Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 295(25): 8550-8559, 2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32376689

RESUMEN

The transcriptional coactivator YAP1 (yes-associated protein 1) regulates cell proliferation, cell-cell interactions, organ size, and tumorigenesis. Post-transcriptional modifications and nuclear translocation of YAP1 are crucial for its nuclear activity. The objective of this study was to elucidate the mechanism by which the steroid hormone androgen regulates YAP1 nuclear entry and functions in several human prostate cancer cell lines. We demonstrate that androgen exposure suppresses the inactivating post-translational modification phospho-Ser-127 in YAP1, coinciding with increased YAP1 nuclear accumulation and activity. Pharmacological and genetic experiments revealed that intact androgen receptor signaling is necessary for androgen's inactivating effect on phospho-Ser-127 levels and increased YAP1 nuclear entry. We also found that androgen exposure antagonizes Ser/Thr kinase 4 (STK4/MST1) signaling, stimulates the activity of protein phosphatase 2A, and thereby attenuates the phospho-Ser-127 modification and promotes YAP1 nuclear localization. Results from quantitative RT-PCR and CRISPR/Cas9-aided gene knockout experiments indicated that androgen differentially regulates YAP1-dependent gene expression. Furthermore, an unbiased computational analysis of the prostate cancer data from The Cancer Genome Atlas revealed that YAP1 and androgen receptor transcript levels correlate with each other in prostate cancer tissues. These findings indicate that androgen regulates YAP1 nuclear localization and its transcriptional activity through the androgen receptor-STK4/MST1-protein phosphatase 2A axis, which may have important implications for human diseases such as prostate cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Andrógenos/farmacología , Núcleo Celular/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos , Línea Celular Tumoral , Bases de Datos Genéticas , Expresión Génica/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Fosforilación/efectos de los fármacos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteína Fosfatasa 2/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Señalizadoras YAP
2.
Prostate ; 80(5): 412-423, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31995655

RESUMEN

BACKGROUND: Mammalian target of rapamycin (mTOR) is a downstream substrate activated by PI3K/AKT pathway and it is essential for cell migration. It exists as two complexes: mTORC1 and mTORC2. mTORC1 is known to be regulated by active AKT, but the activation of mTORC2 is poorly understood. In this study, we investigated the roles and differential activation of the two mTOR complexes during cell migration in prostate cancer cells. METHODS: We used small interfering RNA to silence the expression of Rac1 and the main components of mTOR complexes (regulatory associated protein of mTOR [RAPTOR] and rapamycin-insensitive companion of mTOR [RICTOR]) in LNCaP, DU145, and PC3 prostate cancer cell lines. We performed transwell migration assay to evaluate the migratory capability of the cells, and Western blot analysis to study the activation levels of mTOR complexes. RESULTS: Specific knockdown of RAPTOR and RICTOR caused a decrease of cell migration, suggesting their essential role in prostate cancer cell movement. Furthermore, epidermal growth factor (EGF) treatments induced the activation of both the mTOR complexes. Lack of Rac1 activity in prostate cancer cells blocked EGF-induced activation of mTORC2, but had no effect on mTORC1 activation. Furthermore, the overexpression of constitutively active Rac1 resulted in significant increase in cell migration and activation of mTORC2 in PC3 cells, but had no effect on mTORC1 activation. Active Rac1 was localized in the plasma membrane and was found to be in a protein complex, with RICTOR, but not RAPTOR. CONCLUSION: We suggest that EGF-induced activation of Rac1 causes the activation of mTORC2 via RICTOR. This mechanism plays a critical role in prostate cancer cell migration.


Asunto(s)
Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Aminoquinolinas/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Factor de Crecimiento Epidérmico/farmacología , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Células PC-3 , Pirimidinas/farmacología , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Proteína Asociada al mTOR Insensible a la Rapamicina/deficiencia , Proteína Asociada al mTOR Insensible a la Rapamicina/genética , Proteína Asociada al mTOR Insensible a la Rapamicina/metabolismo , Proteína Reguladora Asociada a mTOR/deficiencia , Proteína Reguladora Asociada a mTOR/genética , Proteína Reguladora Asociada a mTOR/metabolismo , Sirolimus/farmacología , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína de Unión al GTP rac1/metabolismo
3.
Carcinogenesis ; 39(4): 546-555, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29474521

RESUMEN

Epidemiological studies show that the incidence and mortality rates of prostate cancer (PCa) are significantly higher in African-American (AA) men when compared with Caucasian (CA) men in the United States. Transforming growth factor ß (TGFß) signaling pathway is linked to health disparities in AAs. Recent studies suggest a role of TGFß3 in cancer metastases and its effect on the migratory and invasive behavior; however, its role in PCa in AA men has not been studied. We determined the circulating levels of TGFß3 in AA and CA men diagnosed with PCa using ELISA. We analyzed serum samples from both AA and CA men diagnosed with and without PCa. We show that AA PCa patients had higher levels of TGFß3 protein compared with AA controls and CA patients. In fact, TGFß3 protein levels in serum were higher in AA men without PCa compared with the CA population, which may correlate with more aggressive disease seen in AA men. Studies on AA-derived PCa cell lines revealed that TGFß3 protein levels were also higher in these cells compared with CA-derived PCa cell lines. Our studies also reveal that TGFß does not inhibit cell proliferation in AA-derived PCa cell lines, but it does induce migration and invasion through activation of PI3K pathway. We suggest that increased TGFß3 levels are responsible for development of aggressive PCa in AA patients as a consequence of development of resistance to inhibitory effects of TGFß on cell proliferation and induction of invasive metastatic behavior.


Asunto(s)
Biomarcadores de Tumor/sangre , Neoplasias de la Próstata/epidemiología , Neoplasias de la Próstata/metabolismo , Factor de Crecimiento Transformador beta3/sangre , Negro o Afroamericano , Anciano , Movimiento Celular/fisiología , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Invasividad Neoplásica/patología , Neoplasias de la Próstata/patología , Transducción de Señal/fisiología , Población Blanca
4.
J Cell Physiol ; 234(1): 802-815, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-30078221

RESUMEN

Tumor cell motility is the essential step in cancer metastasis. Previously, we showed that oxytocin and epidermal growth factor (EGF) effects on cell migration in prostate cancer cells require Giα2 protein. In the current study, we investigated the interactions among G-protein coupled receptor (GPCR), Giα2, PI3-kinase, and Rac1 activation in the induction of migratory and invasive behavior by diverse stimuli. Knockdown and knockout of endogenous Giα2 in PC3 cells resulted in attenuation of transforming growth factor ß1 (TGFß1), oxytocin, SDF-1α, and EGF effects on cell migration and invasion. In addition, knockdown of Giα2 in E006AA cells attenuated cell migration and overexpression of Giα2 in LNCaP cells caused significant increase in basal and EGF-stimulated cell migration. Pretreatment of PC3 cells with Pertussis toxin resulted in attenuation of TGFß1- and oxytocin-induced migratory behavior and PI3-kinase activation without affecting EGF-induced PI3-kinase activation and cell migration. Basal- and EGF-induced activation of Rac1 in PC3 and DU145 cells were not affected in cells after Giα2 knockdown. On the other hand, Giα2 knockdown abolished the migratory capability of PC3 cells overexpressing constitutively active Rac1. The knockdown or knockout of Giα2 resulted in impaired formation of lamellipodia at the leading edge of the migrating cells. We conclude that Giα2 protein acts at two different levels which are both dependent and independent of GPCR signaling to induce cell migration and invasion in prostate cancer cells and its action is downstream of PI3-kinase-AKT-Rac1 axis.


Asunto(s)
Movimiento Celular/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Neoplasias de la Próstata/genética , Proteína de Unión al GTP rac1/genética , Línea Celular Tumoral , Proliferación Celular/genética , Quimiocina CXCL12/genética , Factor de Crecimiento Epidérmico/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Proteína Oncogénica v-akt/genética , Oxitocina/genética , Fosfatidilinositol 3-Quinasas/genética , Fosforilación , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/patología , Factor de Crecimiento Transformador beta1/genética
5.
Prostate ; 78(5): 377-389, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29341212

RESUMEN

BACKGROUND: Transforming growth factor-ß (TGF-ß) acts as a tumor suppressor in normal epithelial cells but as a tumor promoter in advanced prostate cancer cells. PI3-kinase pathway mediates TGF-ß effects on prostate cancer cell migration and invasion. PTEN inhibits PI3-kinase pathway and is frequently mutated in prostate cancers. We investigated possible role(s) of PTEN in TGF-ß effects on proliferation and migration in prostate cancer cells. METHODS: Expression of PTEN mRNA and proteins were determined using RT-PCR and Western blotting in RWPE1 and DU145 cells. We also studied the role of PTEN in TGF-ß effects on cell proliferation and migration in DU145 cells after transient silencing of endogenous PTEN. Conversely, we determined the role of PTEN in cell proliferation and migration after over-expression of PTEN in PC3 cells which lack endogenous PTEN. RESULTS: TGF-ß1 and TGF-ß3 had no effect on PTEN mRNA levels but both isoforms increased PTEN protein levels in DU145 and RWPE1 cells indicating that PTEN may mediate TGF-ß effects on cell proliferation. Knockdown of PTEN in DU145 cells resulted in significant increase in cell proliferation which was not affected by TGF-ß isoforms. PTEN overexpression in PC3 cells inhibited cell proliferation. Knockdown of endogenous PTEN enhanced cell migration in DU145 cells, whereas PTEN overexpression reduced migration in PC3 cells and reduced phosphorylation of AKT in response to TGF-ß. CONCLUSION: We conclude that PTEN plays a role in inhibitory effects of TGF-ß on cell proliferation whereas its absence may enhance TGF-ß effects on activation of PI3-kinase pathway and cell migration.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Fosfohidrolasa PTEN/metabolismo , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Factor de Crecimiento Transformador beta1/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Masculino , Fosfohidrolasa PTEN/biosíntesis , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas Recombinantes/farmacología , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Factor de Crecimiento Transformador beta3/antagonistas & inhibidores , Factor de Crecimiento Transformador beta3/farmacología
6.
J Biol Chem ; 291(34): 17964-76, 2016 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-27358408

RESUMEN

TGF-ß inhibits proliferation of prostate epithelial cells. However, prostate cancer cells in advanced stages become resistant to inhibitory effects of TGF-ß. The intracellular signaling mechanisms involved in differential effects of TGF-ß during different stages are largely unknown. Using cell line models, we have shown that TGF-ß inhibits proliferation in normal (RWPE-1) and prostate cancer (DU145) cells but does not have any effect on proliferation of prostate cancer (PC3) cells. We have investigated the role of Jun family proteins (c-Jun, JunB, and JunD) in TGF-ß effects on cell proliferation. Jun family members were expressed at different levels and responded differentially to TGF-ß treatment. TGF-ß effects on JunD protein levels, but not mRNA levels, correlated with its effects on cell proliferation. TGF-ß induced significant reduction in JunD protein in RWPE-1 and DU145 cells but not in PC3 cells. Selective knockdown of JunD expression using siRNA in DU145 and PC3 cells resulted in significant reduction in cell proliferation, and forced overexpression of JunD increased the proliferation rate. On the other hand, knockdown of c-Jun or JunB had little, if any, effect on cell proliferation; overexpression of c-Jun and JunB decreased the proliferation rate in DU145 cells. Further studies showed that down-regulation of JunD in response to TGF-ß treatment is mediated via the proteasomal degradation pathway. In conclusion, we show that specific Jun family members exert differential effects on proliferation in prostate cancer cells in response to TGF-ß, and inhibition of cell proliferation by TGF-ß requires degradation of JunD protein.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-jun/biosíntesis , Factor de Crecimiento Transformador beta/farmacología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Masculino , Neoplasias de la Próstata/genética , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-jun/genética
7.
Prostate ; 77(1): 72-81, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27604827

RESUMEN

BACKGROUND: Activator Protein-1 (AP-1) family (cJun, JunB, JunD, cFos, FosB, Fra1, and Fra2) plays a central role in the transcriptional regulation of many genes that are associated with cell proliferation, differentiation, migration, metastasis, and survival. Many oncogenic signaling pathways converge at the AP-1 transcription complex. Transforming growth factor beta (TGF-ß) is a multifunctional regulatory cytokine that regulates many aspects of cellular function, including cellular proliferation, differentiation, migration, apoptosis, adhesion, angiogenesis, immune surveillance, and survival. METHODS: This study investigated, the role of FOS proteins in TGF-ß signaling in prostate cancer cell proliferation, migration, and invasion. Steady state expression levels of FOS mRNA and proteins were determined using RT-PCR and western blotting analyses. DU145 and PC3 prostate cancer cells were exposed to TGF-ß1 at varying time and dosage, RT-PCR, western blot, and immunofluorescence analyses were used to determine TGF-ß1 effect on FOS mRNA and protein expression levels as well as FosB subcellular localization. Transient silencing of FosB protein was used to determine its role in cell proliferation, migration, and invasion. RESULTS: Our data show that FOS mRNA and proteins were differentially expressed in human prostate epithelial (RWPE-1) and prostate cancer cell lines (LNCaP, DU145, and PC3). TGF-ß1 induced the expression of FosB at both the mRNA and protein levels in DU145 and PC3 cells, whereas cFos and Fra1 were unaffected. Immunofluorescence analysis showed an increase in the accumulation of FosB protein in the nucleus of PC3 cells after treatment with exogenous TGF-ß1. Selective knockdown of endogenous FosB by specific siRNA did not have any effect on cell proliferation in PC3 and DU145 cells. However, basal and TGF-ß1- and EGF-induced cell migration was significantly reduced in DU145 and PC3 cells lacking endogenous FosB. TGF-ß1- and EGF-induced cell invasion were also significantly decreased after FosB knockdown in PC3 cells. CONCLUSION: Our data suggest that FosB is required for migration and invasion in prostate cancer cells. We also conclude that TGF-ß1 effect on prostate cancer cell migration and invasion may be mediated through the induction of FosB. Prostate 77:72-81, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Movimiento Celular/fisiología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Factor de Crecimiento Transformador beta/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Masculino , Invasividad Neoplásica/patología , Proteínas Proto-Oncogénicas c-fos/agonistas
8.
Mol Carcinog ; 54(8): 618-31, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24395385

RESUMEN

p66Shc functions as a longevity protein in murine and exhibits oxidase activity in regulating diverse biological activities. In this study, we investigated the role of p66Shc protein in regulating ovarian cancer (OCa) cell proliferation. Among three cell lines examined, the slowest growing OVCAR-3 cells have the lowest level of p66Shc protein. Transient transfection with p66Shc cDNA expression vector in OVCAR-3 cells increases cell proliferation. Conversely, knock-down of p66Shc by shRNA in rapidly growing SKOV-3 cells results in decreased cell growth. In estrogen (E2)-treated CaOV-3 cells, elevated p66Shc protein level correlates with ROS level, ErbB-2 and ERK/MAPK activation, and cell proliferation. Further, the E2-stimulated proliferation of CaOV-3 cells was blocked by antioxidants and ErbB-2 inhibitor. Additionally, in E2-stimulated cells, the tartrate-sensitive, but not the tartrate-resistant, phosphatase activity decreases; concurrently, the tyrosine phosphorylation of ErbB-2 increases. Conversely, inhibition of phosphatase activity by L(+)-tartrate treatment increases p66Shc protein level, ErbB-2 tyrosine phosphorylation, ERK/MAPK activation, and cell growth. Further, inhibition of the ERK/MAPK pathway by PD98059 blocks E2-induced ERK/MAPK activation and cell proliferation in CaOV-3 cells. Moreover, immunohistochemical analyses showed that the p66Shc protein level was significantly higher in cancerous cells than in noncancerous cells in archival OCa tissues (n = 76; P = 0.00037). These data collectively indicate that p66Shc protein plays a critical role in up-regulating OCa progression.


Asunto(s)
Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Receptor ErbB-2/metabolismo , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Estrógenos/farmacología , Femenino , Flavonoides/farmacología , Humanos , Neoplasias Ováricas/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Adaptadoras de la Señalización Shc/genética , Transducción de Señal/efectos de los fármacos , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Regulación hacia Arriba
9.
J Nanosci Nanotechnol ; 15(8): 5733-40, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26369146

RESUMEN

Gold nanoparticles (AuNPs) have been chemically functionalized onto multiwalled carbon nanotubes (MWCNT) through a metallopolymer linker-bis (2,2':6'2"-terpyridine) ruthenium(II)-connected diblock poly(N-isopropyacryamide). A "nano-snowflower" pattern was formed by self-assembly MWCNT-AuNP nanocomposite with anti-DNP IgE antibody. MWCNT-AuNP nanohybrid has unique biocompatibility and electronic current-voltage properties. This nanohybrid shows the potential application for IgE biosensor to diagnose cancer cells. We represent a step towards building complex electronic circuits response by providing molecular recognition properties.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Oro/química , Inmunoensayo/instrumentación , Inmunoglobulina E/inmunología , Nanopartículas del Metal/química , Rutenio/química , Adsorción , Anticuerpos Monoclonales/química , Conductometría/instrumentación , Conductividad Eléctrica , Diseño de Equipo , Análisis de Falla de Equipo , Inmunoglobulina E/análisis , Ensayo de Materiales , Nanocompuestos/química , Nanocompuestos/ultraestructura , Tamaño de la Partícula , Unión Proteica , Propiedades de Superficie
10.
J BUON ; 20(5): 1250-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26537072

RESUMEN

PURPOSE: JAK/STAT is an evolutionarily conserved pathway and very important for second messenger system. This pathway is important in malignant transformation and accumulated evidence indicates that this pathway is involved in tumorigenesis and progression of several cancers. It was possible to assume that activation of JAK/STAT pathway is associated with increase in the expressions of ICAM/1 and VCAM-1. In this study we hypothesized that when cells were maintained as spheroids or monolayers, the structure of cancer stem cells (CSCs) could show differentiation when compared with non-CSCs. METHODS: DU-145 human prostate cancer cells were cultured using the Ege University molecular embryology laboratory medium supplemented with 10% fetal bovine serum. Clusters of differentiation 133 (CD133)(+high)/CD44(+high) prostate CSCs were isolated from the DU145 cell line by using BD FACSAria. CD133//CD44+ CSCs were cultured until confluent with 3% noble agar. The expression of these proteins in CSCs and non-CSCs was analyzed by immunohistochemistry. RESULTS: Different expression profiles were observed in the conventional two-dimensional (2D) and three-dimensional (3D) experimental model system when CSCs and non-CSCs were compared. Human prostate CSCs exhibited intense ICAM-1 and VCAM-1 immunoreaction when compared with non-CSCs. These findings were supported by the fact that VCAM-1 on the surface of cancer cells binds to its counterreceptor, the α4ß1 integrin (also known as very-late antigen, VLA-4), on metastasis-associated macrophages, triggering VCAM-1-mediated activation of the phosphoinositide 3-kinase growth and survival pathway in cancer cells. CONCLUSIONS: The results of this study showed that changes in JAK/STAT pathway are related with adhesion molecules and could affect cancer progression.


Asunto(s)
Molécula 1 de Adhesión Intercelular/fisiología , Quinasas Janus/fisiología , Células Madre Neoplásicas/patología , Neoplasias de la Próstata/patología , Factores de Transcripción STAT/fisiología , Transducción de Señal/fisiología , Esferoides Celulares/patología , Molécula 1 de Adhesión Celular Vascular/fisiología , Línea Celular Tumoral , Humanos , Inmunohistoquímica , Molécula 1 de Adhesión Intercelular/análisis , Masculino , Molécula 1 de Adhesión Celular Vascular/análisis
11.
J Pharm Biomed Anal ; 245: 116183, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38744031

RESUMEN

A sensitive and selective LC-MS/MS method was developed and validated for the quantitation of a novel Gαi2 inhibitor, GT-14, in rat plasma using a SCIEX 6500+ triple QUAD LC-MS system equipped with an ExionLC UHPLC unit. GT-14 (m/z 265.2 → 134.1) and griseofulvin (Internal Standard, IS) (m/z 353.1 → 285.1) were detected in a positive mode by electrospray ionization (ESI) using multiple reaction monitoring (MRM). The assay was linear in the concentration range of 0.78-1000 ng/mL in rat plasma. Both accuracy and precision values were within the acceptance criteria of ±15 %, as established by FDA guidance. The matrix effect was negligible from plasma, with signal percentages of 98.5-106.9 %. The mean recovery was 104.5 %, indicating complete extraction of GT-14 from plasma. GT-14 was found to be stable under different experimental conditions. The validated method was successfully applied to evaluate plasma protein binding and in vivo pharmacokinetics of GT-14 in rats.


Asunto(s)
Ratas Sprague-Dawley , Espectrometría de Masas en Tándem , Animales , Espectrometría de Masas en Tándem/métodos , Ratas , Reproducibilidad de los Resultados , Masculino , Cromatografía Líquida de Alta Presión/métodos , Espectrometría de Masa por Ionización de Electrospray/métodos , Griseofulvina/farmacocinética , Griseofulvina/sangre , Unión Proteica , Cromatografía Liquida/métodos , Cromatografía Líquida con Espectrometría de Masas
12.
Cancers (Basel) ; 16(2)2024 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-38254786

RESUMEN

We have previously shown that heterotrimeric G-protein subunit alphai2 (Gαi2) is essential for cell migration and invasion in prostate, ovarian and breast cancer cells, and novel small molecule inhibitors targeting Gαi2 block its effects on migratory and invasive behavior. In this study, we have identified potent, metabolically stable, second generation Gαi2 inhibitors which inhibit cell migration in prostate cancer cells. Recent studies have shown that chemotherapy can induce the cancer cells to migrate to distant sites to form metastases. In the present study, we determined the effects of taxanes (docetaxel), anti-androgens (enzalutamide and bicalutamide) and histone deacetylase (HDAC) inhibitors (SAHA and SBI-I-19) on cell migration in prostate cancer cells. All treatments induced cell migration, and simultaneous treatments with new Gαi2 inhibitors blocked their effects on cell migration. We concluded that a combination treatment of Gαi2 inhibitors and chemotherapy could blunt the capability of cancer cells to migrate and form metastases.

13.
Prostate ; 73(6): 624-33, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23060149

RESUMEN

BACKGROUND: In prostate cancer cells, transforming growth factor ß (TGFß) inhibits proliferation in earlier stages of the disease; however, the cancer cells become refractory to growth inhibitory effects in advanced stages where TGFß promotes cancer progression and metastasis. Inhibitor of differentiation (Id) family of closely related proteins (Id1-Id4) are dominant negative regulators and basic helix loop helix (bHLH) transcription factors and in general promote proliferation, and inhibit differentiation. In the present study, we have investigated the role of Id1 and Id3 proteins in the growth inhibitory effects of TGFß on prostate cancer cells. METHODS: The effect of TGF ß on proliferation and Id1 and Id3 expression were investigated in PZ-HPV7, DU145, and PC3 cells. Id1 silencing through siRNA was also used in DU145 and PC3 cells to examine its role in anti-proliferative and migratory effects of TGFß. RESULTS: TGFß increased expression of Id1 and Id3 in all cell lines followed by a later down regulation of Id1 in PZ-HPV7 expression and DU145 cells but not in PC3 cells. Id3 expression remained elevated in all three cell lines. This loss of Id1 protein correlated with an increase of CDKNI p21. Id1 knockdown in both DU145 and PC3 cells resulted in decreased proliferation. However, while TGFß caused a further decrease in proliferation of DU145, but had no further effects in PC3 cells. Knockdown of Id1 or Id3 inhibited TGFß1induced migration in PC3 cells. CONCLUSIONS: These findings suggest an essential role of Id1 and Id3 in TGFß1 effects on proliferation and migration in prostate cancer cells.


Asunto(s)
Movimiento Celular/fisiología , Proteína 1 Inhibidora de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/genética , Proteínas de Neoplasias/genética , Neoplasias de la Próstata , Factor de Crecimiento Transformador beta1/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Células Epiteliales/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Inhibidores de Crecimiento/metabolismo , Inhibidores de Crecimiento/farmacología , Humanos , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Proteínas Inhibidoras de la Diferenciación/metabolismo , Masculino , Proteínas de Neoplasias/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , ARN Interferente Pequeño/genética , Factor de Crecimiento Transformador beta1/farmacología
14.
Carcinogenesis ; 33(11): 2054-64, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22843506

RESUMEN

Transforming growth factor-beta (TGF-ß) signaling pathways contain both tumor suppressor and tumor promoting activities. We have demonstrated that Nodal, another member of the TGF-ß superfamily, and its receptors are expressed in prostate cancer cells. Nodal and TGF-ß exerted similar biological effects on prostate cells; both inhibited proliferation in WPE, RWPE1 and DU145 cells, whereas neither had any effect on the proliferation of LNCaP or PC3 cells. Interestingly, Nodal and TGF-ß induced migration in PC3 cells, but not in DU145 cells. TGF-ß induced predominantly phosphorylation of Smad3, whereas Nodal induced phosphorylation of only Smad2. We also determined the expression and differential role of Ski, a corepressor of Smad2/3, in Nodal and TGF-ß signaling in prostate cancer cells. Similar levels of Ski mRNA were found in several established prostate cell lines; however, high levels of Ski protein were only detected in prostate cancer cells and prostate cancer tissue samples. Exogenous Nodal and TGF-ß had no effects on Ski mRNA levels. On the other hand, TGF-ß induced a rapid degradation of Ski protein mediated by the proteasomal pathway, whereas Nodal had no effect on Ski protein. Reduced Ski levels correlated with increased basal and TGF-ß-induced Smad2/3 phosphorylation. Knockdown of endogenous Ski reduced proliferation in DU145 cells and enhanced migration of PC3 cells. We conclude that high levels of Ski expression in prostate cancer cells may be responsible for repression of TGF-ß and Smad3 signaling, but Ski protein levels do not influence Nodal and Smad2 signaling.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteína Nodal/metabolismo , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Western Blotting , Movimiento Celular , Proliferación Celular , Células Cultivadas , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Masculino , Proteína Nodal/genética , Fosforilación , Próstata/patología , Unión Proteica , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Proteína Smad2/genética , Proteína smad3/genética , Factor de Crecimiento Transformador beta/genética , Cicatrización de Heridas
15.
Prostate ; 71(10): 1084-96, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21557273

RESUMEN

BACKGROUND: Nodal, a TGFß like growth factor, functions as an embryonic morphogen that maintains the pluripotency of embryonic stem cells. Nodal has been implicated in cancer progression; however, there is no information on expression and functions of Nodal in prostate cancer. In this study, we have investigated the expression of Nodal, its receptors, and its effects on proliferation and migration of human prostate cells. METHODS: RT-PCR, qPCR, and Western blot analyses were performed to analyze expression of Nodal and Nodal receptors and its effects on phosphorylation of Smad2/3 in prostate cells. The effects on proliferation and migration were determined by (3) H-Thymidine incorporation and cell migration assays in the presence or absence of Nodal receptor inhibitor (SB431542). RESULTS: Nodal was highly expressed in WPE, DU145, LNCaP, and LNCaP-C81 cells with low expression in RWPE1 and RWPE2 cells, but not in PREC, PC3 and PC3M cells. Nodal receptors are expressed at varying levels in all prostate cells. Treatment with exogenous Nodal induced phosphorylation of Smad2/3 in WPE, DU145, and PC3 cells, which was blocked by SB431542. Nodal dose-dependently inhibited proliferation of WPE, RWPE1 and DU145 cells, but not LNCaP and PC3 cells. Nodal induced cell migration in PC3 cells, which was inhibited by SB431542; Nodal had no effect on cell migration in WPE and DU145 cells. The effects of Nodal on cell proliferation and migration are mediated via ALK4 and ActRII/ActRIIB receptors and Smad 2/3 phosphorylation. CONCLUSIONS: Nodal may function as an autocrine regulator of proliferation and migration of prostate cancer cells.


Asunto(s)
Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Proteína Nodal/metabolismo , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Análisis de Varianza , Western Blotting , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Células Madre Neoplásicas/patología , Proteína Nodal/genética , Proteína Nodal/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Próstata/efectos de los fármacos , Próstata/patología , Neoplasias de la Próstata/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo
16.
Am J Clin Exp Urol ; 9(4): 318-328, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34541030

RESUMEN

During prostate cancer progression, TGF-ß acts as both a tumor suppressor and tumor promoter. TGF-ß inhibits cell proliferation in normal and early-stage prostate cancer cells, but during later stages of the disease the cancer cells develop resistance to inhibitory effects on cell proliferation. In these cells, TGF-ß promotes cancer progression due to its effects on epithelial to mesenchymal transition (EMT), cell migration and invasion, and immune suppression. The intracellular mechanisms involved in the development of resistance to TGF-ß effects on cell proliferation are largely unknown. In this review, we summarized the roles of several intracellular proteins including PTEN, Id1 and JunD, which may play a role in this transition. The role of Ski/SnoN proteins in inhibition of Smad2/3 signaling is highlighted.

17.
Artículo en Inglés | MEDLINE | ID: mdl-33800316

RESUMEN

Inter-institutional collaborations and partnerships play fundamental roles in developing and diversifying the basic biomedical, behavioral, and clinical research enterprise at resource-limited, minority-serving institutions. In conjunction with the Research Centers in Minority Institutions (RCMI) Program National Conference in Bethesda, Maryland, in December 2019, a special workshop was convened to summarize current practices and to explore future strategies to strengthen and sustain inter-institutional collaborations and partnerships with research-intensive majority-serving institutions. Representative examples of current inter-institutional collaborations at RCMI grantee institutions are presented. Practical approaches used to leverage institutional resources through collaborations and partnerships within regional and national network programs are summarized. Challenges and opportunities related to such collaborations are provided.


Asunto(s)
Grupos Minoritarios , Investigación , Humanos , Maryland
18.
Cancers (Basel) ; 12(6)2020 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-32575572

RESUMEN

Heterotrimeric G-proteins are ubiquitously expressed in several cancers, and they transduce signals from activated G-protein coupled receptors. These proteins have numerous biological functions, and they are becoming interesting target molecules in cancer therapy. Previously, we have shown that heterotrimeric G-protein subunit alphai2 (Gαi2) has an essential role in the migration and invasion of prostate cancer cells. Using a structure-based approach, we have synthesized optimized small molecule inhibitors that are able to prevent specifically the activation of the Gαi2 subunit, keeping the protein in its inactive GDP-bound state. We observed that two of the compounds (13 and 14) at 10 µΜ significantly inhibited the migratory behavior of the PC3 and DU145 prostate cancer cell lines. Additionally, compound 14 at 10 µΜ blocked the activation of Gαi2 in oxytocin-stimulated prostate cancer PC3 cells, and inhibited the migratory capability of DU145 cells overexpressing the constitutively active form of Gαi2, under basal and EGF-stimulated conditions. We also observed that the knockdown or inhibition of Gαi2 negatively regulated migration of renal and ovarian cancer cell lines. Our results suggest that small molecule inhibitors of Gαi2 have potential as leads for discovering novel anti-metastatic agents for attenuating the capability of cancer cells to spread and invade to distant sites.

19.
Mol Cell Endocrinol ; 295(1-2): 115-20, 2008 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-18782595

RESUMEN

The proliferation and differentiation of normal prostate epithelial cells depends upon the action of androgens produced by the testis. Prostate cancers retain the ability to respond to androgens in the initial stages of cancer development, but progressively become independent of exogenous androgens in advanced stages of the disease while maintaining the expression of functional androgen receptor (AR). In the present study, we have determined the potential of prostate cancer cells to synthesize androgens from cholesterol which may be involved in intracrine regulation of AR in advanced stages of the disease. Established androgen-independent prostate cancer cell lines, PC3 and DU145 cells, expressed mRNA and proteins for scavenger receptor type B1 (SRB1), steroidogenic acute regulatory (StAR) protein, cytochrome P450 cholesterol side chain cleavage (P450scc), 3beta-hydroxysteroid dehydrogenase (3beta-HSD) and other enzymes involved in androgen biosynthesis. Expression of all these proteins and enzymes was significantly higher in the androgen-independent derivative of LNCaP prostate cancer cells (C81) than in the androgen-dependent cell line (C33). In serum-free cultures, the androgen-independent C81 cells secreted approximately 5-fold higher testosterone than C33 cells as determined in the conditioned media by immunoassays. These cells could also directly convert radioactive cholesterol into testosterone which was identified by thin layer chromatography. These results for the first time show that prostate cancer cells in advanced stages of the disease could synthesize androgens from cholesterol and hence are not dependent upon testicular and/or adrenal androgens.


Asunto(s)
Andrógenos/metabolismo , Colesterol/metabolismo , Neoplasias de la Próstata/metabolismo , Testosterona/metabolismo , Comunicación Autocrina , Línea Celular Tumoral , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , ARN Mensajero/metabolismo , Regulación hacia Arriba
20.
Mol Endocrinol ; 20(3): 608-18, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16269516

RESUMEN

Postnatal development and function of testicular Sertoli cells are regulated primarily by FSH. During this early period of development, estrogens play a role in proliferation of somatic cells, which contributes significantly to testicular development. Growth factors like epidermal growth factor (EGF) are produced in the testis and play a role in regulation of estradiol production and male fertility. Although these divergent factors modulate gonadal function, little is known about their mechanism of action in Sertoli cells. The present study investigates the intracellular events that take place down-stream of FSH and EGF receptors in Sertoli cells isolated from immature (10-d-old) rats, and examines which intracellular signals may be involved in their effects on aromatase activity and estradiol production in immature rat Sertoli cells. Primary cultures of rat Sertoli cells were treated with FSH in combination with EGF and signaling pathway-specific inhibitors. Levels of estradiol production, aromatase mRNA (Cyp19a1), and aromatase protein (CYP19A1) were determined. Western blot analysis was performed to determine the effects of FSH and EGF on levels of activated (phosphorylated) AKT1 and p42 ERK2 and p44 ERK1, also named MAPK1 and MAPK3, respectively. The stimulatory actions of FSH on aromatase mRNA, aromatase protein, and estradiol production were blocked by inhibition of the phosphatidylinositol 3-kinase/AKT1 signaling pathway. In contrast, inhibition of ERK signaling augmented the stimulatory effects of FSH on estradiol production, aromatase mRNA, and protein levels. Furthermore, EGF inhibited the expression of aromatase mRNA and protein in response to FSH, and these inhibitory effects of EGF were critically dependent on the activation of the ERK signaling pathway. We conclude that an active phosphatidylinositol 3-kinase /AKT signaling pathway is required for the stimulatory actions of FSH, whereas an active ERK/MAPK pathway inhibits estradiol production and aromatase expression in immature Sertoli cells.


Asunto(s)
Aromatasa/metabolismo , Hormona Folículo Estimulante/metabolismo , Sistema de Señalización de MAP Quinasas , Fosfatidilinositol 3-Quinasas/metabolismo , Células de Sertoli/metabolismo , Animales , Aromatasa/efectos de los fármacos , Aromatasa/genética , Células Cultivadas , Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Factor de Crecimiento Epidérmico/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Estradiol/metabolismo , Flavonoides/farmacología , Hormona Folículo Estimulante/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/metabolismo , Masculino , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Células de Sertoli/citología , Células de Sertoli/efectos de los fármacos , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA