Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Mol Cancer Res ; 16(12): 1940-1951, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30042175

RESUMEN

Prostate cancer is a prevalent public health problem, especially because noncutaneous advanced malignant forms significantly affect the lifespan and quality of life of men worldwide. New therapeutic targets and approaches are urgently needed. The current study reports elevated expression of R1 (CDCA7L/RAM2/JPO2), a c-Myc-interacting protein and transcription factor, in human prostate cancer tissue specimens. In a clinical cohort, high R1 expression is associated with disease recurrence and decreased patient survival. Overexpression and knockdown of R1 in human prostate cancer cells indicate that R1 induces cell proliferation and colony formation. Moreover, silencing R1 dramatically reduces the growth of prostate tumor xenografts in mice. Mechanistically, R1 increases c-Myc protein stability by inhibiting ubiquitination and proteolysis through transcriptional suppression of HUWE1, a c-Myc-targeting E3 ligase, via direct interaction with a binding element in the promoter. Moreover, transcriptional repression is supported by a negative coexpression correlation between R1 and HUWE1 in a prostate cancer clinical dataset. Collectively, these findings, for the first time, characterize the contribution of R1 to prostate cancer pathogenesis. IMPLICATIONS: These findings provide evidence that R1 is a novel regulator of prostate tumor growth by stabilizing c-Myc protein, meriting further investigation of its therapeutic and prognostic potential.


Asunto(s)
Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas Supresoras de Tumor/genética , Ubiquitina-Proteína Ligasas/genética , Regulación hacia Arriba , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Mutación , Trasplante de Neoplasias , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-myc/química , Proteínas Proto-Oncogénicas c-myc/genética , Análisis de Supervivencia
2.
Cancer Cell ; 31(3): 368-382, 2017 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-28292438

RESUMEN

Metastasis is a predominant cause of death for prostate cancer (PCa) patients; however, the underlying mechanisms are poorly understood. We report that monoamine oxidase A (MAOA) is a clinically and functionally important mediator of PCa bone and visceral metastases, activating paracrine Shh signaling in tumor-stromal interactions. MAOA provides tumor cell growth advantages in the bone microenvironment by stimulating interleukin-6 (IL6) release from osteoblasts, and triggers skeletal colonization by activating osteoclastogenesis through osteoblast production of RANKL and IL6. MAOA inhibitor treatment effectively reduces metastasis and prolongs mouse survival by disengaging the Shh-IL6-RANKL signaling network in stromal cells in the tumor microenvironment. These findings provide a rationale for targeting MAOA and its associated molecules to treat PCa metastasis.


Asunto(s)
Comunicación Celular , Proteínas Hedgehog/fisiología , Interleucina-6/fisiología , Monoaminooxidasa/fisiología , Neoplasias de la Próstata/patología , Ligando RANK/fisiología , Transducción de Señal/fisiología , Animales , Neoplasias Óseas/secundario , Humanos , Masculino , Ratones , Ratones SCID , Monoaminooxidasa/análisis , Osteoblastos/fisiología , Células del Estroma/fisiología , Microambiente Tumoral
3.
Oncotarget ; 7(51): 84645-84657, 2016 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-27835867

RESUMEN

Lethal progression of prostate cancer metastasis can be improved by developing animal models that recapitulate the clinical conditions. We report here that cytokeratin 13 (KRT13), an intermediate filament protein, plays a directive role in prostate cancer bone, brain, and soft tissue metastases. KRT13 expression was elevated in bone, brain, and soft tissue metastatic prostate cancer cell lines and in primary and metastatic clinical prostate, lung, and breast cancer specimens. When KRT13 expression was determined at a single cell level in primary tumor tissues of 44 prostate cancer cases, KRT13 level predicted bone metastasis and the overall survival of prostate cancer patients. Genetically enforced KRT13 expression in human prostate cancer cell lines drove metastases toward mouse bone, brain and soft tissues through a RANKL-independent mechanism, as KRT13 altered the expression of genes associated with EMT, stemness, neuroendocrine/neuromimicry, osteomimicry, development, and extracellular matrices, but not receptor activator NF-κB ligand (RANKL) signaling networks in prostate cancer cells. Our results suggest new inhibitors targeting RANKL-independent pathways should be developed for the treatment of prostate cancer bone and soft tissue metastases.


Asunto(s)
Adenocarcinoma/metabolismo , Neoplasias Óseas/metabolismo , Neoplasias Encefálicas/metabolismo , Queratina-13/metabolismo , Neoplasias de la Próstata/metabolismo , Adenocarcinoma/secundario , Animales , Neoplasias Óseas/secundario , Neoplasias Encefálicas/secundario , Línea Celular Tumoral , Movimiento Celular , Reprogramación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Queratina-13/genética , Masculino , Ratones , Ratones SCID , Pronóstico , Neoplasias de la Próstata/mortalidad , Neoplasias de la Próstata/patología , Ligando RANK/metabolismo , Análisis de Supervivencia , Transcriptoma , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Transl Androl Urol ; 4(4): 438-54, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26816842

RESUMEN

Prostate cancer (PCa) metastasizes to bone and soft tissues, greatly decreasing quality of life, causing bone pain, skeletal complications, and mortality in PCa patients. While new treatment strategies are being developed, the molecular and cellular basis of PCa metastasis and the "cross-talk" between cancer cells and their microenvironment and crucial cell signaling pathways need to be successfully dissected for intervention. In this review, we introduce a new concept of the mechanism of PCa metastasis, the recruitment and reprogramming of bystander and dormant cells (DCs) by a population of metastasis-initiating cells (MICs). We provide evidence that recruited and reprogrammed DCs gain MICs phenotypes and can subsequently metastasize to bone and soft tissues. We show that MICs can also recruit and reprogram circulating tumor cells (CTCs) and this could contribute to cancer cell evolution and the acquisition of therapeutic resistance. We summarize relevant molecular signaling pathways, including androgen receptors (ARs) and their variants and growth factors (GFs) and cytokines that could contribute to the predilection of PCa for homing to bone and soft tissues. To understand the etiology and the biology of PCa and the effectiveness of therapeutic targeting, we briefly summarize the animal and cell models that have been employed. We also report our experience in the use of three-dimensional (3-D) culture and co-culture models to understand cell signaling networks and the use of these attractive tools to conduct drug screening exercises against already-identified molecular targets. Further research into PCa growth and metastasis will improve our ability to target cancer metastasis more effectively and provide better rationales for personalized oncology.

5.
Oncotarget ; 6(42): 44072-83, 2015 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-26624980

RESUMEN

FYN is a SRC family kinase (SFK) that has been shown to be up-regulated in human prostate cancer (PCa) tissues and cell lines. In this study, we observed that FYN is strongly up-regulated in human neuroendocrine PCa (NEPC) tissues and xenografts, as well as cells derived from a NEPC transgenic mouse model. In silico analysis of FYN expression in prostate cancer cell line databases revealed an association with the expression of neuroendocrine (NE) markers such as CHGA, CD44, CD56, and SYP. The loss of FYN abrogated the invasion of PC3 and ARCaPM cells in response to MET receptor ligand HGF. FYN also contributed to the metastatic potential of NEPC cells in two mouse models of visceral metastasis with two different cell lines (PC3 and TRAMPC2-RANKL). The activation of MET appeared to regulate neuroendocrine (NE) features as evidenced by increased expression of NE markers in PC3 cells with HGF. Importantly, the overexpression of FYN protein in DU145 cells was directly correlated with the increase of CHGA. Thus, our data demonstrated that the neuroendocrine differentiation that occurs in PCa cells is, at least in part, regulated by FYN kinase. Understanding the role of FYN in the regulation of NE markers will provide further support for ongoing clinical trials of SFK and MET inhibitors in castration-resistant PCa patients.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Diferenciación Celular , Movimiento Celular , Neoplasias Hepáticas/enzimología , Tumores Neuroendocrinos/enzimología , Neoplasias de la Próstata/enzimología , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Animales , Biomarcadores de Tumor/genética , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular , Cromogranina A/metabolismo , Simulación por Computador , Bases de Datos Genéticas , Relación Dosis-Respuesta a Droga , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundario , Masculino , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos , Invasividad Neoplásica , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/secundario , Fenotipo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-fyn/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal , Factores de Tiempo , Transfección , Carga Tumoral , Regulación hacia Arriba
6.
Mol Cancer Ther ; 9(12): 3289-301, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21159612

RESUMEN

As p53 loss of function (LOF) confers high-level drug resistance in neuroblastoma, p53-independent therapies might have superior activity in recurrent neuroblastoma. We tested the activity of vorinostat, a histone deacetylase inhibitor, and flavopiridol, a pan-Cdk inhibitor, in a panel of multidrug-resistant neuroblastoma cell lines that included lines with wild-type (wt) and transcriptionally active TP53 (n = 3), mutated (mt), and LOF TP53 (n = 4) or p14(ARF) deletion (n = 1). The combination of vorinostat and flavopiridol was synergistic and significantly more cytotoxic (P < 0.001) in cell lines with p53-LOF and in the clones stably transfected with dominant-negative p53 plasmids. Cell cycle analysis by flow cytometry showed prominent cell-cycle arrest in G(2)/M (37%) for a cell line with wt TP53 (SK-N-RA) at 16 to 20 hours, while cells with mt TP53 (CHLA-90) slipped into sub-G(1) at 6 to 24 hours (25%-40% specific cell death). The morphological hallmarks of mitotic cell death, including defective spindle formation and abnormal cytokinesis, were detected by confocal microscopy after the treatment with vorinostat + flavopiridol combination in CHLA-90. The combination caused reduction in the expression of G(2)/M proteins (cyclin B1, Mad2, MPM2) in 2 cell lines with mt TP53 but not in those with wt TP53. Plk1 expression was reduced in all treated lines. Small interfering RNA knockdown of Mad2 and cyclin B1 or Plk1 synergistically reduced the clonogenicity of CHLA-90 cells. The combination of HDAC inhibitor and flavopiridol may be a unique approach to treating neuroblastomas with p53 LOF, one that evokes induction of mitotic failure.


Asunto(s)
Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Flavonoides/farmacología , Ácidos Hidroxámicos/farmacología , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/patología , Piperidinas/farmacología , Proteína p53 Supresora de Tumor/genética , Biomarcadores de Tumor/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Sinergismo Farmacológico , Flavonoides/química , Flavonoides/uso terapéutico , Fase G2/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/química , Ácidos Hidroxámicos/uso terapéutico , Mitosis/efectos de los fármacos , Proteínas Mutantes/metabolismo , Mutación/genética , Piperidinas/química , Piperidinas/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Vorinostat
7.
J Biol Chem ; 280(17): 16821-8, 2005 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-15664986

RESUMEN

TFII-I is a signal-induced multi-functional transcription factor that has recently been implicated as a regulatory component of the endoplasmic reticulum (ER) stress response. TFII-I acts through ER stress-induced binding to the ER stress element, which is highly conserved in promoters of ER stress-inducible genes such as Grp78/BiP. Interestingly, its tyrosine phosphorylation sites are required for its activation of the Grp78 promoter. Toward understanding the link between TFII-I, the tyrosine kinase signaling pathway, and Grp78 activation, we discovered that Tg stress induces a dramatic increase of TFII-I phosphorylation at Tyr248 and localization of this form of TFII-I to the nucleus. Chromatin immunoprecipitation analysis further reveals enhanced TFII-I binding to the Grp78 promoter in vivo upon ER stress. Previously, we reported that genistein, a general inhibitor of tyrosine kinase, could suppress ER stress induction of Grp78 by inhibiting complex formation on the ER stress element; however, the mechanism is not known. Consistent with TFII-I being a target of genistein suppression, we observed that genistein could suppress Tg stress-induced phosphorylation of TFII-I. We further demonstrate that c-Src, which is one of kinases identified to mediate phosphorylation of TFII-I at Tyr248, is activated by Tg stress and is able to stimulate the Grp78 promoter activity. Lastly, using stable cell lines with suppressed TFII-I levels, we show that TFII-I is required for optimal induction of Grp78 by ER stress. Our studies provide a molecular link that connects the c-Src tyrosine kinase transduction pathway to ER stress-induced transcriptional activation of Grp78 mediated by TFII-I.


Asunto(s)
Retículo Endoplásmico/metabolismo , Proteínas de Choque Térmico/fisiología , Chaperonas Moleculares/fisiología , Factores de Transcripción TFII/fisiología , Transcripción Genética , Tirosina/metabolismo , Animales , Northern Blotting , Western Blotting , Proteína Tirosina Quinasa CSK , Línea Celular , Inmunoprecipitación de Cromatina , Chaperón BiP del Retículo Endoplásmico , Genes Reporteros , Genisteína/farmacología , Proteínas de Choque Térmico/metabolismo , Humanos , Cinética , Ratones , Microscopía Fluorescente , Modelos Biológicos , Modelos Genéticos , Chaperonas Moleculares/metabolismo , Células 3T3 NIH , Fosforilación , Plásmidos/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal , Factores de Tiempo , Activación Transcripcional , Transfección , Tirosina/química , Familia-src Quinasas
8.
Biochem Biophys Res Commun ; 306(4): 912-7, 2003 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-12821128

RESUMEN

A novel truncated form of Bcl-2, termed Bcl-2psi, was discovered in invasive prostate cancer cells, using laser capture microdissection, RNA-polymerase cycling reaction, and microarray analysis. The expression of Bcl-2psi increased prior to metastasis in higher-grade prostate cancer. The immunoreactive Bcl-2psi was specifically identified in higher-grade prostate cancer cells. These findings suggest that Bcl-2psi may be a potential pre-metastatic marker for detection, diagnosis, and therapy during the initiation of metastasis in prostate cancer.


Asunto(s)
Biomarcadores de Tumor , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/química , Secuencia de Bases , Northern Blotting , Western Blotting , Disección , Electroforesis en Gel de Poliacrilamida , Biblioteca de Genes , Humanos , Inmunohistoquímica , Rayos Láser , Masculino , Datos de Secuencia Molecular , Mutación , Metástasis de la Neoplasia , Análisis de Secuencia por Matrices de Oligonucleótidos , Estructura Terciaria de Proteína , ARN/metabolismo , Homología de Secuencia de Ácido Nucleico , Células Tumorales Cultivadas
9.
J Biol Chem ; 279(12): 11354-63, 2004 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-14699159

RESUMEN

ATF6 is a key transcriptional activator of the unfolded protein response (UPR), which allows mammalian cells to maintain cellular homeostasis when they are subjected to a variety of environmental and physiological stresses that target the endoplasmic reticulum (ER). ATF6, a 90-kDa ER transmembrane protein, contains three evolutionarily conserved N-linked glycosylation sites within its carboxyl luminal domain. Although it is well established that p90ATF6 activation requires transit from the ER to the Golgi, where it is cleaved by the S1P/S2P protease system to generate a nuclear form p60ATF6 that acts as a transcriptional activator, the functional significance of p90ATF6 N-linked glycosylation is unknown. Here we show that ER Ca(2+) depletion stress, a triggering mechanism for the UPR, induces the formation of ATF6(f), which represents de novo partial glycosylation of newly synthesized p90ATF6. By mutating a single amino acid within the N-linked glycosylation site closest to the carboxyl terminus of p90ATF6, we recreated ATF6(f). This mutation sharply reduces p90ATF6 association with calreticulin, a major Ca(2+)-binding chaperone for N-glycoprotein. We further determined that ATF6(f) exhibits a faster rate of constitutive transport to the Golgi, resulting in a higher level of p60ATF6 in the nucleus and stronger transactivating activity in the absence of ER stress. Additional analysis of p90ATF6 mutants targeting single or multiple N-glycosylation sites also showed higher constitutive transactivating activity than wild type ATF6. Because accumulation of underglycosylated proteins in the ER is a potent inducer for the UPR, these studies uncover a novel mechanism whereby the glycosylation status of p90ATF6 can serve as a sensor for ER homeostasis, resulting in ATF6 activation to trigger the UPR.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/metabolismo , Factor de Transcripción Activador 6 , Animales , Secuencia de Bases , Calcio/metabolismo , Línea Celular , Cartilla de ADN , Proteínas de Unión al ADN/genética , Retículo Endoplásmico/metabolismo , Técnica del Anticuerpo Fluorescente , Glicosilación , Homeostasis , Humanos , Mutación , Desnaturalización Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA