Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
PLoS Genet ; 17(7): e1009708, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34314419

RESUMO

The TMPRSS2/ERG gene rearrangement occurs in 50% of prostate tumors and results in expression of the transcription factor ERG, which is normally silent in prostate cells. ERG expression promotes prostate tumor formation and luminal epithelial cell fates when combined with PI3K/AKT pathway activation, however the mechanism of synergy is not known. In contrast to luminal fates, expression of ERG alone in immortalized normal prostate epithelial cells promotes cell migration and epithelial to mesenchymal transition (EMT). Migration requires ERG serine 96 phosphorylation via endogenous Ras/ERK signaling. We found that a phosphomimetic mutant, S96E ERG, drove tumor formation and clonogenic survival without activated AKT. S96 was only phosphorylated on nuclear ERG, and differential recruitment of ERK to a subset of ERG-bound chromatin associated with ERG-activated, but not ERG-repressed genes. S96E did not alter ERG genomic binding, but caused a loss of ERG-mediated repression, EZH2 binding and H3K27 methylation. In contrast, AKT activation altered the ERG cistrome and promoted expression of luminal cell fate genes. These data suggest that, depending on AKT status, ERG can promote either luminal or EMT transcription programs, but ERG can promote tumorigenesis independent of these cell fates and tumorigenesis requires only the transcriptional activation function.


Assuntos
Regulação Neoplásica da Expressão Gênica/genética , Neoplasias da Próstata/metabolismo , Animais , Carcinogênese/genética , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Transformação Celular Neoplásica/genética , Transição Epitelial-Mesenquimal , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Genes Reguladores , Xenoenxertos , Humanos , Sistema de Sinalização das MAP Quinases/genética , Masculino , Camundongos , Camundongos Nus , Fosfatidilinositol 3-Quinases/genética , Próstata/patologia , Neoplasias da Próstata/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/genética , Fatores de Transcrição/genética , Ativação Transcricional , Regulador Transcricional ERG/genética , Regulador Transcricional ERG/metabolismo , Proteínas ras/metabolismo
2.
Prostate ; 77(10): 1066-1075, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28497488

RESUMO

BACKGROUND: Inflammation is the most prevalent and widespread histological finding in the human prostate, and associates with the development and progression of benign prostatic hyperplasia and prostate cancer. Several factors have been hypothesized to cause inflammation, yet the role each may play in the etiology of prostatic inflammation remains unclear. This study examined the possibility that the common protozoan parasite Toxoplasma gondii induces prostatic inflammation and reactive hyperplasia in a mouse model. METHODS: Male mice were infected systemically with T. gondii parasites and prostatic inflammation was scored based on severity and focality of infiltrating leukocytes and epithelial hyperplasia. We characterized inflammatory cells with flow cytometry and the resulting epithelial proliferation with bromodeoxyuridine (BrdU) incorporation. RESULTS: We found that T. gondii infects the mouse prostate within the first 14 days of infection and can establish parasite cysts that persist for at least 60 days. T. gondii infection induces a substantial and chronic inflammatory reaction in the mouse prostate characterized by monocytic and lymphocytic inflammatory infiltrate. T. gondii-induced inflammation results in reactive hyperplasia, involving basal and luminal epithelial proliferation, and the exhibition of proliferative inflammatory microglandular hyperplasia in inflamed mouse prostates. CONCLUSIONS: This study identifies the common parasite T. gondii as a new trigger of prostatic inflammation, which we used to develop a novel mouse model of prostatic inflammation. This is the first report that T. gondii chronically encysts and induces chronic inflammation within the prostate of any species. Furthermore, T. gondii-induced prostatic inflammation persists and progresses without genetic manipulation in mice, offering a powerful new mouse model for the study of chronic prostatic inflammation and microglandular hyperplasia.


Assuntos
Próstata , Hiperplasia Prostática , Prostatite , Toxoplasma , Animais , Modelos Animais de Doenças , Masculino , Camundongos , Próstata/microbiologia , Próstata/patologia , Hiperplasia Prostática/etiologia , Hiperplasia Prostática/microbiologia , Hiperplasia Prostática/patologia , Prostatite/etiologia , Prostatite/microbiologia , Prostatite/patologia , Toxoplasma/isolamento & purificação , Toxoplasma/patogenicidade
3.
Am J Physiol Renal Physiol ; 311(4): F817-F821, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27440781

RESUMO

Lower urinary tract symptoms (LUTS) in aging men are extremely common. They have historically been attributed to benign prostatic hyperplasia (BPH), enlargement of the prostate, and bladder outlet obstruction. However, recent studies have revealed acute and chronic inflammation to be highly associated with LUTS, correlated with prostatic enlargement, and implicated as a cause of prostatic fibrosis that contributes to bladder outlet obstruction. This review examines the evidence implicating inflammation and fibrosis in BPH/LUTS. It identifies potential mechanisms by which inflammation may drive nociceptive signaling as well as hyperplastic growth and fibrosis and identifies targets for pharmacological intervention. This is a promising area for research and development of novel therapies to prevent or more effectively treat LUTS in aging men.


Assuntos
Fibrose/patologia , Inflamação/patologia , Sintomas do Trato Urinário Inferior/patologia , Próstata/patologia , Humanos , Sintomas do Trato Urinário Inferior/tratamento farmacológico , Masculino
4.
Prostate ; 76(8): 722-34, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27088546

RESUMO

PURPOSE: Both prostate cancer and benign prostatic hyperplasia are associated with inflammatory microenvironments. Inflammation is damaging to tissues, but it is unclear how the inflammatory microenvironment protects specialized epithelial cells that function to proliferate and repair the tissue. The objective of this study is to characterize the cell death and cell survival response of the prostatic epithelium in response to inflammation. METHODS: We assessed induction of cell death (TNF, TRAIL, TWEAK, FasL) and cell survival factors (IGFs, hedgehogs, IL-6, FGFs, and TGFs) in inflamed and control mouse prostates by ELISA. Cell death mechanisms were determined by immunoblotting and immunofluorescence for cleavage of caspases and TUNEL. Survival pathway activation was assessed by immunoblotting and immunofluorescence for Mcl-1, Bcl-2, Bcl-XL, and survivin. Autophagy was determined by immunoblotting and immunofluorescence for free and membrane associated light chain 3 (LC-3). RESULTS: Cleavage of all four caspases was significantly increased during the first 2 days of inflammation, and survival protein expression was substantially increased subsequently, maximizing at 3 days. By 5 days of inflammation, 50% of prostatic epithelial cells expressed survivin. Autophagy was also evident during the recovery phase (3 days). Finally, immunofluorescent staining of human specimens indicates strong activation of survival proteins juxtaposed to inflammation in inflamed prostate specimens. CONCLUSIONS: The prostate responds to deleterious inflammation with induction of cell survival mechanisms, most notably survivin and autophagy, demonstrating a coordinated induction of survival factors that protects and expands a specialized set of prostatic epithelial cells as part of the repair and recovery process during inflammation.


Assuntos
Sobrevivência Celular/fisiologia , Inflamação/metabolismo , Próstata/metabolismo , Hiperplasia Prostática/metabolismo , Neoplasias da Próstata/metabolismo , Transdução de Sinais/fisiologia , Microambiente Tumoral/fisiologia , Idoso , Animais , Apoptose/fisiologia , Caspases/metabolismo , Citocina TWEAK , Proteína Ligante Fas/metabolismo , Humanos , Inflamação/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Próstata/patologia , Hiperplasia Prostática/patologia , Neoplasias da Próstata/patologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fatores de Necrose Tumoral/metabolismo
5.
Am J Physiol Renal Physiol ; 308(12): F1421-30, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25925259

RESUMO

Prostatic inflammation is a nearly ubiquitous pathological feature observed in specimens from benign prostate hyperplasia and prostate cancer patients. The microenvironment of the inflamed prostate is highly reactive, and epithelial hyperplasia is a hallmark feature of inflamed prostates. How inflammation orchestrates epithelial proliferation as part of its repair and recovery action is not well understood. Here, we report that a novel epithelial progenitor cell population is induced to expand during inflammation. We used sphere culture assays, immunofluorescence, and flow cytometry to show that this population is increased in bacterially induced inflamed mouse prostates relative to naïve control prostates. We confirmed from previous reports that this population exclusively possesses the ability to regrow entire prostatic structures from single cell culture using renal grafts. In addition, putative progenitor cells harvested from inflamed animals have greater aggregation capacity than those isolated from naïve control prostates. Expansion of this critical cell population requires IL-1 signaling, as IL-1 receptor 1-null mice exhibit inflammation similar to wild-type inflamed animals but exhibit significantly reduced progenitor cell proliferation and hyperplasia. These data demonstrate that inflammation promotes hyperplasia in the mouse prostatic epithelium by inducing the expansion of a selected epithelial progenitor cell population in an IL-1 receptor-dependent manner. These findings may have significant impact on our understanding of how inflammation promotes proliferative diseases such as benign prostatic hyperplasia and prostate cancer, both of which depend on expansion of cells that exhibit a progenitor-like nature.


Assuntos
Proliferação de Células/fisiologia , Hiperplasia Prostática/patologia , Células-Tronco/citologia , Animais , Modelos Animais de Doenças , Inflamação/patologia , Interleucina-1/metabolismo , Masculino , Camundongos , Camundongos Knockout
6.
Prostate ; 75(14): 1620-31, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26174474

RESUMO

BACKGROUND: The presence of inflammation in prostate cancer (PCa) and benign prostate hyperplasia (BPH) has been well described but the cellular mechanisms by which inflammation modulates the prostate are currently unclear. Prostate stem cells (PSC) not only maintain prostate homeostasis but also are considered to be the cell of origin of PCa and an important contributor to BPH. However, the impact of inflammation on PSC is not well understood. Therefore, we initiated studies to evaluate the effect of inflammation on PSC. METHOD: Ovalbumin specific CD8(+) T cells were intravenously delivered to intact and castrated prostate ovalbumin expressing transgenic-3 (POET-3) mice to induce inflammation. Lin (CD45/CD31)(-) Sca1(+) CD49f(+) cells (LSC) and progenitor cells within LSC were determined by flow cytometry. Sorted LSC were subjected to a prostate sphere forming assay to evaluate PSC clonal propagation, proliferation, immediate differentiation, and self-renewal ability. Density of individual spheres was measured by a cantilever-based resonator weighing system. Morphology and characterization of prostate spheres was determined by hematoxylin and eosin (H&E) staining and immunohistochemistry (IHC). Finally, immediate PSC differentiation in sphere formation was determined by immunofluorescence for epithelial cytokeratin markers cytokeratin (CK) 5 and CK8. RESULT: Data presented here demonstrate a significant expansion of the proliferative (BrdU(+) ) LSC population, including CK5(+) , p63(+) , CK18(+) cells, as well as intermediate cells (CK5(+) /CK8(+) ) in inflamed prostates. Histological images reveal that PSC from inflamed prostates produce significantly larger spheres, indicating that the enhanced proliferation observed in LSC is sustained in vitro in the absence of inflammatory mediators. In addition, cultures from inflamed PSC yielded increased number of tubule-like spheres. These tube-like spheres grown from PSCs isolated from inflamed mice exhibited stratification of a CK8(+) luminal-like layer and a CK5(+) basal-like layer. Notably, the numbers of spheres formed by inflamed and non-inflamed PSC were equal, suggesting that even though proliferation is enhanced by inflammation, the homeostatic level of PSC is maintained. CONCLUSION: Induction of inflammation promotes PSC expansion and immediate differentiation through highly proliferative progenitor cells while the homeostasis of PSC is maintained.


Assuntos
Autoimunidade/imunologia , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/patologia , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia , Animais , Proliferação de Células/fisiologia , Inflamação/imunologia , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
7.
Mol Cancer ; 13: 61, 2014 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-24642271

RESUMO

BACKGROUND: The RAS/ERK and PI3K/AKT pathways induce oncogenic gene expression programs and are commonly activated together in cancer cells. Often, RAS/ERK signaling is activated by mutation of the RAS or RAF oncogenes, and PI3K/AKT is activated by loss of the tumor suppressor PTEN. In prostate cancer, PTEN deletions are common, but, unlike other carcinomas, RAS and RAF mutations are rare. We have previously shown that over-expression of "oncogenic" ETS transcription factors, which occurs in about one-half of prostate tumors due to chromosome rearrangement, can bypass the need for RAS/ERK signaling in the activation of a cell migration gene expression program. In this study we test the role of RAS/ERK and PI3K/AKT signaling in the function of oncogenic ETS proteins. RESULTS: We find that oncogenic ETS expression negatively correlates with RAS and RAF mutations in prostate tumors. Furthermore, the oncogenic ETS transcription factors only increased cell migration in the absence of RAS/ERK activation. In contrast to RAS/ERK, it has been reported that oncogenic ETS expression positively correlates with PI3K/AKT activation. We identified a mechanistic explanation for this finding by showing that oncogenic ETS proteins required AKT signaling to activate a cell migration gene expression program through ETS/AP-1 binding sequences. Levels of pAKT correlated with the ability of oncogenic ETS proteins to increase cell migration, but this process did not require mTORC1. CONCLUSIONS: Our findings indicate that oncogenic ETS rearrangements cause a cell migration gene expression program to switch from RAS/ERK control to PI3K/AKT control and provide a possible explanation for the high frequency of PTEN, but not RAS/RAF mutations in prostate cancer.


Assuntos
Movimento Celular , Elafina/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , Neoplasias da Próstata/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas ras/genética , Western Blotting , Linhagem Celular Tumoral , Elafina/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Rearranjo Gênico , Humanos , Masculino , Neoplasias da Próstata/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-ets/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Transdução Genética , Proteínas ras/metabolismo
8.
J Pharmacol Exp Ther ; 351(3): 605-15, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25292180

RESUMO

Prostatic inflammation is of considerable importance to urologic research because of its association with benign prostatic hyperplasia and prostate cancer. However, the mechanisms by which inflammation leads to proliferation and growth remain obscure. Here, we show that insulin-like growth factors (IGFs), previously known as critical developmental growth factors during prostate organogenesis, are induced by inflammation as part of the proliferative recovery to inflammation. Using genetic models and in vivo IGF receptor blockade, we demonstrate that the hyperplastic response to inflammation depends on interleukin-1-driven IGF signaling. We show that human prostatic hyperplasia is associated with IGF pathway activation specifically localized to foci of inflammation. This demonstrates that mechanisms of inflammation-induced epithelial proliferation and hyperplasia involve the induction of developmental growth factors, further establishing a link between inflammatory and developmental signals and providing a mechanistic basis for the management of proliferative diseases by IGF pathway modulation.


Assuntos
Interleucina-1/deficiência , Hiperplasia Prostática/metabolismo , Somatomedinas/biossíntese , Idoso , Animais , Humanos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Hiperplasia Prostática/induzido quimicamente , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Somatomedinas/toxicidade
9.
Res Sq ; 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38168414

RESUMO

The majority of patients with benign prostate hyperplasia (BPH) exhibit chronic prostate inflammation and the extent of inflammation correlates with the severity of symptoms. How inflammation contributes to prostate enlargement and/or BPH symptoms and the underlying mechanisms are not clearly understood. We established a unique mouse model Prostate Ovalbumin Expressing Transgenic 3 (POET3) that mimics chronic non-bacterial prostatitis in men to study the role of inflammation in prostate hyperplasia. After the injection of ovalbumin peptide-specific T cells, POET3 prostates exhibited an influx of inflammatory cells and an increase in pro-inflammatory cytokines that led to epithelial and stromal hyperplasia. We have previously demonstrated with the POET3 model that inflammation expands the basal prostate stem cell (bPSC) population and promotes bPSC differentiation in organoid cultures. In this study, we investigated the mechanisms underlying the impact of inflammation on bPSC. We found that AR activity was enhanced in inflamed bPSC and was essential for bPSC differentiation in organoid cultures. Most importantly, we identified, for the first time, interleukin 1 receptor antagonist (IL-1RA) as a key regulator of AR in basal stem cells. IL-1RA was one of the top genes upregulated by inflammation and inhibition of IL-1RA abrogated the enhanced AR nuclear accumulation and activity in organoids derived from inflamed bPSC. The mirroring effects of IL-1RA recombinant protein and IL-1α neutralizing antibody suggest that IL-1RA may function by antagonizing IL-1α inhibition of AR expression. Furthermore, we established a lineage tracing model to follow bPSC during inflammation and under castrate conditions. We found that inflammation induced bPSC proliferation and differentiation into luminal cells even under castrate conditions, indicating that AR activation driven by inflammation in bPSC is sufficient for their proliferation and differentiation under androgen-deprived conditions. However, proliferation of the differentiated bPSC in the luminal layer significantly diminished with castration, suggesting inflammation may not maintain AR activity in stromal cells, as stromal cells deprived of androgen after castration could no longer provide paracrine growth factors essential for luminal proliferation. Taken together, we have discovered novel mechanisms through which inflammation modulates AR signaling in bPSC and induces bPSC luminal differentiation that contributes to prostate hyperplasia.

10.
Prostate ; 72(3): 307-17, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21681776

RESUMO

BACKGROUND: Prostatic inflammation is gaining increasing attention as a potential etiologic factor in prostate cancer, benign prostatic hyperplasia, lower urinary tract symptoms, and CPPS. This study was performed to address the need for a well characterized model of acute prostatic inflammation that may be used to study the effect of acute inflammation on epithelial and stromal cell proliferation, voiding behavior, and neurovascular physiology. METHODS: Uropathogenic E. coli 1677 was instilled transurethrally into adult C57BL/6J male mice. Prostates were analyzed at 1, 2, 3, 5, 7, or 14 days post-instillation and compared to saline-instilled and naïve controls. Time course and severity of inflammation were characterized by the quantity and type of inflammatory infiltrate present, hemorrhage, proliferation, and reactive hyperplasia. RT-PCR was performed to characterize inflammatory mediators including IL-1α, IL-1ß, IL-1RA, IL-18, IL-6, IL-10, IL-8, TNFα, and COX-2. RESULTS: Inflammation was evident in all lobes of the prostate with the DLP most severely affected. Infection consistently led to a significant increase in neutrophils and macrophages in the early stages of prostate infection, followed by lymphocytic inflammation at the later time points. Inflammation was accompanied by induction of several inflammatory genes, including IL-1 family members, IL-6, and COX-2, and induced a significant increase in epithelial proliferation and reactive hyperplasia in all three prostate lobes. CONCLUSIONS: Transurethral inoculation of uropathogenic E. coli 1677 reliably infects the mouse prostate, produces a significant inflammatory response, and induces quantifiable epithelial proliferation and reactive hyperplasia.


Assuntos
Infecções por Escherichia coli/complicações , Escherichia coli/isolamento & purificação , Próstata/microbiologia , Próstata/patologia , Prostatite/microbiologia , Prostatite/patologia , Animais , Proliferação de Células , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Hiperplasia , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Próstata/metabolismo , Prostatite/metabolismo , Índice de Gravidade de Doença , Fatores de Tempo
11.
Differentiation ; 82(4-5): 237-43, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21864972

RESUMO

The condition known as benign prostatic hyperplasia may be defined as a benign enlargement of the prostate gland resulting from a proliferation of both benign epithelial and stromal elements. It might also be defined clinically as a constellation of lower urinary tract symptoms (LUTSs) in aging men. The purpose of this review is to consider the ways in which inflammatory cytokines belonging to the interleukin family, members of the IFG family, and stem cells may contribute to the development and progression of BPH-LUTS. This might occur in three mechanisms: One, interleukin signaling, IFG signaling and stem cells may contribute to reactivation of developmental growth mechanisms in the adult prostate leading to tissue growth. Two, given that epidemiologic studies indicate an increased incidence of BPH-LUTS in association with obesity and diabetes, IFG signaling may provide the mechanistic basis for the effect of diabetes and obesity on prostate growth. Three, expression of interleukins in association with inflammation in the prostate may induce sensitization of afferent fibers innervating the prostate and result in increased sensitivity to pain and noxious sensations in the prostate and bladder and heightened sensitivity to bladder filling.


Assuntos
Interleucinas/metabolismo , Sintomas do Trato Urinário Inferior/metabolismo , Próstata/metabolismo , Hiperplasia Prostática/metabolismo , Receptores de Somatomedina/metabolismo , Somatomedinas/metabolismo , Citocinas/metabolismo , Complicações do Diabetes/metabolismo , Complicações do Diabetes/patologia , Humanos , Inflamação/metabolismo , Sintomas do Trato Urinário Inferior/complicações , Sintomas do Trato Urinário Inferior/patologia , Masculino , Obesidade/complicações , Obesidade/patologia , Próstata/crescimento & desenvolvimento , Próstata/patologia , Hiperplasia Prostática/complicações , Hiperplasia Prostática/patologia , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/metabolismo
12.
Prostate ; 71(8): 791-800, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21456062

RESUMO

BACKGROUND: Phosphatase homologue of tensin (PTEN) is the most commonly mutated gene in prostate cancer. Bone morphogenetic proteins (BMPs) are known to promote differentiation and inhibit proliferation. Previously published reports from other organ systems led us to investigate a mechanistic relationship between PTEN and BMP signaling in prostate epithelial cells. METHODS: We analyzed growth rate and PTEN expression in E6, BPH-1, and C4-2B prostate epithelial cells treated with BMP-4. We also treated doxacyclin-inducible PTEN-C4-2B cells with BMP-4 and doxacyclin to determine the effect of BMP on growth and PTEN expression in conditions of increasing PTEN expression. We determined the dependency of BMP-mediated growth inhibition via siRNA knockdown of PTEN expression and BMP treatment. We determined PTEN protein stability by determining the effect of BMP-4 on PTEN protein at time points after treatment with cyclohexamide, a translation inhibitor. RESULTS: We found that BMP-4 induces PTEN in E6 and BPH-1 cells and reduces proliferation. Knockdown of PTEN attenuated the growth-inhibiting effects of BMP-4 in these cells. BMP-4 had no effect in PTEN-negative C4-2B cells, but doxacyclin-driven PTEN C4-2B cells responded to BMP-4 with enhanced PTEN and growth inhibition. BMP-4 also increased PTEN protein stability. CONCLUSIONS: BMP signaling induces PTEN expression and sustains PTEN protein expression resulting in inhibition of prostate epithelial cell growth. These data are the first to identify a mechanistic linkage between BMP signaling and PTEN in the prostate, both of which are independently identified as tumor suppressors and suggest possible coordinate dysregulation in prostate cancer.


Assuntos
Proteína Morfogenética Óssea 4/fisiologia , PTEN Fosfo-Hidrolase/biossíntese , Próstata/metabolismo , Proteína Morfogenética Óssea 4/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Humanos , Masculino , Próstata/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , RNA Interferente Pequeno/metabolismo
13.
NAR Cancer ; 3(1): zcaa046, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33554122

RESUMO

The TMPRSS2-ERG gene fusion and subsequent overexpression of the ERG transcription factor occurs in ∼50% of prostate tumors, making it the most common abnormality of the prostate cancer genome. While ERG has been shown to drive tumor progression and cancer-related phenotypes, as a transcription factor it is difficult to target therapeutically. Using a genetic screen, we identified the toll-like receptor 4 (TLR4) signaling pathway as important for ERG function in prostate cells. Our data confirm previous reports that ERG can transcriptionally activate TLR4 gene expression; however, using a constitutively active ERG mutant, we demonstrate that the critical function of TLR4 signaling is upstream, promoting ERG phosphorylation at serine 96 and ERG transcriptional activation. The TLR4 inhibitor, TAK-242, attenuated ERG-mediated migration, clonogenic survival, target gene activation and tumor growth. Together these data indicate a mechanistic basis for inhibition of TLR4 signaling as a treatment for ERG-positive prostate cancer.

14.
Mol Cancer Ther ; 18(11): 1947-1960, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31413178

RESUMO

Bladder cancer is the ninth most common cause of cancer-related deaths worldwide. Although cisplatin is used routinely in treating bladder cancer, refractory disease remains lethal for many patients. The recent addition of immunotherapy has improved patient outcomes; however, a large cohort of patients does not respond to these treatments. Therefore, identification of innovative molecular targets for bladder cancer is crucial. Apurinic/apyrimidinic endonuclease 1/redox factor-1 (APE1/Ref-1) is a multifunctional protein involved in both DNA repair and activation of transcription factors through reduction-oxidation (redox) regulation. High APE1/Ref-1 expression is associated with shorter patient survival time in many cancer types. In this study, we found high APE1/Ref-1 expression in human bladder cancer tissue relative to benign urothelium. Inhibition of APE1/Ref-1 redox signaling using APE1/Ref-1-specific inhibitors attenuates bladder cancer cell proliferation in monolayer, in three-dimensional cultures, and in vivo. This inhibition corresponds with an increase in apoptosis and decreased transcriptional activity of NF-κB and STAT3, transcription factors known to be regulated by APE1/Ref-1, resulting in decreased expression of downstream effectors survivin and Cyclin D1 in vitro and in vivo. We also demonstrate that in vitro treatment of bladder cancer cells with APE1/Ref-1 redox inhibitors in combination with standard-of-care chemotherapy cisplatin is more effective than cisplatin alone at inhibiting cell proliferation. Collectively, our data demonstrate that APE1/Ref-1 is a viable drug target for the treatment of bladder cancer, provide a mechanism of APE1/Ref-1 action in bladder cancer cells, and support the use of novel redox-selective APE1/Ref-1 inhibitors in clinical studies. SIGNIFICANCE: This work identifies a critical mechanism for APE1/Ref-1 in bladder cancer growth and provides compelling preclinical data using selective redox activity inhibitors of APE1/Ref-1 in vitro and in vivo.


Assuntos
Benzoquinonas/administração & dosagem , Cisplatino/administração & dosagem , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/genética , Propionatos/administração & dosagem , Neoplasias da Bexiga Urinária/tratamento farmacológico , Idoso , Animais , Benzoquinonas/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/farmacologia , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/antagonistas & inibidores , Sinergismo Farmacológico , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Propionatos/farmacologia , Regulação para Cima/efeitos dos fármacos , Neoplasias da Bexiga Urinária/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Mol Pharmacol ; 73(1): 18-26, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18094075

RESUMO

Prostanoid synthesis via cyclooxygenase (COX)-2 induction during urothelial stretch is central to nociception, inflammation, contractility, and proliferation caused by urinary tract obstruction. We used our primary human urothelial cell stretch model published previously to evaluate the signaling mechanisms responsible for stretch-induced COX-2 expression in urothelial cells. To determine intracytosolic calcium concentrations ([Ca(2+)](i)), primary human urothelial cells were grown on flexible membranes and loaded with Fura-2 acetoxymethyl ester (AM). We determined [Ca(2+)](i) using a fluorescent scope during stretch. Additional cells were treated with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA)-AM, stretched, and COX-2 mRNA and protein were evaluated by real-time polymerase chain reaction and immunoblotting. To evaluate protein kinase C (PKC) in this system, cells were stretched and fractionated into membrane, cytosol, and nucleus. Fractions were immunoblotted for PKCalpha, beta1, and zeta, the predominant isoforms in urothelial cells. We treated additional cells with increasing concentrations of either bisindolylmaleimide-I or a peptide PKC pseudosubstrate inhibitor, and COX-2 mRNA and protein were evaluated after stretching. Furthermore, we transfected urothelial cells with siRNA against each of the inducible PKC isoforms in these cells and evaluated the stretch-induced COX-2 response. Stretch of urothelial cells activated calcium flux and PKC translocation to membrane and nucleus. Pharmacological inhibition indicated that stretch-induced COX-2 expression is dependent on calcium and PKC, and biochemical knockdown experiments indicated that PKCzeta is the predominant isoform mediating stretch-induced COX-2 expression. Elucidating the signaling mechanism of stretch-induced COX-2 expression may identify therapeutic targets.


Assuntos
Cálcio/metabolismo , Ciclo-Oxigenase 2/metabolismo , Proteína Quinase C/metabolismo , Urotélio/metabolismo , Sequência de Bases , Primers do DNA , Humanos , Urotélio/citologia , Urotélio/enzimologia
16.
J Endourol ; 22(4): 739-42, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18324898

RESUMO

INTRODUCTION: The use of ketorolac in the management of painful symptoms associated with urinary stones is well supported in the literature; however, the gastric and renal adverse effects limit the dose and duration of administration. As a nonselective cyclooxygenase inhibitor, ketorolac can act locally to help control renal colic by inhibiting smooth muscle contractions and inflammation. We sought to confirm ketorolac's inhibition of ureteral contractility and determine a dose response relationship to identify an effectiveness range. MATERIALS AND METHODS: Porcine ureter strips attached to force displacement transducers were suspended in organ tissue baths that contained aerated Krebs buffer. Tissues equilibrated for 1 hour, and a spontaneous contractility rate was established. Tissues were incubated with a concentration-response curve of ketorolac (0.1 nM-10 microM) for 90 minutes and compared with indomethacin (1 muM) and dimethyl sulfoxide (DMSO) 0.1%. Contractility rates were recorded on a polygraph and analyzed for changes over exposure time. RESULTS: Ketorolac inhibition of ureteral contractility was dose dependent. At 90 minutes, the average percent decrease from the spontaneous contraction rate for 0.1 nM ketorolac was 18.2%; 1 nM, 34.3%; 10 nM, 56.0%; 100 nM, 69.9%; 1 microM, 88.7%; and 10 microM, 98.3%. Ureteral contractility was significantly reduced by 1 microM ketorolac (39.0%; P = 0.016) at 15 minutes when compared with DMSO. In addition, 1 microM ketorolac was not significantly different at any time point from any of the higher doses studied. CONCLUSION: Ketorolac inhibition of stretch-induced ureteral contractility is concentration-dependent between 1 nM and 1 microM. Local administration of ketorolac at these doses may be useful during the management of stones while at the same time limiting the risk for adverse effects.


Assuntos
Inibidores de Ciclo-Oxigenase/farmacologia , Cetorolaco/farmacologia , Contração Muscular/efeitos dos fármacos , Ureter/efeitos dos fármacos , Animais , Inibidores de Ciclo-Oxigenase/administração & dosagem , Relação Dose-Resposta a Droga , Técnicas In Vitro , Indometacina/farmacologia , Cetorolaco/administração & dosagem , Suínos
17.
Oncotarget ; 9(13): 10962-10977, 2018 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-29541389

RESUMO

A key feature of prostate cancer progression is the induction and activation of survival proteins, including the Inhibitor of Apoptosis (IAP) family member survivin. Apurinic/apyrimidinic endonuclease 1/redox effector factor 1 (APE1/Ref-1) is a multifunctional protein that is essential in activating oncogenic transcription factors. Because APE1/Ref-1 is expressed and elevated in prostate cancer, we sought to characterize APE1/Ref-1 expression and activity in human prostate cancer cell lines and determine the effect of selective reduction-oxidation (redox) function inhibition on prostate cancer cells in vitro and in vivo. Due to the role of oncogenic transcriptional activators NFĸB and STAT3 in survivin protein expression, and APE1/Ref-1 redox activity regulating their transcriptional activity, we assessed selective inhibition of APE1/Ref-1's redox function as a novel method to halt prostate cancer cell growth and survival. Our study demonstrates that survivin and APE1/Ref-1 are significantly higher in human prostate cancer specimens compared to noncancerous controls and that APE1/Ref-1 redox-specific inhibition with small molecule inhibitor, APX3330 and a second-generation inhibitor, APX2009, decreases prostate cancer cell proliferation and induces cell cycle arrest. Inhibition of APE1/Ref-1 redox function significantly reduced NFĸB transcriptional activity, survivin mRNA and survivin protein levels. These data indicate that APE1/Ref-1 is a key regulator of survivin and a potentially viable target in prostate cancer.

18.
Cell Rep ; 17(5): 1289-1301, 2016 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-27783944

RESUMO

More than 50% of prostate tumors have a chromosomal rearrangement resulting in aberrant expression of an oncogenic ETS family transcription factor. However, mechanisms that differentiate the function of oncogenic ETS factors expressed in prostate tumors from non-oncogenic ETS factors expressed in normal prostate are unknown. Here, we find that four oncogenic ETS (ERG, ETV1, ETV4, and ETV5), and no other ETS, interact with the Ewing's sarcoma breakpoint protein, EWS. This EWS interaction was necessary and sufficient for oncogenic ETS functions including gene activation, cell migration, clonogenic survival, and transformation. Significantly, the EWS interacting region of ERG has no homology with that of ETV1, ETV4, and ETV5. Therefore, this finding may explain how divergent ETS factors have a common oncogenic function. Strikingly, EWS is fused to various ETS factors by the chromosome translocations that cause Ewing's sarcoma. Therefore, these findings link oncogenic ETS function in both prostate cancer and Ewing's sarcoma.


Assuntos
Rearranjo Gênico/genética , Oncogenes , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteína Proto-Oncogênica c-ets-1/metabolismo , Proteína EWS de Ligação a RNA/metabolismo , Sarcoma de Ewing/patologia , Animais , Carcinogênese/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos Nus , Fenótipo , Regiões Promotoras Genéticas/genética , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Fatores de Transcrição/metabolismo
19.
J Endourol ; 19(9): 1088-91, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16283845

RESUMO

BACKGROUND AND PURPOSE: We previously found that prostaglandin (PG) E2 contracts acutely obstructed ureters while relaxing normal ureters. This study investigated the procontractile effects of the PG EP3 receptor in PGE(2)-mediated contractility in obstructed and normal porcine ureters. MATERIALS AND METHODS: We created unilateral ureteral obstruction laparoscopically using titanium clips in farm pigs; the contralateral ureters were dissected as sham controls. Ureters were harvested 48 hours post-obstruction, cut into 5-mm segments, and suspended in water-jacketed tissue baths in Krebs buffer. Tissues were equilibrated for 1 hour, and spontaneous contractile rates were recorded. After 2 hours of incubation in Krebs (controls) or pertussis toxin (G(alpha)i signaling-protein inhibitor [EP-3 blockade]) 500 ng/mL, a concentration- response curve (10(-9) M-10(-5) M) to PGE(2), PGF(2), sulprostone (EP 3 agonist), or 0.01% ethanol (vehicle) was created (N = 4). RESULTS: In the normal ureters, PGE(2) relaxed both pertussis toxin-treated and control tissues. In obstructed segments, PGE(2) increased contractions by 60%; this was reversed by pertussis toxin to a 67% reduction in contractile rate. In both obstructed and contralateral segments, sulprostone induced contractility in the controls; this was attenuated by pertussis toxin. The PGF(2) produced a contractile effect in both the controls and the pertussis toxin-treated segments, demonstrating the selectivity of pertussis toxin for EP3 receptors. CONCLUSION: Our data indicate that the EP3 receptor is involved in hypercontractility during ureteral obstruction. However, it may not be the sole factor behind the condition-dependent effect of PGE(2).


Assuntos
Contração Muscular/fisiologia , Músculo Liso/fisiopatologia , Receptores de Prostaglandina E/fisiologia , Obstrução Ureteral/fisiopatologia , Animais , Feminino , Receptores de Prostaglandina E Subtipo EP3 , Suínos , Ureter/fisiopatologia
20.
J Endourol ; 19(2): 183-7, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15798415

RESUMO

BACKGROUND AND PURPOSE: Although our understanding of ureteral physiology during acute obstruction remains limited, we believe that prostanoids (prostaglandins [PGs], thromboxanes, prostacyclins) play a major role in modulation of ureteral contractility and that inhibition of prostanoid synthesis causes substantial reduction in in-vitro and in-vivo ureteral contractility rates. The purpose of this study was to determine the in-vitro effects of PGE2 on chronically obstructed human and acutely obstructed porcine ureters. MATERIALS AND METHODS: Female pigs underwent unilateral laparoscopic ureteral obstruction. Following 1, 2, 6, 24, and 48 hours of obstruction (n = 3 at all points), animals were euthanized, and obstructed, contralateral nonobstructed, and normal (from unobstructed pigs) ureters were harvested. Chronically obstructed human ureter was obtained from subjects who underwent nephrectomy to remove nonfunctioning kidneys. Normal human ureter was obtained from the discarded portion of excess distal ureter in patients undergoing elective donor nephrectomy. Rings 4 to 5 mm long were suspended in aerated Krebs buffer, and their spontaneous contractions and contraction in response to various concentrations of PGE2 were recorded. RESULTS: Prostaglandin E2 increased contractility in chronically obstructed human ureters. In acutely obstructed porcine ureteral segments, low concentrations of PGE2 inhibited ureteral contractility in a dose-dependent fashion, similar to controls. At higher concentrations of PGE2, contractility was increased. This increase was more pronounced with longer intervals of obstruction in a time-dependent manner. CONCLUSION: Prostaglandin E2 increased contractility in obstructed ureters while relaxing normal and nonobstructed ureters. The response to PGE2 was accentuated by a longer duration of obstruction. Prostaglandin E2 may be a unique target for pharmacologic modulation in the treatment of symptoms associated with acute urinary obstruction.


Assuntos
Dinoprostona/farmacologia , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Obstrução Ureteral/fisiopatologia , Animais , Feminino , Humanos , Técnicas In Vitro , Contração Muscular/fisiologia , Músculo Liso/fisiopatologia , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA