/farmacologia , Nitrilas/farmacologia , Proteínas Nucleares/metabolismo , Próstata/metabolismo , Neoplasias de Próstata Resistentes à Castração/metabolismo , Transporte Proteico , Proteínas Repressoras/metabolismo , Compostos de Tosil/farmacologia , Fatores de Transcrição , Fatores de Elongação da Transcrição/metabolismo
5.
Blood ; 119(12): 2789-98, 2012 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-22308296

RESUMO

Cited2 is a transcriptional modulator involved in various biologic processes including fetal liver hematopoiesis. In the present study, the function of Cited2 in adult hematopoiesis was investigated in conditional knockout mice. Deletion of Cited2 using Mx1-Cre resulted in increased hematopoietic stem cell (HSC) apoptosis, loss of quiescence, and increased cycling, leading to a severely impaired reconstitution capacity as assessed by 5-fluorouracil treatment and long-term transplantation. Transcriptional profiling revealed that multiple HSC quiescence- and hypoxia-related genes such as Egr1, p57, and Hes1 were affected in Cited2-deficient HSCs. Because Cited2 is a negative regulator of HIF-1, which is essential for maintaining HSC quiescence, and because we demonstrated previously that decreased HIF-1α gene dosage partially rescues both cardiac and lens defects caused by Cited2 deficiency, we generated Cited2 and HIF-1α double-knockout mice. Additional deletion of HIF-1α in Cited2-knockout BM partially rescued impaired HSC quiescence and reconstitution capacity. At the transcriptional level, deletion of HIF-1α restored expression of p57 and Hes1 but not Egr1 to normal levels. Our results suggest that Cited2 regulates HSC quiescence through both HIF-1-dependent and HIF-1-independent pathways.


Assuntos
Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Proteínas Repressoras/deficiência , Transativadores/deficiência , Animais , Apoptose/fisiologia , Western Blotting , Citometria de Fluxo , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Repressoras/genética , Transativadores/genética
6.
Proc Natl Acad Sci U S A ; 108(21): 8668-73, 2011 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-21555587

RESUMO

Oncogene-induced senescence (OIS), the proliferative arrest engaged in response to persistent oncogene activation, serves as an important tumor-suppressive barrier. We show here that finite lifespan human mammary epithelial cells (HMEC) undergo a p16/RB- and p53-independent OIS in response to oncogenic RAS that requires TGF-ß signaling. Suppression of TGF-ß signaling by expression of a dominant-negative TGF-ß type II receptor, use of a TGF-ß type I receptor inhibitor, or ectopic expression of MYC permitted continued proliferation upon RAS expression. Surprisingly, unlike fibroblasts, shRNA-mediated knockdown of ATM or CHK2 was unable to prevent RAS-mediated OIS, arguing that the DNA damage response is not required for OIS in HMEC. Abrogation of TGF-ß signaling not only allowed HMEC lacking p53 to tolerate oncogenic RAS but also conferred the capacity for anchorage-independent growth. Thus, the OIS engaged after dysregulated RAS expression provides an early barrier to malignant progression and is mediated by TGF-ß receptor activation in HMEC. Understanding the mechanisms that initiate and maintain OIS in epithelial cells may provide a foundation for future therapies aimed at reengaging this proliferative barrier as a cancer therapy.


Assuntos
Transformação Celular Neoplásica , Senescência Celular , Glândulas Mamárias Humanas/citologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Proteínas ras/fisiologia , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Células Cultivadas , Quinase do Ponto de Checagem 2 , Proteínas de Ligação a DNA/metabolismo , Células Epiteliais , Humanos , Glândulas Mamárias Humanas/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo
7.
Pharmaceuticals (Basel) ; 17(4)2024 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-38675493

RESUMO

The TGF-ß family is a group of 25 kDa secretory cytokines, in mammals consisting of three dimeric isoforms (TGF-ßs 1, 2, and 3), each encoded on a separate gene with unique regulatory elements. Each isoform plays unique, diverse, and pivotal roles in cell growth, survival, immune response, and differentiation. However, many researchers in the TGF-ß field often mistakenly assume a uniform functionality among all three isoforms. Although TGF-ßs are essential for normal development and many cellular and physiological processes, their dysregulated expression contributes significantly to various diseases. Notably, they drive conditions like fibrosis and tumor metastasis/progression. To counter these pathologies, extensive efforts have been directed towards targeting TGF-ßs, resulting in the development of a range of TGF-ß inhibitors. Despite some clinical success, these agents have yet to reach their full potential in the treatment of cancers. A significant challenge rests in effectively targeting TGF-ßs' pathological functions while preserving their physiological roles. Many existing approaches collectively target all three isoforms, failing to target just the specific deregulated ones. Additionally, most strategies tackle the entire TGF-ß signaling pathway instead of focusing on disease-specific components or preferentially targeting tumors. This review gives a unique historical overview of the TGF-ß field often missed in other reviews and provides a current landscape of TGF-ß research, emphasizing isoform-specific functions and disease implications. The review then delves into ongoing therapeutic strategies in cancer, stressing the need for more tools that target specific isoforms and disease-related pathway components, advocating mechanism-based and refined approaches to enhance the effectiveness of TGF-ß-targeted cancer therapies.

8.
Artigo em Inglês | MEDLINE | ID: mdl-35005610

RESUMO

The imidazolium compound Sepantronium Bromide (YM155) successfully promotes tumor regression in various pre-clinical models but has shown modest responses in human clinical trials. We provide evidence to support that the hypoxic milieu of tumors may limit the clinical usefulness of YM155. Hypoxia (1% O2) strongly (>16-fold) represses the cytotoxic activity of YM155 on prostate and renal cancer cells in vitro. Hypoxia also represses all early signaling responses associated with YM155, including activation of AMPK and retinoblastoma protein (Rb), inactivation of the mechanistic target of rapamycin complex 1 (mTORC1), inhibition of phospho-ribosomal protein S6 (rS6), and suppression of the expression of Cyclin Ds, Mcl-1 and Survivin. Cells pre-incubated with hypoxia for 24 â€‹h are desensitized to YM155 even when they are treated with YM155 under atmospheric oxygen conditions, supporting that cells at least temporarily retain hypoxia-induced resistance to YM155. We tested the role of hypoxia-inducible factor (HIF)-1α and HIF-2α in the hypoxia-induced resistance to YM155 by comparing responses of YM155 in VHL-proficient versus VHL-deficient RCC4 and 786-O renal cancer cells and silencing HIF expression in PC-3 prostate cancer cells. Those studies suggested that hypoxia-induced resistance to YM155 occurs independent of HIF-1α and HIF-2α. Moreover, the hypoxia mimetics deferoxamine and dimethyloxalylglycine, which robustly induce HIF-1α levels in PC-3 â€‹cells under atmospheric oxygen, did not diminish their early cellular responses to YM155. Collectively, our data support that hypoxia induces resistance of cells to YM155 through a HIF-1α and HIF-2α-independent mechanism. We hypothesize that a hypothetical hypoxia-inducer factor (HIF-X) represses early signaling responses to YM155.

9.
Artigo em Inglês | MEDLINE | ID: mdl-35992379

RESUMO

The mammalian target of rapamycin (mTOR) plays an important role in the aggressiveness and therapeutic resistance of many cancers. Targeting mTOR continues to be under clinical investigation for cancer therapy. Despite the notable clinical success of mTOR inhibitors in extending the overall survival of patients with certain malignancies including metastatic renal cell carcinomas (RCCs), the overall impact of mTOR inhibitors on cancers has been generally disappointing and attributed to various compensatory responses. Here we provide the first report that expression of the Notch ligand Jagged-1 (JAG1), which is associated with aggressiveness of RCCs, is induced by several inhibitors of mTOR (rapamycin (Rap), BEZ235, KU-0063794) in human clear cell RCC (ccRCC) cells. Using both molecular and chemical inhibitors of PI3K, Akt, and TGF-ß signaling, we provide evidence that the induction of JAG1 expression by mTOR inhibitors in ccRCC cells depends on the activation of Akt and occurs through an ALK5 kinase/Smad4-dependent mechanism. Furthermore, we show that mTOR inhibitors activate Notch1 and induce the expression of drivers of epithelial-mesenchymal transition, notably Hic-5 and Slug. Silencing JAG1 with selective shRNAs blocked the ability of KU-0063794 and Rap to induce Hic-5 in ccRCC cells. Moreover, Rap enhanced TGF-ß-induced expression of Hic-5 and Slug, both of which were repressed in JAG1-silenced ccRCC cells. Silencing JAG1 selectively decreased the motility of ccRCC cells treated with Rap or TGF-ß1. Moreover, inhibition of Notch signaling with γ-secretase inhibitors enhanced or permitted mTOR inhibitors to suppress the motility of ccRCC cells. We suggest targeting JAG1 may enhance therapeutic responses to mTOR inhibitors in ccRCCs.

10.
J Biol Chem ; 285(46): 35578-89, 2010 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-20826775

RESUMO

Prostate cancer is a major cause of mortality in men in developed countries. It has been reported that the naturally occurring antioxidant α-tocopherol (vitamin E) attenuates prostate cancer cell proliferation in cultured cells and mouse models. We hypothesized that overexpression of the tocopherol transfer protein (TTP), a vitamin E-binding protein that regulates tocopherol status, will sensitize prostate cancer cells to the anti-proliferative actions of the vitamin. To test this notion, we manipulated the expression levels of TTP in cultured prostate cells (LNCaP, PC3, DU145, and RWPE-1) using overexpression and knockdown approaches. Treatment of cells with tocopherol caused a time- and dose-dependent inhibition of cell proliferation. Overexpression of TTP dramatically sensitized the cells to the apoptotic effects of α-tocopherol, whereas reduction ("knockdown") of TTP expression resulted in resistance to the vitamin. TTP levels also augmented the inhibitory effects of vitamin E on proliferation in semi-solid medium. The sensitizing effects of TTP were paralleled by changes in the intracellular accumulation of a fluorescent analog of vitamin E and by a reduction in intracellular levels of reactive oxygen species and were not observed when a naturally occurring, ligand binding-defective mutant of TTP was used. We conclude that TTP sensitizes prostate cancer cells to the anti-proliferative effects of vitamin E and that this activity stems from the ability of protein to increase the intracellular accumulation of the antioxidant. These observations support the notion that individual changes in the expression level or activity of TTP may determine the responsiveness of prostate cancer patients to intervention strategies that utilize vitamin E.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Proliferação de Células/efeitos dos fármacos , Vitamina E/farmacologia , Western Blotting , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Células HEK293 , Células Hep G2 , Humanos , Masculino , Microscopia de Fluorescência , Mitocôndrias/metabolismo , Mutação , Poli(ADP-Ribose) Polimerases/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo , Vitaminas/farmacologia , alfa-Tocoferol/farmacologia
12.
Am J Cancer Res ; 11(10): 5063-5075, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34765312

RESUMO

Chondrosarcoma (CS) is the second most common skeletal malignancy in humans. High-grade CS is aggressive and extremely resistant to chemo- and radio-therapies. The lack of effective treatment options warrants the development of novel therapies. The evolutionarily conserved transcriptional co-factor JAB1 (also known as COPS5/CSN5) has emerged as a novel regulator of tumorigenesis. JAB1 overexpression occurs in many common cancers and is associated with poor prognosis. However, the role of JAB1 in CS pathogenesis was completely unknown. To study JAB1's function in CS, we performed shRNA knockdown (KD) of JAB1 in two high-grade human CS cell lines, SW1353 and Hs819.T, and observed significantly decreased proliferation and colony formations, and increased apoptosis in both CS cell lines upon JAB1-KD. Interestingly, we found that endogenous JAB1 interacted with endogenous SOX9, a potent oncogene and a master regulator of skeletogenesis, in chondrosarcoma cells, but not in primary chondrocytes. JAB1 also binds to the same SOX9-mediated chondrocyte-specific enhancer elements in CS cells. Furthermore, we found that a recently developed, novel, potent, and JAB1-specific small molecule inhibitor, CSN5i-3, can significantly increase apoptosis, drastically alter the activities of several signaling pathways, and modulates the expression of specific Cullin-ring-ligases (CRLs) in CS cells. Finally, our RNA-sequencing analysis in JAB1-KD CS cells identified a total of 2945 differentially expressed genes. Gene set enrichment analysis revealed that JAB1 regulates several essential pathways such as DNA damage response and cell cycle regulation. In conclusion, our study showed that JAB1 might regulate a distinct pro-tumorigenic regulatory network to promote chondrosarcoma pathogenesis.

13.
Oncogene ; 39(23): 4581-4591, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32390003

RESUMO

Osteosarcoma (OS) is the most common primary bone cancer and ranks amongst the leading causes of cancer mortality in young adults. Jun activation domain-binding protein 1 (JAB1) is overexpressed in many cancers and has recently emerged as a novel target for cancer treatment. However, the role of JAB1 in osteosarcoma was virtually unknown. In this study, we demonstrate that JAB1-knockdown in malignant osteosarcoma cell lines significantly reduced their oncogenic properties, including proliferation, colony formation, and motility. We also performed RNA-sequencing analysis in JAB1-knockdown OS cells and identified 4110 genes that are significantly differentially expressed. This demonstrated for the first time that JAB1 regulates a large and specific transcriptome in cancer. We also found that JAB1 is overexpressed in human OS and correlates with a poor prognosis. Moreover, we generated a novel mouse model that overexpresses Jab1 specifically in osteoblasts upon a TP53 heterozygous sensitizing background. Interestingly, by 13 months of age, a significant proportion of these mice spontaneously developed conventional OS. Finally, we demonstrate that a novel, highly specific small molecule inhibitor of JAB1, CSN5i-3, reduces osteosarcoma cell viability, and has specific effects on the ubiquitin-proteasome system in OS. Thus, we show for the first time that the overexpression of JAB1 in vivo can result in accelerated spontaneous tumor formation in a p53-dependent manner. In summary, JAB1 might be a unique target for the treatment of osteosarcoma and other cancers.


Assuntos
Neoplasias Ósseas/patologia , Complexo do Signalossomo COP9/metabolismo , Carcinogênese/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Osteossarcoma/patologia , Peptídeo Hidrolases/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Neoplasias Ósseas/genética , Complexo do Signalossomo COP9/genética , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Reparo do DNA/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Osteossarcoma/genética , Peptídeo Hidrolases/genética
14.
Mol Cancer Res ; 6(2): 231-42, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18314484

RESUMO

Androgen withdrawal induces the regression of human prostate cancers, but such cancers eventually become androgen-independent and metastasize. Thus, deciphering the mechanism of androgen withdrawal-induced apoptosis is critical to designing new therapies for prostate cancer. Previously, we showed that in the rat, castration-induced apoptosis is accompanied by a reduction in the expression of the apical caspase inhibitor FLICE-like inhibitory protein (FLIP). To test the functional role of FLIP in inhibiting prostate epithelial cell apoptosis, we employed the rat prostate epithelial cell line NRP-152, which differentiates to a secretory phenotype in a low-mitogen medium and then undergoes apoptosis following the addition of transforming growth factor beta1 (TGFbeta1), mimicking androgen withdrawal-induced apoptosis. FLIP levels decline with TGFbeta1 treatment, suggesting that apoptosis is mediated by caspase-8 and indeed the caspase inhibitor crmA blocks TGFbeta1-induced apoptosis. Small interfering RNA-mediated knockdown of FLIP recapitulates and enhances TGFbeta1-induced cell death. NRP-152 cells stably transfected with constitutively expressed FLIP were refractory to TGFbeta1-induced apoptosis. TGFbeta1-induced caspase-3 activity is proportional to the level of cell death and inversely proportional to the level of FLIP expression in various clones. Moreover, neither caspase-3 nor PARP is cleaved in clones expressing high levels of FLIP. Furthermore, insulin, which inhibits differentiation, increases FLIP and inhibits TGFbeta-induced death in a FLIP-dependent manner. Although neither Fas-Fc, sTNFRII-Fc, nor DR5-Fc blocked TGFbeta1-induced cell death, there is a significant increase in tumor necrosis factor mRNA following TGFbeta stimulation, suggesting both an unexpected role for tumor necrosis factor in this model system and the possibility that FLIP blocks another unknown caspase-dependent mediator of apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Caspases/metabolismo , Células Epiteliais/citologia , Células Epiteliais/enzimologia , Próstata/enzimologia , Fator de Crescimento Transformador beta1/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Etanercepte , Proteína Ligante Fas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Imunoglobulina G/farmacologia , Insulina/farmacologia , Masculino , Camundongos , Próstata/citologia , Próstata/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Receptores do Fator de Necrose Tumoral , Solubilidade/efeitos dos fármacos , Transfecção
15.
Cytokine Growth Factor Rev ; 17(1-2): 59-74, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16297654

RESUMO

Insulin-like growth factor-I (IGF-I) has gained broad recognition as an important survival factor for epithelial cells in numerous tissues. The IGF-I receptor signaling pathway is deregulated in the majority of carcinomas, and such deregulation has also been reported to be tightly associated with enhanced tumor progression and metastasis. One of the key proteins that transduces IGF-I signals and is phospho-activated downstream of the IGF-I receptor, is the non-receptor serine/threonine kinase proto-oncogene protein kinase B (PKB, also known as Akt). This kinase serves as a major molecular node to control the function of many cell survival and death proteins through phosphorylation-mediated protein modification. The end result of the activation of Akt is enhanced cell survival and proliferation, pre-requisites for malignant transformation. Recent studies show that IGF-I signals cross-talk at multiple levels with various components of the TGF-beta signaling pathway, which depending on context may function either as tumor suppressor or as tumor promoter. Thus, a better understanding of how the IGF-I and TGF-beta signaling pathways are mutually interconnected is likely to unveil novel targets for the therapeutic intervention of many cancers.


Assuntos
Comunicação Celular/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Humanos , Proto-Oncogene Mas , Receptor IGF Tipo 1/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/fisiologia
16.
Mol Cancer Ther ; 7(6): 1347-54, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18566209

RESUMO

The mammalian target of rapamycin (mTOR) is a protein kinase that regulates protein translation, cell growth, and apoptosis. Recently, there has been an enormous increase in our understanding on molecular mechanisms underlying the therapeutics of rapamycin in cancer. Alterations in the pathway regulating mTOR occur in many solid malignancies including prostate, bladder, and kidney cancer; in vitro and in vivo models of prostate and bladder cancer have established the importance of the mTOR pathway in control of cancer progression and metastasis. Temsirolimus (Torisel) and everolimus (RAD-001), two ester analogues of rapamycin, as well as rapamycin itself have clear antitumor activity in in vitro and in vivo models and are under clinical trial investigations for prostate and bladder cancer. Phase II and III trials have already established the clinical efficacy of temsirolimus in renal cancer, and current renal trials are evaluating the combined effects of vascular endothelial growth factor and mTOR inhibition. Ongoing studies in prostate and bladder cancer will soon define the activity and safety profiles of everolimus and temsirolimus. Recent molecular advances have uncovered a startling complexity in the macromolecular function of mTOR complexes, with the identification of new mTOR partners (raptor, rictor, FKBP38, PRAS40, and mSIN1), putative cancer therapeutic/prognostic targets for future clinical trials.


Assuntos
Proteínas Quinases/metabolismo , Neoplasias Urológicas/enzimologia , Neoplasias Urológicas/terapia , Humanos , Transdução de Sinais/efeitos dos fármacos , Sirolimo/análogos & derivados , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Proteínas Supressoras de Tumor/metabolismo
17.
Sci Rep ; 9(1): 14826, 2019 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-31597941

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

18.
Sci Rep ; 9(1): 11541, 2019 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-31395901

RESUMO

The imidazolium compound YM155, first discovered as a potent inhibitor of Survivin, effectively kills many carcinomas in preclinical models. However, the upstream signaling mechanism triggered by YM155 remains unclear. Here we studied early signaling responses in vitro in prostate and renal cancer cell lines in a dose-dependent manner. We found that YM155 rapidly activates the retinoblastoma protein, correlating with the loss of expression of all three Cyclin Ds. Using Western blot, various selective chemical inhibitors and q-PCR, we show that YM155-mediated decrease in protein levels of Cyclin Ds, Survivin and Mcl-1 is independent of transcription or proteasomal control mechanisms. Moreover, we provide the first evidence that YM155 changes the phosphorylation status of known mTOR-target proteins involved in translational control, namely ribosomal protein S6 (rS6) and 4E-BP1. Our data support that YM155 achieves this by blocking mTORC1 via the phosphorylation of Raptor at S792 through activated AMPKα (T172). Furthermore, we also used a polysome profile, supporting that YM155 markedly suppresses cap-dependent translation of mRNAs which include Survivin, Cyclin D1 and Mcl-1. We provide the first evidence that YM155 functions as a potent activator of AMPKα, a robust suppressor of mTORC1 and an attenuator of global protein synthesis.


Assuntos
Carcinoma/tratamento farmacológico , Imidazóis/farmacologia , Naftoquinonas/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Proteínas Quinases/genética , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Adaptadoras de Transdução de Sinal/genética , Apoptose/efeitos dos fármacos , Carcinoma/genética , Carcinoma/patologia , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclina D/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Próstata/efeitos dos fármacos , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Transdução de Sinais/efeitos dos fármacos , Survivina/genética
19.
J Cell Physiol ; 216(1): 172-9, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18247359

RESUMO

Store-operated Ca2+ channels control homeostasis between extracellular Ca2+ reservoir and intracellular Ca2+ storage and play important roles in apoptosis in a wide variety of cells, including prostate epithelia. Recent studies have shown that the acquired apoptosis-resistant nature of androgen-independent prostate cancer is associated with reduced function of store-operated Ca2+ entry (SOCE). This study investigates the functional interaction between Bax and SOCE in the apoptosis signaling cascade in prostate cancer. Our previous findings show that NRP-154, an androgen-independent prostate cancer cell line, could sustain overexpression of exogenous Bax without undergoing apoptosis. Here we show that sustained overexpression of Bax in NRP-154 cells leads to down-regulation of SOCE and reduced Ca2+ storage inside the endoplasmic reticulum. While reduced SOCE may represent an adaptive mechanism for cell survival, increased levels of Bax in the latent state enhances the sensitivity of NRP-154 cells to TGF-beta and thapsigargin-induced apoptosis. This enhanced apoptosis can be reduced by 2-aminoethoxydiphenyl borate (2-APB), an inhibitor of SOCE, or reversed under conditions where SOCE is only partially activated. Our results demonstrate a functional interaction between Bax and SOCE in apoptosis of prostate cancer, and support the concept that improving this interaction has therapeutic implications for prostate cancer.


Assuntos
Apoptose/fisiologia , Cálcio/metabolismo , Neoplasias da Próstata/metabolismo , Proteína X Associada a bcl-2/metabolismo , Animais , Linhagem Celular Tumoral , Regulação para Baixo , Regulação da Expressão Gênica , Homeostase , Humanos , Masculino , Camundongos , Ratos , Ratos Wistar , Transdução de Sinais/fisiologia , Proteína X Associada a bcl-2/genética
20.
Endocrinology ; 148(3): 1235-45, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17138648

RESUMO

Axis positioning and tissue determination during development involve coordinated expression of Hox genes throughout the body. The most posterior Hox gene clusters are involved in prostate organogenesis. In the present study, we characterized and compared the expression profiles of posterior (5') Hox genes in the separate lobes of the adult rat prostate gland, the coagulating gland, seminal vesicles, and epididymis using quantitative real-time RT-PCR. These genes include Hoxa9-11, Hoxa13, Hoxd13, and Hoxb13. We identified a unique Hox code for each of these organs and propose that this contributes to the organ-specific and prostate lobe-specific identities in the adult rat. Using the ventral prostate (VP) as a model, we characterized the Hox genes expression patterns over time from birth through adulthood. Expression levels of the three Hox13 genes and Hoxa10 were significantly higher in the adult VP compared with the neonatal developing VP suggesting an important role during adult homeostasis. In contrast, Hoxa9 and Hoxa11 levels declined after morphogenesis suggesting a specific developmental role. Overall, the Hoxb13 gene exhibited the most striking temporal and organ-specific differences. Using in situ hybridization and immunohistochemistry, a distinct Hoxb13 anterior-to-posterior expression gradient was observed with the highest expression levels in the VP luminal epithelial cells, moderate levels in the lateral prostate, and low expression in the dorsal prostate. An expression gradient was also observed along the ductal length in all three prostate lobes with strongest expression at the distal tips and limited expression in the proximal ducts. After infection with a lentivirus expressing the Hoxb13 gene, NRP-152 cells cultured under nondifferentiating conditions exhibited robust cytokeratin 8 immunostain indicating that Hoxb13 expression drives luminal cell differentiation in the rat epithelium. Androgen regulation of prostatic Hox gene expression was examined during development in vitro and after castration in the adult rat. In the neonatal VP, all six Hox genes were significantly up-regulated by androgens, whereas none of the genes were affected by testosterone in the lateral prostate. In the adult rat, castration resulted in up-regulation of Hoxa9 and Hoxa13 in the VP and down-regulation of Hoxb13 in the dorsal prostate and lateral prostate. Taken together, we conclude that the prostatic Hox genes reach a destined expression level at specific developmental time points in the prostate gland and possess differential androgenic regulation in a temporal and lobe-specific manner. We suggest that this timely Hox code participates in determining lobe-specific prostatic identity and cellular differentiation.


Assuntos
Androgênios/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteínas de Homeodomínio/metabolismo , Próstata/crescimento & desenvolvimento , Próstata/metabolismo , Animais , Animais Geneticamente Modificados , Animais Recém-Nascidos , Diferenciação Celular , Células Cultivadas , Células Epiteliais/citologia , Feminino , Proteínas de Homeodomínio/fisiologia , Humanos , Masculino , Gravidez , Próstata/citologia , Próstata/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA