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

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Prostate ; 79(15): 1739-1751, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31449679

RESUMEN

BACKGROUND: Castrate resistant prostate cancer (CRPC) accounts for almost all prostate cancer (PCa) deaths. Aberrant activation of ERK/MEK and PI3K/AKT signaling pathways plays an important role in subsets of patients with CRPC. The role of protocadherin 7 (PCDH7) in modulating these signaling pathways is investigated for the first time in PCa in the present investigation. METHODS: PCDH7 expression was analyzed in CRPC/neuroendocrine prostate cancer (NEPC) dataset. Protein expression was assessed by Western blotting and immunohistochemistry, and messenger RNA (mRNA) by quantitative real-time polymerase chain reaction. Small hairpin ribonucleic acid was used to knockdown PCDH7. Colony formation, cell migration, and invasion studies were done using standard protocols. RESULTS: PCDH7 amplification/mRNA upregulation was observed in 41% of patients in CRPC/NEPC dataset. PCDH7 was also overexpressed in CRPC cells. Increased PCDH protein expression was observed during tumor progression in PCa tissues and in TRAMP mice. Epidermal growth factor treatment resulted in aberrant activation of ERK/AKT. Knockdown of PCDH7 decreased ERK, AKT, and RB phosphorylation and reduced colony formation, decreased cell invasion, and cell migration. CONCLUSIONS: These data show for the first time that PCDH7 is overexpressed in a large number of patients with CRPC and suggest that PCDH7 may be an attractive target in subsets of patients with CRPC for whom there is no cure to-date.


Asunto(s)
Cadherinas/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Transducción de Señal/fisiología , Animales , Cadherinas/genética , Línea Celular Tumoral , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Protocadherinas
2.
Cancer Cell Int ; 19: 224, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31496918

RESUMEN

BACKGROUND: Cap-dependent mRNA translation is essential for the translation of key oncogenic proteins at optimal levels and is highly regulated by the rate limiting, initiation step in protein synthesis. Eukaryotic Translation Initiation Factor 4 Gamma 1 (EIF4G1) serves as a scaffold for assembly of cap-dependent translation components in EIF4F complex formation. In the current study, we analyzed the role and expression of EIF4G1 in Pan human cancer panels through various approaches. METHODS: Immunohistochemistry analysis of EIF4G1 protein was done on high-density multi-organ Human Cancer tissue microarray (TMA) derived from the patient samples from different cancers. We used multiple clinical cohorts to analyze the EIF4G1 mRNA expression across human cancers. TCGA data analysis of EIF4G1 was done through Ualcan and c-bioportal web servers. Western blots for EIF4G1 protein was done for different cell lines in representing the multiple cancer types. Dependency score was calculated through Cancer Dependency Map. Clonogenic, tumorosphere assay and cell invasion assay were done with EIF4G complex inhibitor. Association of EIF4G1 mRNA and Kaplan-Meier survival analysis was done on available TCGA datasets. RESULTS: We observed an increase in EIF4G1 protein levels in tissue sections from different cancers as compared to their respective normal tissue. Our analysis of the TCGA data revealed that EIF4G1 mRNA expression is significantly increased in tumor tissues compared to respective control tissues across human cancers and variable expression was observed among different datasets. We discovered that alteration frequency in EIF4G1 is prevalent in human cancers e.g. prostate cancer (~ 25%), ovarian cancer (~ 15%), Head and Neck cancer (~ 13%) and cervical cancer (~ 12.5%). EIF4G1 mRNA and protein levels were high across cancer cell lines from multiple organs. Our analysis of DepMap datasets utilizing depletion assays revealed that EIF4G1 is critical for cancer cell survival. Treatment with EIF4G complex inhibitor impaired clonogenic, tumorosphere formation potential and inhibited cell invasion. Moreover, higher EIF4G1 mRNA level was associated with a lower median survival of patients in multiple tumor types. CONCLUSIONS: These studies show that EIF4G1 is amplified/over-expressed in multiple cancers and plays an essential role in cancer cell survival, as such EIF4G1 could serve as a novel potential target for therapeutic intervention across many cancers.

4.
J Biol Chem ; 288(17): 12222-31, 2013 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-23449978

RESUMEN

Loss of E-cadherin is one of the key steps in tumor progression. Our previous studies demonstrate that SAM pointed domain-containing ETS transcription factor (SPDEF) inhibited prostate cancer metastasis in vitro and in vivo. In the present study, we evaluated the relationship between SPDEF and E-cadherin expression in an effort to better understand the mechanism of action of SPDEF in prostate tumor cell invasion and metastasis. The results presented here demonstrate a direct correlation between expression of E-cadherin and SPDEF in prostate cancer cells. Additional data demonstrate that modulation of E-cadherin and SPDEF had similar effects on cell migration/invasion. In addition, siRNA-mediated knockdown of E-cadherin was sufficient to block the effects of SPDEF on cell migration and invasion. We also show that stable forced expression of SPDEF results in increased expression of E-cadherin, whereas down-regulation of SPDEF decreased E-cadherin expression. In addition, we demonstrate that SPDEF expression is not regulated by E-cadherin. Moreover, our chromatin immunoprecipitation and luciferase reporter assay revealed that SPDEF occupies E-cadherin promoter site and acts as a direct transcriptional inducer of E-cadherin in prostate cancer cells. Taken together, to the best of our knowledge, these studies are the first demonstrating requirement of SPDEF for expression of E-cadherin, an essential epithelial cell junction protein. Given that loss of E-cadherin is a central tenant in tumor metastasis, the results of our studies, by providing a new mechanism for regulation of E-cadherin expression, could have far reaching impact.


Asunto(s)
Cadherinas/biosíntesis , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo , Activación Transcripcional , Cadherinas/genética , Línea Celular Tumoral , Movimiento Celular , Humanos , Masculino , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-ets/genética
5.
Am J Physiol Renal Physiol ; 306(9): F1039-46, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24523387

RESUMEN

The role of inflammation in oxalate-induced nephrolithiasis is debated. Our gene expression study indicated an increase in interleukin-2 receptor ß (IL-2Rß) mRNA in response to oxalate (Koul S, Khandrika L, Meacham RB, Koul HK. PLoS ONE 7: e43886, 2012). Herein, we evaluated IL-2Rß expression and its downstream signaling pathway in HK-2 cells in an effort to understand the mechanisms of oxalate nephrotoxicity. HK-2 cells were exposed to oxalate for various time points in the presence or absence of SB203580, a specific p38 MAPK inhibitor. Gene expression data were analyzed by Ingenuity Pathway Analysis software. mRNA expression was quantitated via real-time PCR, and changes in protein expression/kinase activation were analyzed by Western blotting. Exposure of HK-2 cells to oxalate resulted in increased transcription of IL-2Rß mRNA and increased protein levels. Oxalate treatment also activated the IL-2Rß signaling pathway (JAK1/STAT5 phosphorylation). Moreover, the increase in IL-2Rß protein was dependent upon p38 MAPK activity. These results suggest that oxalate-induced activation of the IL-2Rß pathway may lead to a plethora of cellular changes, the most common of which is the induction of inflammation. These results suggest a central role for the p38 MAPK pathway in mediating the effects of oxalate in renal cells, and additional studies may provide the key to unlocking novel biochemical targets in stone disease.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Subunidad beta del Receptor de Interleucina-2/efectos de los fármacos , Riñón/efectos de los fármacos , Ácido Oxálico/toxicidad , Transducción de Señal/efectos de los fármacos , Western Blotting , Línea Celular , Activación Enzimática , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Humanos , Mediadores de Inflamación/metabolismo , Subunidad beta del Receptor de Interleucina-2/genética , Subunidad beta del Receptor de Interleucina-2/metabolismo , Janus Quinasa 1/metabolismo , Riñón/inmunología , Riñón/metabolismo , Riñón/patología , Nefritis/inducido químicamente , Nefritis/inmunología , Nefritis/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción STAT5/metabolismo , Factores de Tiempo , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
Curr Top Med Chem ; 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38445699

RESUMEN

During the last five decades, there has been tremendous development in our understanding of cancer biology and the development of new and novel therapeutics to target cancer. However, despite these advances, cancer remains the second leading cause of death across the globe. Most cancer deaths are attributed to the development of resistance to current therapies. There is an urgent and unmet need to address cancer therapy resistance. Tetrandrine, a bis-benzyl iso-quinoline, has shown a promising role as an anti-cancer agent. Recent work from our laboratory and others suggests that tetrandrine and its derivatives could be an excellent adjuvant to the current arsenal of anti-cancer drugs. Herein, we provide an overview of resistance mechanisms to current therapeutics and review the existing literature on the anti-cancer effects of tetrandrine and its potential use for overcoming therapy resistance in cancer.

7.
J Biol Chem ; 287(35): 29968-78, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22761428

RESUMEN

Emerging evidence suggests that the SAM pointed domain containing ETS transcription factor (SPDEF) plays a significant role in tumorigenesis in prostate, breast, colon, and ovarian cancer. However, there are no in vivo studies with respect to the role of SPDEF in tumor metastasis. The present study examined the effects of SPDEF on tumor cell metastasis using prostate tumor cells as a model. Utilizing two experimental metastasis models, we demonstrate that SPDEF inhibits cell migration and invasion in vitro and acts a tumor metastasis suppressor in vivo. Using stable expression of SPDEF in PC3-Luc cells and shRNA-mediated knockdown of SPDEF in LNCaP-Luc cells, we demonstrate for the first time that SPDEF diminished the ability of disseminated tumors cells to survive at secondary sites and establish micrometastases. These effects on tumor metastasis were not a result of the effect of SPDEF on cell growth as SPDEF expression had no effect on cell growth in vitro or subcutaneous tumor xenograft-growth in vivo. Transcriptional analysis of several genes associated with tumor metastasis, invasion, and the epithelial-mesenchymal transition demonstrated that SPDEF expression selectively down-regulated MMP9 and MMP13 in prostate cancer cells. Further analysis indicated that forced MMP9 or MMP13 expression rescued the invasive phenotype in SPDEF expressing PC3 cells in vitro, suggesting that the effects of SPDEF on tumor invasion are mediated, in part, through the suppression of MMP9 and MMP13 expression. These results demonstrate for the first time, in any system, that SPDEF functions as a tumor metastasis suppressor in vivo.


Asunto(s)
Movimiento Celular , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo , Animales , Línea Celular Tumoral , Regulación hacia Abajo , Transición Epitelial-Mesenquimal/genética , Regulación Enzimológica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Masculino , Metaloproteinasa 13 de la Matriz/biosíntesis , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/genética , Ratones , Ratones Desnudos , Invasividad Neoplásica , Metástasis de la Neoplasia , Trasplante de Neoplasias , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-ets/genética , Transcripción Genética/genética , Trasplante Heterólogo
8.
Front Endocrinol (Lausanne) ; 14: 1156120, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37900138

RESUMEN

Introduction: Prostate cancer (PCa) presents a significant health challenge in men, with a substantial number of deaths attributed to metastatic castration resistant PCa (mCRPC). Moreover, African American men experience disproportionately high mortality rates due to PCa. This study delves into the pivotal role of SPDEF, a prostate specific Ets transcription factor, and its regulation by DNA methylation in the context of PCa progression. Methods: We performed Epigenetic reprogramming using daily treatment with non-toxic dose of 5Aza-2-deoxycytidine (5Aza-dC) for two weeks to assess its impact on PDEF expression in prostate cancer cells. Next, we conducted functional studies on reprogrammed cells, including cell migration (wound-healing assay), invasion (Boyden-Chamber test), and proliferation (MTT assay) to comprehensively evaluate the consequences of altered PDEF expression. We used bisulfite sequencing (BSP) to examine DNA methylation at SPDEF promoter. Simultaneously, we utilized siRNA-mediated targeting of key DNMTs (DNMT1, DNMT3A, and DNMT3B) to elucidate their specific role in regulating PDEF. We measured mRNA and protein expressions using qRT-PCR and immune-blotting techniques, respectively. Results: In this report, we observed that: a) there is a gradual decrease in SPDEF expression with a concomitant increase in methylated CpG sites within the SPDEF gene during prostate cancer progression from lower to higher Gleason grade; b) Expression of DNMT's (DNMT1, 3a and 3b) is increased during prostate cancer progression, and there is an inverse correlation between SPDEF and DNMT expression; c) SPDEF levels are decreased in RC77/T, a line of PCa cells from African American origin similar to PC3 and DU145 cells (CRPC cells), as compared to LNCaP cells , a line of androgen dependent cells,; d) the 5' CpG island of SPDEF gene are hypermethylated in SPDEF-negative CRPC ( PC3, DU145 and RC77/T) cell lines but the same regions are hypomethylated in SPDEF-positive castrate sensitive (LNCaP) cell line ; (e) expression of SPDEF in PCa cells lacking SPDEF decreases cell migration and invasion, but has no significant effect on cell proliferation, and; (f) treatment with the demethylating agent, 5-aza-2'-deoxycytidine, or silencing of the DNMT's by siRNA, partially restores SPDEF expression in SPDEF-negative PCa cell lines, and decreases cell migration and invasion. Discussion: These results indicate hypermethylation is a prevalent mechanism for decreasing SPDEF expression during prostate cancer progression. The data demonstrate that loss of SPDEF expression in prostate cancer cells, a critical step in cellular plasticity, results from a potentially reversible process of aberrant DNA methylation. These studies suggest DMNT activity as a potential therapeutic vulnerability that can be exploited for limiting cellular plasticity, tumor progression, and therapy resistance in prostate cancer.


Asunto(s)
Metilación de ADN , Neoplasias de la Próstata Resistentes a la Castración , Masculino , Humanos , Neoplasias de la Próstata Resistentes a la Castración/genética , Línea Celular Tumoral , Islas de CpG/genética , Decitabina , ARN Interferente Pequeño , Proteínas Proto-Oncogénicas c-ets/genética
10.
J Urol ; 186(6): 2426-33, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22019171

RESUMEN

PURPOSE: Histone deacetylase inhibitors represent promising cancer treatments since they offer improved access to target DNA/protein complexes by cytotoxic agents. We hypothesized that histone deacetylase inhibitors would be most effective when combined with DNA damaging agents such as mitomycin C. Valproic acid is a safe, affordable histone deacetylase inhibitor. We examined the effect of the combination of valproic acid and mitomycin C on human bladder cancer cells in vitro and compared this to the effect of valproic acid or mitomycin C alone on the cells. MATERIALS AND METHODS: We used HTB5 and HTB9 cells derived from low and high grade bladder tumors, respectively. HTB5 and HTB9 cells were grown in modified Eagle's and RPMI medium, respectively. Cell growth and proliferation were measured by standard methods. Apoptosis was evaluated microscopically after dual staining of cells with annexin V-fluorescein isothiocyanate/propidium iodide. The change in protein expression was analyzed by Western blot. RESULTS: Treatment of HTB5 and HTB9 bladder cancer cells for 24 to 72 hours with valproic acid and mitomycin C resulted in concentration and time dependent decreases in viability and proliferation. HTB9 cells showed marked sensitivity to mitomycin C with a 48-hour 50% median inhibitory concentration of 1 µg. Cells were less sensitive to valproic acid alone with a 48-hour 50% median inhibitory concentration of 2.5 mM. The chromatin structure relaxation induced by valproic acid pretreatment sensitized the bladder cancer cell lines, augmenting the cytotoxic action of mitomycin C. Valproic acid potentiated the induction of cell death by mitomycin C in each cell line in synergistic fashion. The effect of combining the 2 drugs was greater than the sum effect of each drug alone. CONCLUSIONS: Results indicate that the valproic acid and mitomycin C combination is effective, likely due to synergistic mechanisms. Animal model validation is needed but early results suggest promising intravesical treatments for superficial bladder cancer.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Carcinoma de Células Transicionales/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/uso terapéutico , Mitomicina/uso terapéutico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Ácido Valproico/uso terapéutico , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Células Tumorales Cultivadas
11.
Cancer Lett ; 519: 343, 2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-34429223

RESUMEN

This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article is being retracted following correspondence from an Investigation Committee at the University of Colorado Denver. An internal investigation into this manuscript by the University of Colorado Denver, found evidence that there was image manipulation and that these actions warrant retraction to correct the scientific record. Bands on blot obscured or removed, apparent when the images are enhanced (Fig 4A p21 band "C" at 24hr, Fig 4B p21 band "C" at 24hr, and Fig 4B Cyclin A band "10" at 48 hr).

12.
Cancer Lett ; 507: 112-123, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33737002

RESUMEN

Hippo pathway is a master regulator of development, cell proliferation, stem cell function, tissue regeneration, homeostasis, and organ size control. Hippo pathway relays signals from different extracellular and intracellular events to regulate cell behavior and functions. Hippo pathway is conserved from Protista to eukaryotes. Deregulation of the Hippo pathway is associated with numerous cancers. Alteration of the Hippo pathway results in cell invasion, migration, disease progression, and therapy resistance in cancers. However, the function of the various components of the mammalian Hippo pathway is yet to be elucidated in detail especially concerning tumor biology. In the present review, we focused on the Hippo pathway in different model organisms, its regulation and deregulation, and possible therapeutic targets for cancer treatment.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Animales , Antineoplásicos/uso terapéutico , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Drosophila , Resistencia a Antineoplásicos , Epigénesis Genética , Exosomas/metabolismo , Exosomas/patología , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Regulación Neoplásica de la Expresión Génica , Vía de Señalización Hippo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral
13.
Mol Cancer ; 9: 148, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20550708

RESUMEN

BACKGROUND: Prostate-derived Ets factor (PDEF) is expressed in tissues of high epithelial content including prostate, although its precise function has not been fully established. Conventional therapies produce a high rate of cure for patients with localized prostate cancer, but there is, at present, no effective treatment for intervention in metastatic prostate cancer. These facts underline the need to develop new approaches for early diagnosis of aggressive prostate cancer patients, and mechanism based anti-metastasis therapies that will improve the outlook for hormone-refractory prostate cancer. In this study we evaluated role of prostate-derived Ets factor (PDEF) in prostate cancer. RESULTS: We observed decreased PDEF expression in prostate cancer cell lines correlated with increased aggressive phenotype, and complete loss of PDEF protein in metastatic prostate cancer cell lines. Loss of PDEF expression was confirmed in high Gleason Grade prostate cancer samples by immuno-histochemical methods. Reintroduction of PDEF profoundly affected cell behavior leading to less invasive phenotypes in three dimensional cultures. In addition, PDEF expressing cells had altered cell morphology, decreased FAK phosphorylation and decreased colony formation, cell migration, and cellular invasiveness. In contrast PDEF knockdown resulted in increased migration and invasion as well as clonogenic activity. Our results also demonstrated that PDEF downregulated MMP9 promoter activity, suppressed MMP9 mRNA expression, and resulted in loss of MMP9 activity in prostate cancer cells. These results suggested that loss of PDEF might be associated with increased MMP9 expression and activity in aggressive prostate cancer. To confirm results we investigated MMP9 expression in clinical samples of prostate cancer. Results of these studies show increased MMP9 expression correlated with advanced Gleason grade. Taken together our results demonstrate decreased PDEF expression and increased MMP9 expression during the transition to aggressive prostate cancer. CONCLUSIONS: These studies demonstrate for the first time negative regulation of MMP9 expression by PDEF, and that PDEF expression was lost in aggressive prostate cancer and was inversely associated with MMP9 expression in clinical samples of prostate cancer. Based on these exciting results, we propose that loss of PDEF along with increased MMP9 expression should serve as novel markers for early detection of aggressive prostate cancer.


Asunto(s)
Metaloproteinasa 9 de la Matriz/metabolismo , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-ets/fisiología , Secuencia de Bases , Western Blotting , Línea Celular Tumoral , Cartilla de ADN , Humanos , Inmunohistoquímica , Masculino , Fenotipo , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-ets/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Mol Cancer Res ; 6(10): 1639-48, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18922979

RESUMEN

Overexpression of focal adhesion kinase (FAK) has been well correlated with tumor development and/or the maintenance of tumor phenotype. In addition, inappropriate activation of the extracellular regulated kinase (ERK) signaling pathway is common to many human cancers. In the present study, we investigated the interplay between FAK and ERK in androgen-independent prostate cancer cells (PC3 and DU145 cells). We observed that suppression of FAK expression using small interfering RNA-mediated knockdown decreased the clonogenic activity, whereas overexpression of FAK increased it. We also observed that detachment of PC3 and DU145 cells from their substrate induced tyrosine phosphorylation of FAK. ERK knockdown diminished FAK protein levels and tyrosine phosphorylation of FAK as well as FAK promoter-reporter activity. We also tested the effect of MEK inhibitors and small interfering RNA-mediated knockdown of ERK1 and/or ERK2 on cell proliferation, invasiveness, and growth in soft agar of PC3 and DU145 cells. Inhibition of ERK signaling grossly impaired clonogenicity as well as invasion through Matrigel. However, inhibition of ERK signaling resulted in only a modest inhibition of 3H-thymidine incorporation and no effect on overall viability of the cells or increased sensitivity to anoikis. Taken together, these data show, for the first time, a requirement for FAK in aggressive phenotype of prostate cancer cells; reveal interdependence of FAK and ERK1/2 for clonogenic and invasive activity of androgen-independent prostate cancer cells; suggest a role for ERK regulation of FAK in substrate-dependent survival; and show for the first time, in any cell type, the regulation of FAK expression by ERK signaling pathway.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Andrógenos/metabolismo , Anoicis , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Activación Enzimática , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Invasividad Neoplásica , Fenotipo , Fosforilación , ARN Interferente Pequeño/metabolismo , Ensayo de Tumor de Célula Madre
15.
J Urol ; 181(4): 1926-31, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19237171

RESUMEN

PURPOSE: We evaluated the effect of partial bladder outlet obstruction on bladder weight, protein synthesis, mitotic markers and the mitogen activated protein kinase pathway in a mouse model. MATERIALS AND METHODS: Mice were divided into 3 groups, including control, sham treated and partially obstructed. Bladders were harvested from the mice in the partially obstructed group 12, 24, 48, 72 and 168 hours after surgical partial outlet obstruction, respectively. Partially obstructed bladders were compared to bladders in the control and sham treated groups by weight, protein content, and expression of proliferating cellular nuclear antigen, cyclin D3, HsP 70, c-jun and phosphorylated c-jun. Bladders were examined histologically for changes occurring with partial obstruction. RESULTS: We tested 3 groups of mice, including control, sham treated and partially obstructed mice, to understand the pathophysiology of the bladder response to partial obstruction. We found no statistical difference in body weight among the groups. Furthermore, there was a significant increase in bladder weight and protein content in partially obstructed mice compared to those in controls and sham operated mice. There was up-regulation of proliferating cellular nuclear antigen, cyclin D3, HsP70, c-jun and phosphorylated c-jun with partial obstruction. Fibrosis was prominent at 168 hours compared to that in controls. CONCLUSIONS: Bladder weight and protein content increase with partial bladder outlet obstruction in mice. Cell cycle proteins and elements of the mitogen activated protein kinase pathway are up-regulated during this process.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/fisiología , Obstrucción del Cuello de la Vejiga Urinaria/enzimología , Animales , Femenino , Ratones , Ratones Endogámicos C57BL , Regulación hacia Arriba
16.
Int J Oncol ; 34(6): 1557-64, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19424573

RESUMEN

Transitional cell carcinoma (TCC) is the most common form of bladder cancer. In bladder cancer, which in terms of its origins and genetics, is a representative of invasive tumors, the differing clinical course and the limited value of established prognostic markers compelled many researchers to look for new molecular parameters in predicting the prognosis and treatment of patients with bladder cancer. Activation of mitogen activated protein kinase (MAPK) is a frequent event in tumor progression and metastasis. In the current study, we investigated the role of two different MAPKs (ERK1/2 and p38) by using their specific inhibitors PD98059 and SB203580 respectively, on bladder cancer growth in two cell lines derived from different tumor stages. Our preliminary work showed that ERK1/2 and p38 MAP kinase are active during the log phase growth of bladder cancer, and inhibition of these pathways could reduce proliferation and growth. Moreover, treatment with these inhibitors hinders DNA synthesis, and has differential effects on the progression of cell cycle. ERK1/2 inhibitor caused cyclin B1-dependent G2/M arrest in both HTB5 and HTB9 bladder cancer cell lines, where as p38 MAPK inhibitor showed G2/M arrest in HTB9 and G1 arrest in HTB5 cell line. Furthermore, decreased proliferation and growth arrest caused by MAPK inhibitors was found to be a reflection of apoptotic induction by these inhibitors in bladder cancer cells. Thus, these studies establish MAPKs as a molecular target in bladder cancer growth which could provide new molecular modalities in clinical application.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Western Blotting , Carcinoma de Células Transicionales/tratamiento farmacológico , Carcinoma de Células Transicionales/metabolismo , Carcinoma de Células Transicionales/patología , Ciclo Celular/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Flavonoides/farmacología , Humanos , Imidazoles/farmacología , Técnicas para Inmunoenzimas , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Piridinas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
17.
Pharmaceuticals (Basel) ; 12(4)2019 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-31835563

RESUMEN

PDEF (prostate-derived ETS factor, also known as SAM-pointed domain containing ETS transcription factor (SPDEF)) is expressed in luminal epithelial cells of the prostate gland and associates with luminal phenotype. The Hippo pathway regulates cell growth/proliferation, cellular homeostasis, and organ development by modulating phosphorylation of its downstream effectors. In previous studies, we observed decreased levels of PDEF during prostate cancer progression. In the present study, we evaluated the effects of the expression of PDEF on total/phosphoprotein levels of YAP1 (a downstream effector of the Hippo pathway). We observed that the PC3 and DU145 cells transfected with PDEF (PDEF-PC3 and PDEF-DU145) showed an increased phospho-YAP1 (Ser127) and total YAP1 levels as compared to the respective PC3 vector control (VC-PC3) and DU145 vector control cells (VC-DU145). We also observed an increased cytoplasmic YAP1 levels in PDEF-PC3 cells as compared to VC-PC3 cells. Moreover, our gene set enrichment analysis (GSEA) of mRNA expression in PDEF-PC3 and VC-PC3 cells revealed that PDEF resulted in inhibition of YAP1 target genes, directly demonstrating that PDEF plays a critical role in modulating YAP1 activity, and by extension in the regulation of the Hippo pathway. We also observed a decrease in YAP1 mRNA levels in prostate cancer tissues as compared to normal prostate tissues. Our analysis of multiple publicly available clinical cohorts revealed a gradual decrease in YAP1 mRNA expression during prostate cancer progression and metastasis. This decrease was similar to the decrease in PDEF levels, which we had reported earlier, and we observed a direct correlation between PDEF and YAP1 expression in CRPC data set. To the best of our knowledge, these results provide the first demonstration of inhibiting YAP1 activity by PDEF in any system and suggest a cross-talk between PDEF and the Hippo signaling pathway.

18.
Cancer Lett ; 452: 132-143, 2019 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-30905813

RESUMEN

Elevated levels of Reactive Oxygen Species (ROS), increased antioxidant ability and the maintenance of redox homeostasis can cumulatively contribute to tumor progression and metastasis. The sources and the role of ROS in a heterogeneous tumor microenvironment can vary at different stages of tumor: initiation, development, and progression, thus making it a complex subject. In this review, we have summarized the sources of ROS generation in cancer cells, its role in the tumor microenvironment, the possible functions of ROS and its important scavenger systems in tumor progression with special emphasis on solid tumors.


Asunto(s)
NADPH Oxidasa 4/metabolismo , Neoplasias/patología , Especies Reactivas de Oxígeno/metabolismo , Microambiente Tumoral/fisiología , Hipoxia de la Célula/fisiología , Humanos , Factor 2 Relacionado con NF-E2/metabolismo , Oxidación-Reducción , Transducción de Señal
19.
J Urol ; 179(5): 2057-63, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18355855

RESUMEN

PURPOSE: Our understanding of physiological and pathophysiological events associated with inner medullary collecting duct epithelium is based on studies in cells isolated from mice and rats. We established primary cultures of hIMCD (human papillary collecting duct epithelial) cells. MATERIALS AND METHODS: Normal papillary tissues were dissected from the surgical waste of consenting patients undergoing renal surgery. Tissues were digested enzymatically. Cells were maintained in Dulbecco's modified Eagle's medium supplemented with glucose and antibiotics. Cultures were treated with ethylenediaminetetraacetic acid and epithelial select medium was also used to obtain a pure epithelial culture. RESULTS: The hIMCD cells grew in a monolayer. Cells showed the expression of epithelial specific markers, including cytokeratin, the tight junction marker zonula occludens 1 and the cytoskeletal protein vimentin. They lacked expression of factor VIII, which is a glycoprotein synthesized by endothelial cells. To our knowledge we also noted for the first time uroplakin expression in collecting duct epithelial cells. This expression was maintained in primary culture. The hIMCD cells in culture were highly resistant to hypertonic solutions and they responded to hypertonicity by cyclooxygenase-2 over expression. Moreover, these cells also survived prolonged periods of hypoxia. CONCLUSIONS: To our knowledge this is the first report of successful culture and characterization of primary cultures of collecting duct epithelial cells from human renal papillae. These cells will serve as essential tools in helping us fill the gaps in our understanding of the events associated with the physiology and pathophysiology of human renal inner medullary collecting duct epithelium.


Asunto(s)
Técnicas de Cultivo de Célula , Médula Renal/citología , Túbulos Renales Colectores/citología , Hipoxia de la Célula , Supervivencia Celular , Medios de Cultivo , Ciclooxigenasa 2/genética , Células Epiteliales/citología , Expresión Génica , Gliceraldehído 3-Fosfato Deshidrogenasa (NADP+)/genética , Humanos , Concentración Osmolar
20.
Mol Cancer Ther ; 17(6): 1217-1228, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29549167

RESUMEN

TNF-related apoptosis-inducing ligand (TRAIL) selectively induces apoptosis in cancer cells, but not in normal cells; as such, it is a promising therapeutic agent. However, therapeutic resistance limits its clinical use in many malignancies, including prostate cancer. Strategies to sensitize cancer cells to TRAIL are urgently needed. We demonstrate here that small-molecule tetrandrine (TET) potentially sensitizes previously resistant (LNCaP and C4-2B cells) and mildly sensitive (PC3 cells) prostate cancer cells to TRAIL-induced apoptosis, and they do so by upregulating mRNA expression and protein levels of death receptors Apo Trail R1 (DR4) and Apo Trail R2 (DR5). Using shRNA knockdown, we show critical requirement of DR4 and DR5 in sensitization of prostate cancer cells to TRAIL. We show that double knockdown of DR4 and DR5 abrogated the apoptotic effects of TET and TRAIL. We also demonstrate that TET-induced DR4 and DR5 expression is independent of p53 status. Given that loss of p53 is associated with progression of prostate cancer to CRPC and NEPC, our results show that TET, by acting as a TRAIL-sensitizing agent in prostate cancer, could serve as a potential therapeutic agent in CRPC and NEPC, for which there is no cure to date. Mol Cancer Ther; 17(6); 1217-28. ©2018 AACR.


Asunto(s)
Apoptosis/efectos de los fármacos , Apoptosis/genética , Bencilisoquinolinas/farmacología , Neoplasias de la Próstata/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ciencias Bioconductuales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Neoplasias de la Próstata/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA